Anticoagulation for atrial fibrillation

Article Type
Changed
Wed, 09/06/2017 - 08:04
Display Headline
Anticoagulation for atrial fibrillation

To the Editor: As a geriatric medicine fellow, I eagerly read Hagerty and Rich’s review “Fall risk and anticoagulation for atrial fibrillation in the elderly: A delicate balance1 and Suh’s editorial, “Whether to anticoagulate: Toward a more reasoned approach2 in the January 2017 issue. Both pieces were helpful and informative.

I appreciate that Dr. Suh encourages shared decision-making between physicians and patients that balances patient preferences and risk stratification to inform whether to anticoagulate. He states, “Unfortunately, there is no similar screening tool to predict bleeding risk from anticoagulation with greater precision in the middle to lower part of the risk spectrum...The patient’s life expectancy and personal preferences are important independent factors that affect the decision of whether to anticoagulate or not.” 

Dr. Mark Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) incorporates patients’ CHA2DS2-VASc and HAS-BLED scores to determine their quality-adjusted life expectancy on or off anticoagulation. The tool helps guide physicians and patients to make shared decisions about anticoagulation.3–5 The AFDST informs clinicians if a patient is undertreated or being treated unnecessarily. Eckman and his colleagues have demonstrated the AFDST’s effective application in clinical practice, including for older adults. I invite readers to learn more about Eckman’s work!

References
  1. Hagerty T, Rich MW. Fall risk and anticoagulation for atrial fibrillation in the elderly: a delicate balance. Cleve Clin J Med 2017; 84:35–40.
  2. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  3. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  4. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  5. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
Article PDF
Author and Disclosure Information

Elise Henning, MD, MEd
Geriatric Medicine Fellow, University of Cincinnati/Christ Hospital

Issue
Cleveland Clinic Journal of Medicine - 84(9)
Publications
Topics
Page Number
658-659
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2Ds@-VASc, HAS-BLED, Mark Eckman, Elise Henning
Sections
Author and Disclosure Information

Elise Henning, MD, MEd
Geriatric Medicine Fellow, University of Cincinnati/Christ Hospital

Author and Disclosure Information

Elise Henning, MD, MEd
Geriatric Medicine Fellow, University of Cincinnati/Christ Hospital

Article PDF
Article PDF
Related Articles

To the Editor: As a geriatric medicine fellow, I eagerly read Hagerty and Rich’s review “Fall risk and anticoagulation for atrial fibrillation in the elderly: A delicate balance1 and Suh’s editorial, “Whether to anticoagulate: Toward a more reasoned approach2 in the January 2017 issue. Both pieces were helpful and informative.

I appreciate that Dr. Suh encourages shared decision-making between physicians and patients that balances patient preferences and risk stratification to inform whether to anticoagulate. He states, “Unfortunately, there is no similar screening tool to predict bleeding risk from anticoagulation with greater precision in the middle to lower part of the risk spectrum...The patient’s life expectancy and personal preferences are important independent factors that affect the decision of whether to anticoagulate or not.” 

Dr. Mark Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) incorporates patients’ CHA2DS2-VASc and HAS-BLED scores to determine their quality-adjusted life expectancy on or off anticoagulation. The tool helps guide physicians and patients to make shared decisions about anticoagulation.3–5 The AFDST informs clinicians if a patient is undertreated or being treated unnecessarily. Eckman and his colleagues have demonstrated the AFDST’s effective application in clinical practice, including for older adults. I invite readers to learn more about Eckman’s work!

To the Editor: As a geriatric medicine fellow, I eagerly read Hagerty and Rich’s review “Fall risk and anticoagulation for atrial fibrillation in the elderly: A delicate balance1 and Suh’s editorial, “Whether to anticoagulate: Toward a more reasoned approach2 in the January 2017 issue. Both pieces were helpful and informative.

I appreciate that Dr. Suh encourages shared decision-making between physicians and patients that balances patient preferences and risk stratification to inform whether to anticoagulate. He states, “Unfortunately, there is no similar screening tool to predict bleeding risk from anticoagulation with greater precision in the middle to lower part of the risk spectrum...The patient’s life expectancy and personal preferences are important independent factors that affect the decision of whether to anticoagulate or not.” 

Dr. Mark Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) incorporates patients’ CHA2DS2-VASc and HAS-BLED scores to determine their quality-adjusted life expectancy on or off anticoagulation. The tool helps guide physicians and patients to make shared decisions about anticoagulation.3–5 The AFDST informs clinicians if a patient is undertreated or being treated unnecessarily. Eckman and his colleagues have demonstrated the AFDST’s effective application in clinical practice, including for older adults. I invite readers to learn more about Eckman’s work!

References
  1. Hagerty T, Rich MW. Fall risk and anticoagulation for atrial fibrillation in the elderly: a delicate balance. Cleve Clin J Med 2017; 84:35–40.
  2. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  3. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  4. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  5. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
References
  1. Hagerty T, Rich MW. Fall risk and anticoagulation for atrial fibrillation in the elderly: a delicate balance. Cleve Clin J Med 2017; 84:35–40.
  2. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  3. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  4. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  5. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Page Number
658-659
Page Number
658-659
Publications
Publications
Topics
Article Type
Display Headline
Anticoagulation for atrial fibrillation
Display Headline
Anticoagulation for atrial fibrillation
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2Ds@-VASc, HAS-BLED, Mark Eckman, Elise Henning
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2Ds@-VASc, HAS-BLED, Mark Eckman, Elise Henning
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

In reply: Anticoagulation for atrial fibrillation

Article Type
Changed
Wed, 09/13/2017 - 15:20
Display Headline
In reply: Anticoagulation for atrial fibrillation

In Reply: I appreciate Dr. Henning’s letter in response to my editorial.1 Indeed, Dr. Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) is useful for determining quality-adjusted life expectancy on or off anticoagulation, and could possibly help with shared decision-making in regard to anticoagulation.2–4 

However, the AFDST does not incorporate personal preferences regarding anticoagulant or medication use in general. Many older adults are on too many medications (ie, polypharmacy) and wish to reduce their overall pill count.

A number of potential barriers to shared decision-making regarding medication use have been identified, including poor physician communication skills, the growing number of available medications, multiple prescribers for the same patient, lack of trust in the prescribing physician, and patients feeling that their preferences are not valued or important.5 Until communication and acceptance between prescribers and patients regarding possible medication choices improves, shared decision-making for medication use in general and anticoagulant use in particular will be an unfulfilled ideal.

References
  1. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  2. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  3. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  4. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
  5. Belcher VN, Fried TR, Agostini JV, Tinetti ME.  Views of older adults on patient participation in medication-related decision making.  J Gen Intern Med 2006; 21:298–303.
Article PDF
Author and Disclosure Information

Theodore T. Suh, MD, PhD, MHS, AGSF
University of Michigan Geriatric Center, Ann Arbor

Issue
Cleveland Clinic Journal of Medicine - 84(9)
Publications
Topics
Page Number
658-659
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2DS2-VASc, HAS-BLED, Mark Eckman, Elise Henning, Theodore Suh
Sections
Author and Disclosure Information

Theodore T. Suh, MD, PhD, MHS, AGSF
University of Michigan Geriatric Center, Ann Arbor

Author and Disclosure Information

Theodore T. Suh, MD, PhD, MHS, AGSF
University of Michigan Geriatric Center, Ann Arbor

Article PDF
Article PDF
Related Articles

In Reply: I appreciate Dr. Henning’s letter in response to my editorial.1 Indeed, Dr. Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) is useful for determining quality-adjusted life expectancy on or off anticoagulation, and could possibly help with shared decision-making in regard to anticoagulation.2–4 

However, the AFDST does not incorporate personal preferences regarding anticoagulant or medication use in general. Many older adults are on too many medications (ie, polypharmacy) and wish to reduce their overall pill count.

A number of potential barriers to shared decision-making regarding medication use have been identified, including poor physician communication skills, the growing number of available medications, multiple prescribers for the same patient, lack of trust in the prescribing physician, and patients feeling that their preferences are not valued or important.5 Until communication and acceptance between prescribers and patients regarding possible medication choices improves, shared decision-making for medication use in general and anticoagulant use in particular will be an unfulfilled ideal.

In Reply: I appreciate Dr. Henning’s letter in response to my editorial.1 Indeed, Dr. Eckman’s Atrial Fibrillation Decision Support Tool (AFDST) is useful for determining quality-adjusted life expectancy on or off anticoagulation, and could possibly help with shared decision-making in regard to anticoagulation.2–4 

However, the AFDST does not incorporate personal preferences regarding anticoagulant or medication use in general. Many older adults are on too many medications (ie, polypharmacy) and wish to reduce their overall pill count.

A number of potential barriers to shared decision-making regarding medication use have been identified, including poor physician communication skills, the growing number of available medications, multiple prescribers for the same patient, lack of trust in the prescribing physician, and patients feeling that their preferences are not valued or important.5 Until communication and acceptance between prescribers and patients regarding possible medication choices improves, shared decision-making for medication use in general and anticoagulant use in particular will be an unfulfilled ideal.

References
  1. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  2. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  3. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  4. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
  5. Belcher VN, Fried TR, Agostini JV, Tinetti ME.  Views of older adults on patient participation in medication-related decision making.  J Gen Intern Med 2006; 21:298–303.
References
  1. Suh TT. Whether to anticoagulate: toward a more reasoned approach. Cleve Clin J Med 2017; 84:41–42.
  2. Eckman MH, Lip GYH, Wise RE, et al. Impact of an atrial fibrillation decision support tool on thromboprophylaxis for atrial fibrillation. Am Heart J 2016; 176:17–27.
  3. Eckman MH, Wise RE, Speer B, et al. Integrating real-time clinical information to provide estimates of net clinical benefit antithrombotic therapy for patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2014; 7:680–686.
  4. Eckman MH, Lip TYH, Wise RE, et al. Using an atrial fibrillation decision support tool for thromboprophylaxis in atrial fibrillation: effect of sex and age. J Am Geriatr Soc 2016; 64:1054–1060.
  5. Belcher VN, Fried TR, Agostini JV, Tinetti ME.  Views of older adults on patient participation in medication-related decision making.  J Gen Intern Med 2006; 21:298–303.
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Page Number
658-659
Page Number
658-659
Publications
Publications
Topics
Article Type
Display Headline
In reply: Anticoagulation for atrial fibrillation
Display Headline
In reply: Anticoagulation for atrial fibrillation
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2DS2-VASc, HAS-BLED, Mark Eckman, Elise Henning, Theodore Suh
Legacy Keywords
anticoagulation, atrial fibrillation, risk, Atrial Fibrillation Decision Support Tool, AFDST, CHA2DS2-VASc, HAS-BLED, Mark Eckman, Elise Henning, Theodore Suh
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Renal denervation: What happened, and why?

Article Type
Changed
Fri, 09/01/2017 - 09:10
Display Headline
Renal denervation: What happened, and why?

Many patients, clinicians, and researchers had hoped that renal denervation would help control resistant hypertension. However, in the SYMPLICITY HTN-3 trial,1 named for the catheter-based system used in the study (Symplicity RDN, Medtronic, Dublin, Ireland), this endovascular procedure failed to meet its primary and secondary efficacy end points, although it was found to be safe. These results were surprising, especially given the results of an earlier randomized trial (SYMPLICITY HTN-2),2 which showed larger reductions in blood pressures 6 months after denervation than in the current trial.

See related editorial

Here, we discuss the results of the SYMPLICITY HTN-3 trial and offer possible explanations for its negative outcomes.

LEAD-UP TO SYMPLICITY HTN-3

Renal denervation consists of passing a catheter through the femoral artery into the renal arteries and ablating their sympathetic nerves using radiofrequency energy. In theory, this should interrupt efferent sympathetic communication between the brain and renal arteries, reducing muscular contraction of these arteries, increasing renal blood flow, reducing activation of the renin-angiotensin-adosterone system, thus reducing sodium retention, reducing afferent sympathetic communication between the kidneys and brain, and in turn reducing further sympathetic activity elsewhere in the body, such as in the heart. Blood pressure should fall.3

The results of the SYMPLICITY HTN-1 and 2 trials were discussed in an earlier article in this Journal,3 and the Medtronic-Ardian renal denervation system has been available in Europe and Australia for clinical use for over 2 years.4 Indeed, after the SYMPLICITY HTN-2 results were published in 2010, Boston Scientific’s Vessix, St. Jude Medical’s EnligHTN, and Covidien’s OneShot radiofrequency renal denervation devices—albeit each with some modifications—received a Conformité Européene (CE) mark and became available in Europe and Australia for clinical use. These devices are not available for clinical use or research in the United States.3,5

Therefore, SYMPLICITY HTN-3, sponsored by Medtronic, was designed to obtain US Food and Drug Administration approval in the United States.6

SYMPLICITY HTN-3 DESIGN

Inclusion criteria were similar to those in the earlier SYMPLICITY trials. Patients had to have resistant hypertension, defined as a systolic blood pressure ≥ 160 mm Hg despite taking at least 3 blood pressure medications at maximum tolerated doses. Patients were excluded if they had a glomerular filtration rate of less than 45 mL/min/1.73 m2, renal artery stenosis, or known secondary hypertension.

A total of 1,441 patients were enrolled, of whom 364 were eventually randomized to undergo renal denervation, and 171 were randomized to undergo a sham procedure. The mean systolic blood pressure at baseline was 188 mm Hg in each group. Most patients were taking maximum doses of blood pressure medications, and almost one-fourth were taking an aldosterone antagonist. Patients in both groups were taking an average of 5 medications.

The 2 groups were well matched for important covariates, including obstructive sleep apnea, diabetes mellitus, and renal insufficiency. Most of the patients were white; 25% of the renal denervation group and 29% of the sham procedure group were black.

The physicians conducting the follow-up appointments did not know which procedure the patients underwent, and neither did the patients. Medications were closely monitored, and patients had close follow-up. The catheter (Symplicity RDS, Medtronic) was of the same design that was used in the earlier SYMPLICITY trials and in clinical practice in countries where renal denervation was available.

Researchers expected that the systolic blood pressure, as measured in the office, would fall in both groups, but they hoped it would fall farther in the denervation group—at least 5 mm Hg farther, the primary end point of the trial. The secondary effectiveness end point was a 2-mm Hg greater reduction in 24-hour ambulatory systolic blood pressure.

 

 

SYMPLICITY HTN-3 RESULTS

No statistically significant difference in safety was observed between the denervation and control groups. However, the procedure was associated with 1 embolic event and 1 case of renal artery stenosis.

Blood pressure fell in both groups. However, at 6 months, office systolic pressure had fallen by a mean of 14.13 mm Hg in the denervation group and 11.74 mm Hg in the sham procedure group, a difference of only 2.39 mm Hg. The mean ambulatory systolic blood pressure had fallen by 6.75 vs 4.79 mm Hg, a difference of only 1.96 mm Hg. Neither difference was statistically significant.

A number of prespecified subgroup analyses were conducted, but the benefit of the procedure was statistically significant in only 3 subgroups: patients who were not black (P = .01), patients who were less than 65 years old (P = .04), and patients who had an estimated glomerular filtration rate of 60 mL/min/1.73 m2 or higher (P = .05).

WHAT WENT WRONG?

The results of SYMPLICITY HTN-3 were disappointing and led companies that were developing renal denervation devices to discontinue or reevaluate their programs.

Although the results were surprising, many observers (including our group) raised concerns about the initial enthusiasm surrounding renal denervation.3–7 Indeed, in 2010, we had concerns about the discrepancy between office-based blood pressure measurements (the primary end point of all renal denervation trials) and ambulatory blood pressure measurements in SYMPLICITY HTN-2.7

The enthusiasm surrounding this procedure led to the publication of 2 consensus documents on this novel therapy based on only 1 small randomized controlled study (SYMPLICITY HTN-2).8,9 Renal denervation was even reported to be useful in other conditions involving the sympathorenal axis, including diabetes mellitus, metabolic syndrome, and obstructive sleep apnea, and also as a potential treatment adjunct in atrial fibrillation and other arrhythmias.5

What went wrong?

Shortcomings in trial design?

The trial was well designed. Both patients and operators were blinded to the procedure, and 24-hour ambulatory blood pressure monitoring was used. We presume that appropriate patients with resistant hypertension were enrolled—the mean baseline systolic blood pressure was 188 mm Hg, and patients in each group were taking an average of 5 medications.

On the other hand, true medication adherence is difficult to ascertain. Further, the term maximal “tolerated” doses of medications is vague, and we cannot rule out the possibility that some patients were enrolled who did not truly have resistant hypertension—they simply did not want to take medications.

Patients were required to be on a stable medication regimen before enrollment and, ideally, to not have any medication changes during the course of the study, but at least 40% of patients did require medication changes during the study. Additionally, it is unclear whether all patients underwent specific testing to rule out secondary hypertension, as this was done at the discretion of the treating physician.

First-generation catheters?

The same type of catheter was used as in the earlier SYMPLICITY trials, and it had been used in many patients in clinical practice in countries where the catheter is routinely available. It is unknown, however, whether newer multisite denervation devices would yield better results than the first-generation devices used in SYMPLICITY HTN-3. But even this would not explain the discrepancies in data between earlier trials and this trial.

Operator inexperience?

It has been suggested that operator inexperience may have played a role, but an analysis of operator volume did not find any association between this variable and the outcomes. Each procedure was supervised by at least 1 and in most cases 2 certified Medtronic representatives, who made certain that meticulous attention was paid to procedure details and that no shortcuts were taken during the procedure.

Inadequate ablation?

While we can assume that the correct technique was followed in most cases, renal denervation is still a “blind” procedure, and there is no nerve mapping to ascertain the degree of ablation achieved. Notably, patients who had the most ablations reportedly had a greater average drop in systolic ambulatory blood pressure than those who received fewer ablations. Sympathetic nervous system activity is a potential marker of adequacy of ablation, but it was not routinely assessed in the SYMPLICITY HTN-3 trial. Techniques to assess sympathetic nerve activity such as norepinephrine spillover and muscle sympathetic nerve activity are highly specialized and available only at a few research centers, and are not available for routine clinical use.

While these points may explain the negative findings of this trial, they fail to account for the discrepant results between this study and previous trials that used exactly the same definitions and techniques.

 

 

Patient demographics?

Is it possible that renal denervation has a differential effect according to race? All previous renal denervation studies were conducted in Europe or Australia; therefore, few data are available on the efficacy of the procedure in other racial groups, such as black Americans. Most of the patients in this trial were white, but approximately 25% were black—a good representation. There was a statistically significant benefit favoring renal denervation in nonblack (mostly white) patients, but not in black patients. This may be related to racial differences in the pathophysiology of hypertension or possibly due to chance alone.

A Hawthorne effect?

A Hawthorne effect (patients being more compliant because physicians are paying more attention to them) is unlikely, since the renal denervation arm did not have any reduction in blood pressure medications. At 6 months, both the sham group and the procedure group were still on an average of 5 medications.

Additionally, while the blood pressure reduction in both treatment groups was significant, the systolic blood pressure at 6 months was still 166 mm Hg in the denervation group and 168 mm Hg in the sham group. If denervation was effective, one would have expected a greater reduction in blood pressure or at least a decrease in the number of medications needed, eg, 1 to 2 fewer medications in the denervation group compared with the sham procedure group.

Regression to the mean?

It is unknown whether the results represent a statistical error such as regression to the mean. But given the run-in period and the confirmatory data from 24-hour ambulatory blood pressure, this would be unlikely.

WHAT NOW?

Is renal denervation dead? SYMPLICITY HTN-3 is only a single trial with multiple shortcomings and lessons to learn from. Since its publication, there have been updates from 2 prospective, randomized, open-label trials concerning the efficacy of catheter-based renal denervation in lowering blood pressure.10,11

DENERHTN (Renal Denervation for Hypertension)10 studied patients with ambulatory systolic blood pressure higher than 135 mm Hg, diastolic blood pressure higher than 80 mm Hg, or both (after excluding secondary etiologies), despite 4 weeks of standardized triple-drug treatment including a diuretic. Patients were randomized to standardized stepped-care antihypertensive treatment alone (control group) or standard care plus renal denervation. The latter resulted in a significant further reduction in ambulatory blood pressure at 6 months.

The Prague-15 trial11 studied patients with resistant hypertension. Secondary etiologies were excluded and adherence to therapy was confirmed by measuring plasma medication levels. It showed that renal denervation along with optimal antihypertensive medical therapy (unchanged after randomization) resulted in a significant reduction in ambulatory blood pressure that was comparable to the effect of intensified antihypertensive medical therapy including spironolactone. (Studies have shown that spironolactone is effective when added on as a fourth-line medication in resistant hypertension.12) At 6 months, patients in the intensive medical therapy group were using an average of 0.3 more antihypertensive medications than those in the procedure group.

These two trials addressed some of the drawbacks of the SYMPLICITY HTN-3 trial. However, both have many limitations including and not limited to being open-label and nonblinded, lacking a sham procedure, using a lower blood pressure threshold than SYMPLICITY HTN-3 did to define resistant hypertension, and using the same catheter as in the SYMPLICITY trials.

 

 

Better technology is coming

Distribution and density of renal sympathetic nerves.
Figure 1. Distribution and density of renal sympathetic nerves. Distribution of nerves stratified according to total number (each green dot represents 10 nerves), relative number as percent per segment, and distance from the lumen in the proximal (A), middle (B), and distal (C) location.
Sakakura et al and Mahfoud et al showed that the concentration of sympathetic periarterial renal nerves is higher in the proximal and ventral areas but closer to the lumen in the distal segment (Figure 1).13,14 Moreover, Id et al15 found that ablating nerves in the renal arteries without addressing accessory arteries resulted in less-optimal blood pressure reduction. Thus, the technical aspects of the procedure are highly important.

Advanced renal denervation catheters are needed that are multielectrode, smaller, easier to manipulate, and capable of providing simultaneous, circumferential, more-intense, and deeper ablations. The ongoing Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPIRED)16 and Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE-HTN: REINFORCE)17 trials are using contemporary innovative ablation catheters to address the limitations of the first-generation Symplicity catheter.

Further, Fischell et al18 reported encouraging results of renal denervation performed by injecting ethanol into the adventitial space of the renal arteries. This is still an invasive procedure; however, ethanol can spread out in all directions and reach all targeted nerves, potentially resulting in a more complete renal artery sympathetic ablation.

As technology advances, the WAVE IV trial19 is examining renal denervation performed from the outside through the skin using high-intensity focused ultrasound, which eliminates the need for femoral arterial catheterization, a promising noninvasive approach.

Proposals for future trials

The European Clinical Consensus Conference for Renal Denervation20 proposed that future trials of renal denervation include patients with moderate rather than resistant hypertension, reflecting the pathogenic importance of sympathetic activity in earlier stages of hypertension. The conference also proposed excluding patients with stiff large arteries, a cause of isolated systolic hypertension. Other proposals included standardizing concomitant antihypertensive therapy, preferably treating all patients with the combination of a renin-angiotensin system blocker, calcium channel blocker, and diuretic in the run-in period; monitoring drug adherence through the use of pill counts, electronic pill dispensers, and drug blood tests; and using change in ambulatory blood pressure as the primary efficacy end point and change in office blood pressure as a secondary end point.

Trials ongoing

To possibly address the limitations posed by the SYMPLICITY HTN-3 trial and to answer other important questions, several sham-controlled clinical trials of renal denervation are currently being conducted:

  • INSPiRED16
  • REDUCE-HTN: REINFORCE17
  • Spyral HTN-Off Med21
  • Spyral HTN-On Med21
  • Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN).22

We hope these new studies can more clearly identify subsets of patients who would benefit from this technology, determine predictors of blood pressure reduction in such patients, and lead to newer devices that may provide more complete ablation.

Obviously, we also need better ways to identify the exact location of these sympathetic nerves within the renal artery and have a clearer sense of procedural success.

Until then, our colleagues in Europe and Australia continue to treat patients with this technology as we appropriately and patiently wait for level 1 clinical evidence of its efficacy.


Acknowledgments: We thank Kathryn Brock, BA, Editorial Services Manager, Heart and Vascular Institute, Cleveland Clinic, for her assistance in the preparation of this paper.

References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al, for the SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Symplicity HTN-2 Investigators, Esler MD, Krum H, Sobotka PA, Schlaich MP, Schmieder RE, Bohm M. Renal sympathetic denervation in patients with treatment-resistant hypertension (the Symplicity HTN-2 trial): a randomised controlled trial. Lancet 2010; 376:1903–1909.
  3. Bunte MC, Infante de Oliveira E, Shishehbor MH. Endovascular treatment of resistant and uncontrolled hypertension: therapies on the horizon. JACC Cardiovasc Interv 2013; 6:1–9.
  4. Thomas G, Shishehbor MH, Bravo EL, Nally JV. Renal denervation to treat resistant hypertension: guarded optimism. Cleve Clin J Med 2012; 79:501–510.
  5. Shishehbor MH, Bunte MC. Anatomical exclusion for renal denervation: are we putting the cart before the horse? JACC Cardiovasc Interv 2014; 7:193–194.
  6. Bhatt DL, Bakris GL. The promise of renal denervation. Cleve Clin J Med 2012; 79:498–500.
  7. Bunte MC. Renal sympathetic denervation for refractory hypertension. Lancet 2011; 377:1074; author reply 1075.
  8. Mahfoud F, Luscher TF, Andersson B, et al; European Society of Cardiology. Expert consensus document from the European Society of Cardiology on catheter-based renal denervation. Eur Heart J 2013; 34:2149–2157.
  9. Schlaich MP, Schmieder RE, Bakris G, et al. International expert consensus statement: percutaneous transluminal renal denervation for the treatment of resistant hypertension. J Am Coll Cardiol 2013; 62:2031–2045.
  10. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  11. Rosa J, Widimsky P, Tousek P, et al. Randomized comparison of renal denervation versus intensified pharmacotherapy including spironolactone in true-resistant hypertension: six-month results from the Prague-15 study. Hypertension 2015; 65:407–413.
  12. Williams B, MacDonald TM, Morant S, et al; British Hypertension Society’s PATHWAY Studies Group. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet 2015; 386:2059–2068.
  13. Sakakura K, Ladich E, Cheng Q, et al. Anatomic assessment of sympathetic peri-arterial renal nerves in man. J Am Coll Cardiol 2014; 64:635–643.
  14. Mahfoud F, Edelman ER, Bohm M. Catheter-based renal denervation is no simple matter: lessons to be learned from our anatomy? J Am Coll Cardiol 2014; 64:644–646.
  15. Id D, Kaltenbach B, Bertog SC, et al. Does the presence of accessory renal arteries affect the efficacy of renal denervation? JACC Cardiovasc Interv 2013; 6:1085–1091.
  16. Jin Y, Jacobs L, Baelen M, et al; Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (Inspired) Investigators. Rationale and design of the Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPiRED) trial. Blood Press 2014; 23:138–146.
  17. ClinicalTrialsgov. Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE HTN: REINFORCE). https://clinicaltrials.gov/ct2/show/NCT02392351?term=REDUCE-HTN%3A+REINFORCE&rank=1. Accessed August 3, 2017.
  18. Fischell TA, Ebner A, Gallo S, et al. Transcatheter alcohol-mediated perivascular renal denervation with the peregrine system: first-in-human experience. JACC Cardiovasc Interv 2016; 9:589–598.
  19. ClinicalTrialsgov. Sham controlled study of renal denervation for subjects with uncontrolled hypertension (WAVE_IV) (NCT02029885). https://clinicaltrials.gov/ct2/show/results/NCT02029885. Accessed August 3, 2017.
  20. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European clinical consensus conference for renal denervation: considerations on future clinical trial design. Eur Heart J 2015; 36:2219–2227.
  21. Kandzari DE, Kario K, Mahfoud F, et al. The SPYRAL HTN Global Clinical Trial Program: rationale and design for studies of renal denervation in the absence (SPYRAL HTN OFF-MED) and presence (SPYRAL HTN ON-MED) of antihypertensive medications. Am Heart J 2016; 171:82–91.
  22. ClinicalTrialsgov. A Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN). https://clinicaltrials.gov/ct2/show/NCT02649426?term=RADIANCE&rank=3. Accessed August 3, 2017.
Article PDF
Author and Disclosure Information

Mehdi H. Shishehbor, DO, MPH, PhD
Professor of Medicine, Case Western Reserve University, Cleveland, OH; Co-Chair, Harring Heart and Vascular Institute; Director, Cardiovascular Interventional Center; Co-Director, Vascular Center, University Hospitals of Cleveland, OH; Site Principal Investigator, SYMPLICITY HTN-3 trial

Tarek A. Hammad, MD
Department of Medicine, Division of Cardiology, The University of Texas Health Center at San Antonio

George Thomas, MD, MPH
Director, Center for Blood Pressure Disorders, Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Investigator, SYMPLICITY HTN-3 trial

Address: Mehdi H. Shishehbor, DO, MPH, PhD, University Hospitals of Cleveland, 11100 Euclid Avenue, Lakeside, 3rd Floor, Cleveland, OH 44107; [email protected]

Issue
Cleveland Clinic Journal of Medicine - 84(9)
Publications
Topics
Page Number
681-686
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Mehdi Shishehbor, Tarek Hammad, George Thomas
Sections
Author and Disclosure Information

Mehdi H. Shishehbor, DO, MPH, PhD
Professor of Medicine, Case Western Reserve University, Cleveland, OH; Co-Chair, Harring Heart and Vascular Institute; Director, Cardiovascular Interventional Center; Co-Director, Vascular Center, University Hospitals of Cleveland, OH; Site Principal Investigator, SYMPLICITY HTN-3 trial

Tarek A. Hammad, MD
Department of Medicine, Division of Cardiology, The University of Texas Health Center at San Antonio

George Thomas, MD, MPH
Director, Center for Blood Pressure Disorders, Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Investigator, SYMPLICITY HTN-3 trial

Address: Mehdi H. Shishehbor, DO, MPH, PhD, University Hospitals of Cleveland, 11100 Euclid Avenue, Lakeside, 3rd Floor, Cleveland, OH 44107; [email protected]

Author and Disclosure Information

Mehdi H. Shishehbor, DO, MPH, PhD
Professor of Medicine, Case Western Reserve University, Cleveland, OH; Co-Chair, Harring Heart and Vascular Institute; Director, Cardiovascular Interventional Center; Co-Director, Vascular Center, University Hospitals of Cleveland, OH; Site Principal Investigator, SYMPLICITY HTN-3 trial

Tarek A. Hammad, MD
Department of Medicine, Division of Cardiology, The University of Texas Health Center at San Antonio

George Thomas, MD, MPH
Director, Center for Blood Pressure Disorders, Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Investigator, SYMPLICITY HTN-3 trial

Address: Mehdi H. Shishehbor, DO, MPH, PhD, University Hospitals of Cleveland, 11100 Euclid Avenue, Lakeside, 3rd Floor, Cleveland, OH 44107; [email protected]

Article PDF
Article PDF
Related Articles

Many patients, clinicians, and researchers had hoped that renal denervation would help control resistant hypertension. However, in the SYMPLICITY HTN-3 trial,1 named for the catheter-based system used in the study (Symplicity RDN, Medtronic, Dublin, Ireland), this endovascular procedure failed to meet its primary and secondary efficacy end points, although it was found to be safe. These results were surprising, especially given the results of an earlier randomized trial (SYMPLICITY HTN-2),2 which showed larger reductions in blood pressures 6 months after denervation than in the current trial.

See related editorial

Here, we discuss the results of the SYMPLICITY HTN-3 trial and offer possible explanations for its negative outcomes.

LEAD-UP TO SYMPLICITY HTN-3

Renal denervation consists of passing a catheter through the femoral artery into the renal arteries and ablating their sympathetic nerves using radiofrequency energy. In theory, this should interrupt efferent sympathetic communication between the brain and renal arteries, reducing muscular contraction of these arteries, increasing renal blood flow, reducing activation of the renin-angiotensin-adosterone system, thus reducing sodium retention, reducing afferent sympathetic communication between the kidneys and brain, and in turn reducing further sympathetic activity elsewhere in the body, such as in the heart. Blood pressure should fall.3

The results of the SYMPLICITY HTN-1 and 2 trials were discussed in an earlier article in this Journal,3 and the Medtronic-Ardian renal denervation system has been available in Europe and Australia for clinical use for over 2 years.4 Indeed, after the SYMPLICITY HTN-2 results were published in 2010, Boston Scientific’s Vessix, St. Jude Medical’s EnligHTN, and Covidien’s OneShot radiofrequency renal denervation devices—albeit each with some modifications—received a Conformité Européene (CE) mark and became available in Europe and Australia for clinical use. These devices are not available for clinical use or research in the United States.3,5

Therefore, SYMPLICITY HTN-3, sponsored by Medtronic, was designed to obtain US Food and Drug Administration approval in the United States.6

SYMPLICITY HTN-3 DESIGN

Inclusion criteria were similar to those in the earlier SYMPLICITY trials. Patients had to have resistant hypertension, defined as a systolic blood pressure ≥ 160 mm Hg despite taking at least 3 blood pressure medications at maximum tolerated doses. Patients were excluded if they had a glomerular filtration rate of less than 45 mL/min/1.73 m2, renal artery stenosis, or known secondary hypertension.

A total of 1,441 patients were enrolled, of whom 364 were eventually randomized to undergo renal denervation, and 171 were randomized to undergo a sham procedure. The mean systolic blood pressure at baseline was 188 mm Hg in each group. Most patients were taking maximum doses of blood pressure medications, and almost one-fourth were taking an aldosterone antagonist. Patients in both groups were taking an average of 5 medications.

The 2 groups were well matched for important covariates, including obstructive sleep apnea, diabetes mellitus, and renal insufficiency. Most of the patients were white; 25% of the renal denervation group and 29% of the sham procedure group were black.

The physicians conducting the follow-up appointments did not know which procedure the patients underwent, and neither did the patients. Medications were closely monitored, and patients had close follow-up. The catheter (Symplicity RDS, Medtronic) was of the same design that was used in the earlier SYMPLICITY trials and in clinical practice in countries where renal denervation was available.

Researchers expected that the systolic blood pressure, as measured in the office, would fall in both groups, but they hoped it would fall farther in the denervation group—at least 5 mm Hg farther, the primary end point of the trial. The secondary effectiveness end point was a 2-mm Hg greater reduction in 24-hour ambulatory systolic blood pressure.

 

 

SYMPLICITY HTN-3 RESULTS

No statistically significant difference in safety was observed between the denervation and control groups. However, the procedure was associated with 1 embolic event and 1 case of renal artery stenosis.

Blood pressure fell in both groups. However, at 6 months, office systolic pressure had fallen by a mean of 14.13 mm Hg in the denervation group and 11.74 mm Hg in the sham procedure group, a difference of only 2.39 mm Hg. The mean ambulatory systolic blood pressure had fallen by 6.75 vs 4.79 mm Hg, a difference of only 1.96 mm Hg. Neither difference was statistically significant.

A number of prespecified subgroup analyses were conducted, but the benefit of the procedure was statistically significant in only 3 subgroups: patients who were not black (P = .01), patients who were less than 65 years old (P = .04), and patients who had an estimated glomerular filtration rate of 60 mL/min/1.73 m2 or higher (P = .05).

WHAT WENT WRONG?

The results of SYMPLICITY HTN-3 were disappointing and led companies that were developing renal denervation devices to discontinue or reevaluate their programs.

Although the results were surprising, many observers (including our group) raised concerns about the initial enthusiasm surrounding renal denervation.3–7 Indeed, in 2010, we had concerns about the discrepancy between office-based blood pressure measurements (the primary end point of all renal denervation trials) and ambulatory blood pressure measurements in SYMPLICITY HTN-2.7

The enthusiasm surrounding this procedure led to the publication of 2 consensus documents on this novel therapy based on only 1 small randomized controlled study (SYMPLICITY HTN-2).8,9 Renal denervation was even reported to be useful in other conditions involving the sympathorenal axis, including diabetes mellitus, metabolic syndrome, and obstructive sleep apnea, and also as a potential treatment adjunct in atrial fibrillation and other arrhythmias.5

What went wrong?

Shortcomings in trial design?

The trial was well designed. Both patients and operators were blinded to the procedure, and 24-hour ambulatory blood pressure monitoring was used. We presume that appropriate patients with resistant hypertension were enrolled—the mean baseline systolic blood pressure was 188 mm Hg, and patients in each group were taking an average of 5 medications.

On the other hand, true medication adherence is difficult to ascertain. Further, the term maximal “tolerated” doses of medications is vague, and we cannot rule out the possibility that some patients were enrolled who did not truly have resistant hypertension—they simply did not want to take medications.

Patients were required to be on a stable medication regimen before enrollment and, ideally, to not have any medication changes during the course of the study, but at least 40% of patients did require medication changes during the study. Additionally, it is unclear whether all patients underwent specific testing to rule out secondary hypertension, as this was done at the discretion of the treating physician.

First-generation catheters?

The same type of catheter was used as in the earlier SYMPLICITY trials, and it had been used in many patients in clinical practice in countries where the catheter is routinely available. It is unknown, however, whether newer multisite denervation devices would yield better results than the first-generation devices used in SYMPLICITY HTN-3. But even this would not explain the discrepancies in data between earlier trials and this trial.

Operator inexperience?

It has been suggested that operator inexperience may have played a role, but an analysis of operator volume did not find any association between this variable and the outcomes. Each procedure was supervised by at least 1 and in most cases 2 certified Medtronic representatives, who made certain that meticulous attention was paid to procedure details and that no shortcuts were taken during the procedure.

Inadequate ablation?

While we can assume that the correct technique was followed in most cases, renal denervation is still a “blind” procedure, and there is no nerve mapping to ascertain the degree of ablation achieved. Notably, patients who had the most ablations reportedly had a greater average drop in systolic ambulatory blood pressure than those who received fewer ablations. Sympathetic nervous system activity is a potential marker of adequacy of ablation, but it was not routinely assessed in the SYMPLICITY HTN-3 trial. Techniques to assess sympathetic nerve activity such as norepinephrine spillover and muscle sympathetic nerve activity are highly specialized and available only at a few research centers, and are not available for routine clinical use.

While these points may explain the negative findings of this trial, they fail to account for the discrepant results between this study and previous trials that used exactly the same definitions and techniques.

 

 

Patient demographics?

Is it possible that renal denervation has a differential effect according to race? All previous renal denervation studies were conducted in Europe or Australia; therefore, few data are available on the efficacy of the procedure in other racial groups, such as black Americans. Most of the patients in this trial were white, but approximately 25% were black—a good representation. There was a statistically significant benefit favoring renal denervation in nonblack (mostly white) patients, but not in black patients. This may be related to racial differences in the pathophysiology of hypertension or possibly due to chance alone.

A Hawthorne effect?

A Hawthorne effect (patients being more compliant because physicians are paying more attention to them) is unlikely, since the renal denervation arm did not have any reduction in blood pressure medications. At 6 months, both the sham group and the procedure group were still on an average of 5 medications.

Additionally, while the blood pressure reduction in both treatment groups was significant, the systolic blood pressure at 6 months was still 166 mm Hg in the denervation group and 168 mm Hg in the sham group. If denervation was effective, one would have expected a greater reduction in blood pressure or at least a decrease in the number of medications needed, eg, 1 to 2 fewer medications in the denervation group compared with the sham procedure group.

Regression to the mean?

It is unknown whether the results represent a statistical error such as regression to the mean. But given the run-in period and the confirmatory data from 24-hour ambulatory blood pressure, this would be unlikely.

WHAT NOW?

Is renal denervation dead? SYMPLICITY HTN-3 is only a single trial with multiple shortcomings and lessons to learn from. Since its publication, there have been updates from 2 prospective, randomized, open-label trials concerning the efficacy of catheter-based renal denervation in lowering blood pressure.10,11

DENERHTN (Renal Denervation for Hypertension)10 studied patients with ambulatory systolic blood pressure higher than 135 mm Hg, diastolic blood pressure higher than 80 mm Hg, or both (after excluding secondary etiologies), despite 4 weeks of standardized triple-drug treatment including a diuretic. Patients were randomized to standardized stepped-care antihypertensive treatment alone (control group) or standard care plus renal denervation. The latter resulted in a significant further reduction in ambulatory blood pressure at 6 months.

The Prague-15 trial11 studied patients with resistant hypertension. Secondary etiologies were excluded and adherence to therapy was confirmed by measuring plasma medication levels. It showed that renal denervation along with optimal antihypertensive medical therapy (unchanged after randomization) resulted in a significant reduction in ambulatory blood pressure that was comparable to the effect of intensified antihypertensive medical therapy including spironolactone. (Studies have shown that spironolactone is effective when added on as a fourth-line medication in resistant hypertension.12) At 6 months, patients in the intensive medical therapy group were using an average of 0.3 more antihypertensive medications than those in the procedure group.

These two trials addressed some of the drawbacks of the SYMPLICITY HTN-3 trial. However, both have many limitations including and not limited to being open-label and nonblinded, lacking a sham procedure, using a lower blood pressure threshold than SYMPLICITY HTN-3 did to define resistant hypertension, and using the same catheter as in the SYMPLICITY trials.

 

 

Better technology is coming

Distribution and density of renal sympathetic nerves.
Figure 1. Distribution and density of renal sympathetic nerves. Distribution of nerves stratified according to total number (each green dot represents 10 nerves), relative number as percent per segment, and distance from the lumen in the proximal (A), middle (B), and distal (C) location.
Sakakura et al and Mahfoud et al showed that the concentration of sympathetic periarterial renal nerves is higher in the proximal and ventral areas but closer to the lumen in the distal segment (Figure 1).13,14 Moreover, Id et al15 found that ablating nerves in the renal arteries without addressing accessory arteries resulted in less-optimal blood pressure reduction. Thus, the technical aspects of the procedure are highly important.

Advanced renal denervation catheters are needed that are multielectrode, smaller, easier to manipulate, and capable of providing simultaneous, circumferential, more-intense, and deeper ablations. The ongoing Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPIRED)16 and Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE-HTN: REINFORCE)17 trials are using contemporary innovative ablation catheters to address the limitations of the first-generation Symplicity catheter.

Further, Fischell et al18 reported encouraging results of renal denervation performed by injecting ethanol into the adventitial space of the renal arteries. This is still an invasive procedure; however, ethanol can spread out in all directions and reach all targeted nerves, potentially resulting in a more complete renal artery sympathetic ablation.

As technology advances, the WAVE IV trial19 is examining renal denervation performed from the outside through the skin using high-intensity focused ultrasound, which eliminates the need for femoral arterial catheterization, a promising noninvasive approach.

Proposals for future trials

The European Clinical Consensus Conference for Renal Denervation20 proposed that future trials of renal denervation include patients with moderate rather than resistant hypertension, reflecting the pathogenic importance of sympathetic activity in earlier stages of hypertension. The conference also proposed excluding patients with stiff large arteries, a cause of isolated systolic hypertension. Other proposals included standardizing concomitant antihypertensive therapy, preferably treating all patients with the combination of a renin-angiotensin system blocker, calcium channel blocker, and diuretic in the run-in period; monitoring drug adherence through the use of pill counts, electronic pill dispensers, and drug blood tests; and using change in ambulatory blood pressure as the primary efficacy end point and change in office blood pressure as a secondary end point.

Trials ongoing

To possibly address the limitations posed by the SYMPLICITY HTN-3 trial and to answer other important questions, several sham-controlled clinical trials of renal denervation are currently being conducted:

  • INSPiRED16
  • REDUCE-HTN: REINFORCE17
  • Spyral HTN-Off Med21
  • Spyral HTN-On Med21
  • Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN).22

We hope these new studies can more clearly identify subsets of patients who would benefit from this technology, determine predictors of blood pressure reduction in such patients, and lead to newer devices that may provide more complete ablation.

Obviously, we also need better ways to identify the exact location of these sympathetic nerves within the renal artery and have a clearer sense of procedural success.

Until then, our colleagues in Europe and Australia continue to treat patients with this technology as we appropriately and patiently wait for level 1 clinical evidence of its efficacy.


Acknowledgments: We thank Kathryn Brock, BA, Editorial Services Manager, Heart and Vascular Institute, Cleveland Clinic, for her assistance in the preparation of this paper.

Many patients, clinicians, and researchers had hoped that renal denervation would help control resistant hypertension. However, in the SYMPLICITY HTN-3 trial,1 named for the catheter-based system used in the study (Symplicity RDN, Medtronic, Dublin, Ireland), this endovascular procedure failed to meet its primary and secondary efficacy end points, although it was found to be safe. These results were surprising, especially given the results of an earlier randomized trial (SYMPLICITY HTN-2),2 which showed larger reductions in blood pressures 6 months after denervation than in the current trial.

See related editorial

Here, we discuss the results of the SYMPLICITY HTN-3 trial and offer possible explanations for its negative outcomes.

LEAD-UP TO SYMPLICITY HTN-3

Renal denervation consists of passing a catheter through the femoral artery into the renal arteries and ablating their sympathetic nerves using radiofrequency energy. In theory, this should interrupt efferent sympathetic communication between the brain and renal arteries, reducing muscular contraction of these arteries, increasing renal blood flow, reducing activation of the renin-angiotensin-adosterone system, thus reducing sodium retention, reducing afferent sympathetic communication between the kidneys and brain, and in turn reducing further sympathetic activity elsewhere in the body, such as in the heart. Blood pressure should fall.3

The results of the SYMPLICITY HTN-1 and 2 trials were discussed in an earlier article in this Journal,3 and the Medtronic-Ardian renal denervation system has been available in Europe and Australia for clinical use for over 2 years.4 Indeed, after the SYMPLICITY HTN-2 results were published in 2010, Boston Scientific’s Vessix, St. Jude Medical’s EnligHTN, and Covidien’s OneShot radiofrequency renal denervation devices—albeit each with some modifications—received a Conformité Européene (CE) mark and became available in Europe and Australia for clinical use. These devices are not available for clinical use or research in the United States.3,5

Therefore, SYMPLICITY HTN-3, sponsored by Medtronic, was designed to obtain US Food and Drug Administration approval in the United States.6

SYMPLICITY HTN-3 DESIGN

Inclusion criteria were similar to those in the earlier SYMPLICITY trials. Patients had to have resistant hypertension, defined as a systolic blood pressure ≥ 160 mm Hg despite taking at least 3 blood pressure medications at maximum tolerated doses. Patients were excluded if they had a glomerular filtration rate of less than 45 mL/min/1.73 m2, renal artery stenosis, or known secondary hypertension.

A total of 1,441 patients were enrolled, of whom 364 were eventually randomized to undergo renal denervation, and 171 were randomized to undergo a sham procedure. The mean systolic blood pressure at baseline was 188 mm Hg in each group. Most patients were taking maximum doses of blood pressure medications, and almost one-fourth were taking an aldosterone antagonist. Patients in both groups were taking an average of 5 medications.

The 2 groups were well matched for important covariates, including obstructive sleep apnea, diabetes mellitus, and renal insufficiency. Most of the patients were white; 25% of the renal denervation group and 29% of the sham procedure group were black.

The physicians conducting the follow-up appointments did not know which procedure the patients underwent, and neither did the patients. Medications were closely monitored, and patients had close follow-up. The catheter (Symplicity RDS, Medtronic) was of the same design that was used in the earlier SYMPLICITY trials and in clinical practice in countries where renal denervation was available.

Researchers expected that the systolic blood pressure, as measured in the office, would fall in both groups, but they hoped it would fall farther in the denervation group—at least 5 mm Hg farther, the primary end point of the trial. The secondary effectiveness end point was a 2-mm Hg greater reduction in 24-hour ambulatory systolic blood pressure.

 

 

SYMPLICITY HTN-3 RESULTS

No statistically significant difference in safety was observed between the denervation and control groups. However, the procedure was associated with 1 embolic event and 1 case of renal artery stenosis.

Blood pressure fell in both groups. However, at 6 months, office systolic pressure had fallen by a mean of 14.13 mm Hg in the denervation group and 11.74 mm Hg in the sham procedure group, a difference of only 2.39 mm Hg. The mean ambulatory systolic blood pressure had fallen by 6.75 vs 4.79 mm Hg, a difference of only 1.96 mm Hg. Neither difference was statistically significant.

A number of prespecified subgroup analyses were conducted, but the benefit of the procedure was statistically significant in only 3 subgroups: patients who were not black (P = .01), patients who were less than 65 years old (P = .04), and patients who had an estimated glomerular filtration rate of 60 mL/min/1.73 m2 or higher (P = .05).

WHAT WENT WRONG?

The results of SYMPLICITY HTN-3 were disappointing and led companies that were developing renal denervation devices to discontinue or reevaluate their programs.

Although the results were surprising, many observers (including our group) raised concerns about the initial enthusiasm surrounding renal denervation.3–7 Indeed, in 2010, we had concerns about the discrepancy between office-based blood pressure measurements (the primary end point of all renal denervation trials) and ambulatory blood pressure measurements in SYMPLICITY HTN-2.7

The enthusiasm surrounding this procedure led to the publication of 2 consensus documents on this novel therapy based on only 1 small randomized controlled study (SYMPLICITY HTN-2).8,9 Renal denervation was even reported to be useful in other conditions involving the sympathorenal axis, including diabetes mellitus, metabolic syndrome, and obstructive sleep apnea, and also as a potential treatment adjunct in atrial fibrillation and other arrhythmias.5

What went wrong?

Shortcomings in trial design?

The trial was well designed. Both patients and operators were blinded to the procedure, and 24-hour ambulatory blood pressure monitoring was used. We presume that appropriate patients with resistant hypertension were enrolled—the mean baseline systolic blood pressure was 188 mm Hg, and patients in each group were taking an average of 5 medications.

On the other hand, true medication adherence is difficult to ascertain. Further, the term maximal “tolerated” doses of medications is vague, and we cannot rule out the possibility that some patients were enrolled who did not truly have resistant hypertension—they simply did not want to take medications.

Patients were required to be on a stable medication regimen before enrollment and, ideally, to not have any medication changes during the course of the study, but at least 40% of patients did require medication changes during the study. Additionally, it is unclear whether all patients underwent specific testing to rule out secondary hypertension, as this was done at the discretion of the treating physician.

First-generation catheters?

The same type of catheter was used as in the earlier SYMPLICITY trials, and it had been used in many patients in clinical practice in countries where the catheter is routinely available. It is unknown, however, whether newer multisite denervation devices would yield better results than the first-generation devices used in SYMPLICITY HTN-3. But even this would not explain the discrepancies in data between earlier trials and this trial.

Operator inexperience?

It has been suggested that operator inexperience may have played a role, but an analysis of operator volume did not find any association between this variable and the outcomes. Each procedure was supervised by at least 1 and in most cases 2 certified Medtronic representatives, who made certain that meticulous attention was paid to procedure details and that no shortcuts were taken during the procedure.

Inadequate ablation?

While we can assume that the correct technique was followed in most cases, renal denervation is still a “blind” procedure, and there is no nerve mapping to ascertain the degree of ablation achieved. Notably, patients who had the most ablations reportedly had a greater average drop in systolic ambulatory blood pressure than those who received fewer ablations. Sympathetic nervous system activity is a potential marker of adequacy of ablation, but it was not routinely assessed in the SYMPLICITY HTN-3 trial. Techniques to assess sympathetic nerve activity such as norepinephrine spillover and muscle sympathetic nerve activity are highly specialized and available only at a few research centers, and are not available for routine clinical use.

While these points may explain the negative findings of this trial, they fail to account for the discrepant results between this study and previous trials that used exactly the same definitions and techniques.

 

 

Patient demographics?

Is it possible that renal denervation has a differential effect according to race? All previous renal denervation studies were conducted in Europe or Australia; therefore, few data are available on the efficacy of the procedure in other racial groups, such as black Americans. Most of the patients in this trial were white, but approximately 25% were black—a good representation. There was a statistically significant benefit favoring renal denervation in nonblack (mostly white) patients, but not in black patients. This may be related to racial differences in the pathophysiology of hypertension or possibly due to chance alone.

A Hawthorne effect?

A Hawthorne effect (patients being more compliant because physicians are paying more attention to them) is unlikely, since the renal denervation arm did not have any reduction in blood pressure medications. At 6 months, both the sham group and the procedure group were still on an average of 5 medications.

Additionally, while the blood pressure reduction in both treatment groups was significant, the systolic blood pressure at 6 months was still 166 mm Hg in the denervation group and 168 mm Hg in the sham group. If denervation was effective, one would have expected a greater reduction in blood pressure or at least a decrease in the number of medications needed, eg, 1 to 2 fewer medications in the denervation group compared with the sham procedure group.

Regression to the mean?

It is unknown whether the results represent a statistical error such as regression to the mean. But given the run-in period and the confirmatory data from 24-hour ambulatory blood pressure, this would be unlikely.

WHAT NOW?

Is renal denervation dead? SYMPLICITY HTN-3 is only a single trial with multiple shortcomings and lessons to learn from. Since its publication, there have been updates from 2 prospective, randomized, open-label trials concerning the efficacy of catheter-based renal denervation in lowering blood pressure.10,11

DENERHTN (Renal Denervation for Hypertension)10 studied patients with ambulatory systolic blood pressure higher than 135 mm Hg, diastolic blood pressure higher than 80 mm Hg, or both (after excluding secondary etiologies), despite 4 weeks of standardized triple-drug treatment including a diuretic. Patients were randomized to standardized stepped-care antihypertensive treatment alone (control group) or standard care plus renal denervation. The latter resulted in a significant further reduction in ambulatory blood pressure at 6 months.

The Prague-15 trial11 studied patients with resistant hypertension. Secondary etiologies were excluded and adherence to therapy was confirmed by measuring plasma medication levels. It showed that renal denervation along with optimal antihypertensive medical therapy (unchanged after randomization) resulted in a significant reduction in ambulatory blood pressure that was comparable to the effect of intensified antihypertensive medical therapy including spironolactone. (Studies have shown that spironolactone is effective when added on as a fourth-line medication in resistant hypertension.12) At 6 months, patients in the intensive medical therapy group were using an average of 0.3 more antihypertensive medications than those in the procedure group.

These two trials addressed some of the drawbacks of the SYMPLICITY HTN-3 trial. However, both have many limitations including and not limited to being open-label and nonblinded, lacking a sham procedure, using a lower blood pressure threshold than SYMPLICITY HTN-3 did to define resistant hypertension, and using the same catheter as in the SYMPLICITY trials.

 

 

Better technology is coming

Distribution and density of renal sympathetic nerves.
Figure 1. Distribution and density of renal sympathetic nerves. Distribution of nerves stratified according to total number (each green dot represents 10 nerves), relative number as percent per segment, and distance from the lumen in the proximal (A), middle (B), and distal (C) location.
Sakakura et al and Mahfoud et al showed that the concentration of sympathetic periarterial renal nerves is higher in the proximal and ventral areas but closer to the lumen in the distal segment (Figure 1).13,14 Moreover, Id et al15 found that ablating nerves in the renal arteries without addressing accessory arteries resulted in less-optimal blood pressure reduction. Thus, the technical aspects of the procedure are highly important.

Advanced renal denervation catheters are needed that are multielectrode, smaller, easier to manipulate, and capable of providing simultaneous, circumferential, more-intense, and deeper ablations. The ongoing Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPIRED)16 and Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE-HTN: REINFORCE)17 trials are using contemporary innovative ablation catheters to address the limitations of the first-generation Symplicity catheter.

Further, Fischell et al18 reported encouraging results of renal denervation performed by injecting ethanol into the adventitial space of the renal arteries. This is still an invasive procedure; however, ethanol can spread out in all directions and reach all targeted nerves, potentially resulting in a more complete renal artery sympathetic ablation.

As technology advances, the WAVE IV trial19 is examining renal denervation performed from the outside through the skin using high-intensity focused ultrasound, which eliminates the need for femoral arterial catheterization, a promising noninvasive approach.

Proposals for future trials

The European Clinical Consensus Conference for Renal Denervation20 proposed that future trials of renal denervation include patients with moderate rather than resistant hypertension, reflecting the pathogenic importance of sympathetic activity in earlier stages of hypertension. The conference also proposed excluding patients with stiff large arteries, a cause of isolated systolic hypertension. Other proposals included standardizing concomitant antihypertensive therapy, preferably treating all patients with the combination of a renin-angiotensin system blocker, calcium channel blocker, and diuretic in the run-in period; monitoring drug adherence through the use of pill counts, electronic pill dispensers, and drug blood tests; and using change in ambulatory blood pressure as the primary efficacy end point and change in office blood pressure as a secondary end point.

Trials ongoing

To possibly address the limitations posed by the SYMPLICITY HTN-3 trial and to answer other important questions, several sham-controlled clinical trials of renal denervation are currently being conducted:

  • INSPiRED16
  • REDUCE-HTN: REINFORCE17
  • Spyral HTN-Off Med21
  • Spyral HTN-On Med21
  • Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN).22

We hope these new studies can more clearly identify subsets of patients who would benefit from this technology, determine predictors of blood pressure reduction in such patients, and lead to newer devices that may provide more complete ablation.

Obviously, we also need better ways to identify the exact location of these sympathetic nerves within the renal artery and have a clearer sense of procedural success.

Until then, our colleagues in Europe and Australia continue to treat patients with this technology as we appropriately and patiently wait for level 1 clinical evidence of its efficacy.


Acknowledgments: We thank Kathryn Brock, BA, Editorial Services Manager, Heart and Vascular Institute, Cleveland Clinic, for her assistance in the preparation of this paper.

References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al, for the SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Symplicity HTN-2 Investigators, Esler MD, Krum H, Sobotka PA, Schlaich MP, Schmieder RE, Bohm M. Renal sympathetic denervation in patients with treatment-resistant hypertension (the Symplicity HTN-2 trial): a randomised controlled trial. Lancet 2010; 376:1903–1909.
  3. Bunte MC, Infante de Oliveira E, Shishehbor MH. Endovascular treatment of resistant and uncontrolled hypertension: therapies on the horizon. JACC Cardiovasc Interv 2013; 6:1–9.
  4. Thomas G, Shishehbor MH, Bravo EL, Nally JV. Renal denervation to treat resistant hypertension: guarded optimism. Cleve Clin J Med 2012; 79:501–510.
  5. Shishehbor MH, Bunte MC. Anatomical exclusion for renal denervation: are we putting the cart before the horse? JACC Cardiovasc Interv 2014; 7:193–194.
  6. Bhatt DL, Bakris GL. The promise of renal denervation. Cleve Clin J Med 2012; 79:498–500.
  7. Bunte MC. Renal sympathetic denervation for refractory hypertension. Lancet 2011; 377:1074; author reply 1075.
  8. Mahfoud F, Luscher TF, Andersson B, et al; European Society of Cardiology. Expert consensus document from the European Society of Cardiology on catheter-based renal denervation. Eur Heart J 2013; 34:2149–2157.
  9. Schlaich MP, Schmieder RE, Bakris G, et al. International expert consensus statement: percutaneous transluminal renal denervation for the treatment of resistant hypertension. J Am Coll Cardiol 2013; 62:2031–2045.
  10. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  11. Rosa J, Widimsky P, Tousek P, et al. Randomized comparison of renal denervation versus intensified pharmacotherapy including spironolactone in true-resistant hypertension: six-month results from the Prague-15 study. Hypertension 2015; 65:407–413.
  12. Williams B, MacDonald TM, Morant S, et al; British Hypertension Society’s PATHWAY Studies Group. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet 2015; 386:2059–2068.
  13. Sakakura K, Ladich E, Cheng Q, et al. Anatomic assessment of sympathetic peri-arterial renal nerves in man. J Am Coll Cardiol 2014; 64:635–643.
  14. Mahfoud F, Edelman ER, Bohm M. Catheter-based renal denervation is no simple matter: lessons to be learned from our anatomy? J Am Coll Cardiol 2014; 64:644–646.
  15. Id D, Kaltenbach B, Bertog SC, et al. Does the presence of accessory renal arteries affect the efficacy of renal denervation? JACC Cardiovasc Interv 2013; 6:1085–1091.
  16. Jin Y, Jacobs L, Baelen M, et al; Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (Inspired) Investigators. Rationale and design of the Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPiRED) trial. Blood Press 2014; 23:138–146.
  17. ClinicalTrialsgov. Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE HTN: REINFORCE). https://clinicaltrials.gov/ct2/show/NCT02392351?term=REDUCE-HTN%3A+REINFORCE&rank=1. Accessed August 3, 2017.
  18. Fischell TA, Ebner A, Gallo S, et al. Transcatheter alcohol-mediated perivascular renal denervation with the peregrine system: first-in-human experience. JACC Cardiovasc Interv 2016; 9:589–598.
  19. ClinicalTrialsgov. Sham controlled study of renal denervation for subjects with uncontrolled hypertension (WAVE_IV) (NCT02029885). https://clinicaltrials.gov/ct2/show/results/NCT02029885. Accessed August 3, 2017.
  20. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European clinical consensus conference for renal denervation: considerations on future clinical trial design. Eur Heart J 2015; 36:2219–2227.
  21. Kandzari DE, Kario K, Mahfoud F, et al. The SPYRAL HTN Global Clinical Trial Program: rationale and design for studies of renal denervation in the absence (SPYRAL HTN OFF-MED) and presence (SPYRAL HTN ON-MED) of antihypertensive medications. Am Heart J 2016; 171:82–91.
  22. ClinicalTrialsgov. A Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN). https://clinicaltrials.gov/ct2/show/NCT02649426?term=RADIANCE&rank=3. Accessed August 3, 2017.
References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al, for the SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Symplicity HTN-2 Investigators, Esler MD, Krum H, Sobotka PA, Schlaich MP, Schmieder RE, Bohm M. Renal sympathetic denervation in patients with treatment-resistant hypertension (the Symplicity HTN-2 trial): a randomised controlled trial. Lancet 2010; 376:1903–1909.
  3. Bunte MC, Infante de Oliveira E, Shishehbor MH. Endovascular treatment of resistant and uncontrolled hypertension: therapies on the horizon. JACC Cardiovasc Interv 2013; 6:1–9.
  4. Thomas G, Shishehbor MH, Bravo EL, Nally JV. Renal denervation to treat resistant hypertension: guarded optimism. Cleve Clin J Med 2012; 79:501–510.
  5. Shishehbor MH, Bunte MC. Anatomical exclusion for renal denervation: are we putting the cart before the horse? JACC Cardiovasc Interv 2014; 7:193–194.
  6. Bhatt DL, Bakris GL. The promise of renal denervation. Cleve Clin J Med 2012; 79:498–500.
  7. Bunte MC. Renal sympathetic denervation for refractory hypertension. Lancet 2011; 377:1074; author reply 1075.
  8. Mahfoud F, Luscher TF, Andersson B, et al; European Society of Cardiology. Expert consensus document from the European Society of Cardiology on catheter-based renal denervation. Eur Heart J 2013; 34:2149–2157.
  9. Schlaich MP, Schmieder RE, Bakris G, et al. International expert consensus statement: percutaneous transluminal renal denervation for the treatment of resistant hypertension. J Am Coll Cardiol 2013; 62:2031–2045.
  10. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  11. Rosa J, Widimsky P, Tousek P, et al. Randomized comparison of renal denervation versus intensified pharmacotherapy including spironolactone in true-resistant hypertension: six-month results from the Prague-15 study. Hypertension 2015; 65:407–413.
  12. Williams B, MacDonald TM, Morant S, et al; British Hypertension Society’s PATHWAY Studies Group. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet 2015; 386:2059–2068.
  13. Sakakura K, Ladich E, Cheng Q, et al. Anatomic assessment of sympathetic peri-arterial renal nerves in man. J Am Coll Cardiol 2014; 64:635–643.
  14. Mahfoud F, Edelman ER, Bohm M. Catheter-based renal denervation is no simple matter: lessons to be learned from our anatomy? J Am Coll Cardiol 2014; 64:644–646.
  15. Id D, Kaltenbach B, Bertog SC, et al. Does the presence of accessory renal arteries affect the efficacy of renal denervation? JACC Cardiovasc Interv 2013; 6:1085–1091.
  16. Jin Y, Jacobs L, Baelen M, et al; Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (Inspired) Investigators. Rationale and design of the Investigator-Steered Project on Intravascular Renal Denervation for Management of Drug-Resistant Hypertension (INSPiRED) trial. Blood Press 2014; 23:138–146.
  17. ClinicalTrialsgov. Renal Denervation Using the Vessix Renal Denervation System for the Treatment of Hypertension (REDUCE HTN: REINFORCE). https://clinicaltrials.gov/ct2/show/NCT02392351?term=REDUCE-HTN%3A+REINFORCE&rank=1. Accessed August 3, 2017.
  18. Fischell TA, Ebner A, Gallo S, et al. Transcatheter alcohol-mediated perivascular renal denervation with the peregrine system: first-in-human experience. JACC Cardiovasc Interv 2016; 9:589–598.
  19. ClinicalTrialsgov. Sham controlled study of renal denervation for subjects with uncontrolled hypertension (WAVE_IV) (NCT02029885). https://clinicaltrials.gov/ct2/show/results/NCT02029885. Accessed August 3, 2017.
  20. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European clinical consensus conference for renal denervation: considerations on future clinical trial design. Eur Heart J 2015; 36:2219–2227.
  21. Kandzari DE, Kario K, Mahfoud F, et al. The SPYRAL HTN Global Clinical Trial Program: rationale and design for studies of renal denervation in the absence (SPYRAL HTN OFF-MED) and presence (SPYRAL HTN ON-MED) of antihypertensive medications. Am Heart J 2016; 171:82–91.
  22. ClinicalTrialsgov. A Study of the ReCor Medical Paradise System in Clinical Hypertension (RADIANCE-HTN). https://clinicaltrials.gov/ct2/show/NCT02649426?term=RADIANCE&rank=3. Accessed August 3, 2017.
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Page Number
681-686
Page Number
681-686
Publications
Publications
Topics
Article Type
Display Headline
Renal denervation: What happened, and why?
Display Headline
Renal denervation: What happened, and why?
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Mehdi Shishehbor, Tarek Hammad, George Thomas
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Mehdi Shishehbor, Tarek Hammad, George Thomas
Sections
Inside the Article

KEY POINTS

  • Renal denervation consists of passing a catheter into the renal arteries and ablating their sympathetic nerves using radiofrequency energy. In theory, it should lower blood pressure and be an attractive option for treating resistant hypertension.
  • SYMPLICITY HTN-3 was a blinded trial in which patients with resistant hypertension were randomized to undergo real or sham renal denervation.
  • At 6 months, office systolic blood pressure had failed to fall more in the renal denervation group than in the sham denervation group by a margin of at least 5 mm Hg, the primary efficacy end point of the trial.
  • Methodologic and technical shortcomings may explain the negative results of the SYMPLICITY HTN-3 trial, but most device manufacturers have put the brakes on future research into this novel therapy.
  • Today, renal denervation is not available in the United States but is available for routine care in Europe and Australia.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Renal denervation: Are we on the right path?

Article Type
Changed
Fri, 09/01/2017 - 09:12
Display Headline
Renal denervation: Are we on the right path?

When renal sympathetic denervation, an endovascular procedure designed to treat resistant hypertension, failed to meet its efficacy goal in the SYMPLICITY HTN-3 trial,1 the news was disappointing.

See related article

In this issue of the Cleveland Clinic Journal of Medicine, Shishehbor et al2 provide a critical review of the findings of that trial and summarize its intricacies, as well as the results of other important trials of renal denervation therapy for hypertension. To their excellent observations, we would like to add some of our own.

HYPERTENSION: COMMON, OFTEN RESISTANT

The worldwide prevalence of hypertension is increasing. In the year 2000, about 26% of the adult world population had hypertension; by the year 2025, the number is projected to rise to 29%—1.56 billion people.3

Only about 50% of patients with hypertension are treated for it and, of those, about half have it adequately controlled. In one report, about 30% of US patients with hypertension had adequate blood pressure control.4

Patients who have uncontrolled hypertension are usually older and more obese, have higher baseline blood pressure and excessive salt intake, and are more likely to have chronic kidney disease, diabetes, obstructive sleep apnea, and aldosterone excess.5 Many of these conditions are also associated with increased sympathetic nervous system activity.6

Resistance and pseudoresistance

But lack of control of blood pressure is not the same as resistant hypertension. It is important to differentiate resistant hypertension from pseudoresistant hypertension, ie, hypertension that only seems to be resistant.7 Resistant hypertension affects 12.8% of all drug-treated hypertensive patients in the United States, according to data from the National Health and Nutrition Examination Survey.8

Factors that can cause pseudoresistant hypertension include:

Suboptimal antihypertensive regimens (truly resistant hypertension means blood pressure that remains high despite concurrent treatment with 3 antihypertensive drugs of different classes, 1 of which is a diuretic, in maximal doses)

The white coat effect (higher blood pressure in the office than at home, presumably due to the stress of an office visit)

  • Suboptimal blood pressure measurement techniques (eg, use of a cuff that is too small, causing falsely high readings)
  • Physician inertia (eg, failure to change a regimen that is not working)
  • Lifestyle factors (eg, excessive sodium intake)
  • Medications that interfere with blood pressure control (eg, nonsteroidal anti-inflammatory drugs)
  • Poor adherence to prescribed medications.

Causes of secondary hypertension such as obstructive sleep apnea, primary aldosteronism, and renal artery stenosis should also be ruled out before concluding that a patient has resistant hypertension.

 

 

Treatment prevents complications

Hypertension causes a myriad of medical diseases, including accelerated atherosclerosis, myocardial ischemia and infarction, both systolic and diastolic heart failure, rhythm problems (eg, atrial fibrillation), and stroke.

Most patients with resistant hypertension have no identifiable reversible causes of it, exhibit increased sympathetic nervous system activity, and have increased risk of cardiovascular events. The risk can be reduced by treatment.9,10

Adequate and sustained treatment of hypertension prevents and mitigates its complications. The classic Veterans Administration Cooperative Study in the 1960s demonstrated a 96% reduction in cardiovascular events over 18 months with the use of 3 antihypertensive medications in patients with severe hypertension.11 A reduction of as little as 2 mm Hg in the mean blood pressure has been associated with a 10% reduction in the risk of stroke mortality and a 7% decrease in ischemic heart disease mortality.12 This is an important consideration when evaluating the clinical end points of hypertension trials.

SYMPLICITY HTN-3 TRIAL: WHAT DID WE LEARN?

As controlling blood pressure is paramount in reducing cardiovascular complications, it is only natural to look for innovative strategies to supplement the medical treatments of hypertension.

The multicenter SYMPLICITY HTN-3 trial1 was undertaken to establish the efficacy of renal-artery denervation using radiofrequency energy delivered by a catheter-based system (Symplicity RDN, Medtronic, Dublin, Ireland). This randomized, sham-controlled, blinded study did not show a benefit from this procedure with respect to either of its efficacy end points—at 6 months, a reduction in office systolic blood pressure of at least 5 mm Hg more than with medical therapy alone, or a reduction in mean ambulatory systolic pressure of at least 2 mm Hg more than with medical therapy alone.

Despite the negative results, this medium-size (N = 535) randomized clinical trial still represents the highest-level evidence in the field, and we ought to learn something from it.

Limitations of SYMPLICITY HTN-3

Several factors may have contributed to the negative results of the trial. 

Patient selection. For the most part, patients enrolled in renal denervation trials, including SYMPLICITY HTN-3, were not selected on the basis of heightened sympathetic nervous system activity. Assessment of sympathetic nervous system activity may identify the population most likely to achieve an adequate response.

Of note, the baseline blood pressure readings of patients in this trial were higher in the office than on ambulatory monitoring. Patients with white coat hypertension have increased sympathetic nervous system activity and thus might actually be good candidates for renal denervation therapy.

Adequacy of ablation was not measured. Many argue that an objective measure of the adequacy of the denervation procedure (qualitative or quantitative) should have been implemented and, if it had been, the results might have been different. For example, when ablation is performed in the main renal artery as well as the branches, the efficacy in reducing levels of norepinephrine is improved.13

Blood pressure fell in both groups. In SYMPLICITY HTN-3 and many other renal denervation trials, patients were assessed using both office and ambulatory blood pressure measurements. The primary end point was the office blood pressure measurement, with a 5-mm Hg difference in reduction chosen to define the superiority margin. This margin was chosen because even small reductions in blood pressure are known to decrease adverse events caused by hypertension. Notably, blood pressure fell significantly in both the control and intervention groups, with an intergroup difference of 2.39 mm Hg (not statistically significant) in favor of denervation.

Medication questions. The SYMPLICITY HTN-3 patients were supposed to be on stable medical regimens with maximal tolerated doses before the procedure. However, it was difficult to assess patients’ adherence to and tolerance of medical therapies. Many (about 40%) of the patients had their medications changed during the study.1

Therefore, a critical look at the study enrollment criteria may shed more light on the reasons for the negative findings. Did these patients truly have resistant hypertension? Before they underwent the treatment, was their prestudy pharmacologic regimen adequately intensified?

 

 

ONGOING STUDIES

After the findings of the SYMPLICITY HTN-3 study were released, several other trials—such as the Renal Denervation for Hypertension (DENERHTN)14 and Prague-15 trials15—reported conflicting results. Notably, these were not sham-controlled trials.

Newer studies with robust trial designs are ongoing. A quick search of www.clinicaltrials.gov reveals that at least 89 active clinical trials of renal denervation are registered as of the date of this writing. Excluding those with unknown status, there are 63 trials open or ongoing.

Clinical trials are also ongoing to determine the effects of renal denervation in patients with heart failure, atrial fibrillation, sleep apnea, and chronic kidney disease, all of which are known to involve heightened sympathetic nervous system activity.

NOT READY FOR CLINICAL USE

Although nonpharmacologic treatments of hypertension continue to be studied and are supported by an avalanche of trials in animals and small, mostly nonrandomized trials in humans, one should not forget that the SYMPLICITY HTN-3 trial simply did not meet its primary efficacy end points. We need definitive clinical evidence showing that renal denervation reduces either blood pressure or clinical events before it becomes a mainstream therapy in humans.

Additional trials are being conducted that were designed in accordance with the recommendations of the European Clinical Consensus Conference for Renal Denervation16 in terms of study population, design, and end points. Well-designed studies that conform to those recommendations are critical.

Finally, although our enthusiasm for renal denervation as a treatment of hypertension is tempered, there have been no noteworthy safety concerns related to the procedure, which certainly helps maintain the research momentum in this field.              

References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al; SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Shishehbor MH, Hammad TA, Thomas G. Renal denervation: what happened, and why? Cleve Clin J Med 2017; 84:681–686.
  3. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet 2005; 365:217–223.
  4. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Worldwide prevalence of hypertension: a systematic review. J Hypertens 2004; 22:11–19.
  5. Calhoun DA, Jones D, Textor S, et al; American Heart Association Professional Education Committee. Resistant hypertension: diagnosis, evaluation, and treatment: a scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research. Circulation 2008; 117:e510–e526.
  6. Tsioufis C, Papademetriou V, Thomopoulos C, Stefanadis C. Renal denervation for sleep apnea and resistant hypertension: alternative or complementary to effective continuous positive airway pressure treatment? Hypertension 2011; 58:e191–e192.
  7. Calhoun DA, Jones D, Textor S, et al. Resistant hypertension: diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research.Hypertension 2008; 51:1403–1419.
  8. Persell SD. Prevalence of resistant hypertension in the United States, 2003–2008. Hypertension 2011; 57:1076–1080.
  9. Papademetriou V, Doumas M, Tsioufis K. Renal sympathetic denervation for the treatment of difficult-to-control or resistant hypertension. Int J Hypertens 2011; 2011:196518.
  10. Doumas M, Faselis C, Papademetriou V. Renal sympathetic denervation in hypertension. Curr Opin Nephrol Hypertens 2011; 20:647–653.
  11. Veterans Administration Cooperative Study Group on Antihypertensive Agents. Effect of treatment on morbidity in hypertension: results in patients with diastolic blood pressures averaging 115 through 129 mm Hg. JAMA 1967; 202:1028–1034.
  12. Lewington S, Clarke R, Qizilbash N, Peto R, Collins R; Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002; 360:1903–1913.
  13. Henegar JR, Zhang Y, Hata C, Narciso I, Hall ME, Hall JE. Catheter-based radiofrequency renal denervation: location effects on renal norepinephrine. Am J Hypertens 2015; 28:909–914.
  14. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  15. Rosa J, Widimsky P, Waldauf P, et al. Role of adding spironolactone and renal denervation in true resistant hypertension: one-year outcomes of randomized PRAGUE-15 study. Hypertension 2016; 67:397–403.
  16. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European Clinical Consensus Conference for Renal Denervation: Considerations on Future Clinical Trial Design. Eur Heart J 2015; 6:2219–2227.
Article PDF
Author and Disclosure Information

Ali E. Denktas, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Cardiac Catheterization Laboratories, Michael E. DeBakey VA Medical Center, Houston, TX; site Principal Investigator for the SYMPLICITY HTN-3 Trial

David Paniagua, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Structural Heart Disease Interventions, Michael E. DeBakey VA Medical Center, Houston, TX

Hani Jneid, MD, FACC, FAHA, FSCAI
Associate Professor of Medicine and Director of Interventional Cardiology Research, Baylor College of Medicine; Director of Interventional Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX

Address: Ali E. Denktas, MD, Section of Cardiology, Baylor College of Medicine, 2002 Holcombe Boulevard, Houston, TX 77004; [email protected]

Issue
Cleveland Clinic Journal of Medicine - 84(9)
Publications
Topics
Page Number
687-689
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Ali Denktas, David Paniagua, Hani Jneid
Sections
Author and Disclosure Information

Ali E. Denktas, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Cardiac Catheterization Laboratories, Michael E. DeBakey VA Medical Center, Houston, TX; site Principal Investigator for the SYMPLICITY HTN-3 Trial

David Paniagua, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Structural Heart Disease Interventions, Michael E. DeBakey VA Medical Center, Houston, TX

Hani Jneid, MD, FACC, FAHA, FSCAI
Associate Professor of Medicine and Director of Interventional Cardiology Research, Baylor College of Medicine; Director of Interventional Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX

Address: Ali E. Denktas, MD, Section of Cardiology, Baylor College of Medicine, 2002 Holcombe Boulevard, Houston, TX 77004; [email protected]

Author and Disclosure Information

Ali E. Denktas, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Cardiac Catheterization Laboratories, Michael E. DeBakey VA Medical Center, Houston, TX; site Principal Investigator for the SYMPLICITY HTN-3 Trial

David Paniagua, MD, FACC, FSCAI
Associate Professor of Medicine, Division of Cardiology, Baylor College of Medicine; Director of Structural Heart Disease Interventions, Michael E. DeBakey VA Medical Center, Houston, TX

Hani Jneid, MD, FACC, FAHA, FSCAI
Associate Professor of Medicine and Director of Interventional Cardiology Research, Baylor College of Medicine; Director of Interventional Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX

Address: Ali E. Denktas, MD, Section of Cardiology, Baylor College of Medicine, 2002 Holcombe Boulevard, Houston, TX 77004; [email protected]

Article PDF
Article PDF
Related Articles

When renal sympathetic denervation, an endovascular procedure designed to treat resistant hypertension, failed to meet its efficacy goal in the SYMPLICITY HTN-3 trial,1 the news was disappointing.

See related article

In this issue of the Cleveland Clinic Journal of Medicine, Shishehbor et al2 provide a critical review of the findings of that trial and summarize its intricacies, as well as the results of other important trials of renal denervation therapy for hypertension. To their excellent observations, we would like to add some of our own.

HYPERTENSION: COMMON, OFTEN RESISTANT

The worldwide prevalence of hypertension is increasing. In the year 2000, about 26% of the adult world population had hypertension; by the year 2025, the number is projected to rise to 29%—1.56 billion people.3

Only about 50% of patients with hypertension are treated for it and, of those, about half have it adequately controlled. In one report, about 30% of US patients with hypertension had adequate blood pressure control.4

Patients who have uncontrolled hypertension are usually older and more obese, have higher baseline blood pressure and excessive salt intake, and are more likely to have chronic kidney disease, diabetes, obstructive sleep apnea, and aldosterone excess.5 Many of these conditions are also associated with increased sympathetic nervous system activity.6

Resistance and pseudoresistance

But lack of control of blood pressure is not the same as resistant hypertension. It is important to differentiate resistant hypertension from pseudoresistant hypertension, ie, hypertension that only seems to be resistant.7 Resistant hypertension affects 12.8% of all drug-treated hypertensive patients in the United States, according to data from the National Health and Nutrition Examination Survey.8

Factors that can cause pseudoresistant hypertension include:

Suboptimal antihypertensive regimens (truly resistant hypertension means blood pressure that remains high despite concurrent treatment with 3 antihypertensive drugs of different classes, 1 of which is a diuretic, in maximal doses)

The white coat effect (higher blood pressure in the office than at home, presumably due to the stress of an office visit)

  • Suboptimal blood pressure measurement techniques (eg, use of a cuff that is too small, causing falsely high readings)
  • Physician inertia (eg, failure to change a regimen that is not working)
  • Lifestyle factors (eg, excessive sodium intake)
  • Medications that interfere with blood pressure control (eg, nonsteroidal anti-inflammatory drugs)
  • Poor adherence to prescribed medications.

Causes of secondary hypertension such as obstructive sleep apnea, primary aldosteronism, and renal artery stenosis should also be ruled out before concluding that a patient has resistant hypertension.

 

 

Treatment prevents complications

Hypertension causes a myriad of medical diseases, including accelerated atherosclerosis, myocardial ischemia and infarction, both systolic and diastolic heart failure, rhythm problems (eg, atrial fibrillation), and stroke.

Most patients with resistant hypertension have no identifiable reversible causes of it, exhibit increased sympathetic nervous system activity, and have increased risk of cardiovascular events. The risk can be reduced by treatment.9,10

Adequate and sustained treatment of hypertension prevents and mitigates its complications. The classic Veterans Administration Cooperative Study in the 1960s demonstrated a 96% reduction in cardiovascular events over 18 months with the use of 3 antihypertensive medications in patients with severe hypertension.11 A reduction of as little as 2 mm Hg in the mean blood pressure has been associated with a 10% reduction in the risk of stroke mortality and a 7% decrease in ischemic heart disease mortality.12 This is an important consideration when evaluating the clinical end points of hypertension trials.

SYMPLICITY HTN-3 TRIAL: WHAT DID WE LEARN?

As controlling blood pressure is paramount in reducing cardiovascular complications, it is only natural to look for innovative strategies to supplement the medical treatments of hypertension.

The multicenter SYMPLICITY HTN-3 trial1 was undertaken to establish the efficacy of renal-artery denervation using radiofrequency energy delivered by a catheter-based system (Symplicity RDN, Medtronic, Dublin, Ireland). This randomized, sham-controlled, blinded study did not show a benefit from this procedure with respect to either of its efficacy end points—at 6 months, a reduction in office systolic blood pressure of at least 5 mm Hg more than with medical therapy alone, or a reduction in mean ambulatory systolic pressure of at least 2 mm Hg more than with medical therapy alone.

Despite the negative results, this medium-size (N = 535) randomized clinical trial still represents the highest-level evidence in the field, and we ought to learn something from it.

Limitations of SYMPLICITY HTN-3

Several factors may have contributed to the negative results of the trial. 

Patient selection. For the most part, patients enrolled in renal denervation trials, including SYMPLICITY HTN-3, were not selected on the basis of heightened sympathetic nervous system activity. Assessment of sympathetic nervous system activity may identify the population most likely to achieve an adequate response.

Of note, the baseline blood pressure readings of patients in this trial were higher in the office than on ambulatory monitoring. Patients with white coat hypertension have increased sympathetic nervous system activity and thus might actually be good candidates for renal denervation therapy.

Adequacy of ablation was not measured. Many argue that an objective measure of the adequacy of the denervation procedure (qualitative or quantitative) should have been implemented and, if it had been, the results might have been different. For example, when ablation is performed in the main renal artery as well as the branches, the efficacy in reducing levels of norepinephrine is improved.13

Blood pressure fell in both groups. In SYMPLICITY HTN-3 and many other renal denervation trials, patients were assessed using both office and ambulatory blood pressure measurements. The primary end point was the office blood pressure measurement, with a 5-mm Hg difference in reduction chosen to define the superiority margin. This margin was chosen because even small reductions in blood pressure are known to decrease adverse events caused by hypertension. Notably, blood pressure fell significantly in both the control and intervention groups, with an intergroup difference of 2.39 mm Hg (not statistically significant) in favor of denervation.

Medication questions. The SYMPLICITY HTN-3 patients were supposed to be on stable medical regimens with maximal tolerated doses before the procedure. However, it was difficult to assess patients’ adherence to and tolerance of medical therapies. Many (about 40%) of the patients had their medications changed during the study.1

Therefore, a critical look at the study enrollment criteria may shed more light on the reasons for the negative findings. Did these patients truly have resistant hypertension? Before they underwent the treatment, was their prestudy pharmacologic regimen adequately intensified?

 

 

ONGOING STUDIES

After the findings of the SYMPLICITY HTN-3 study were released, several other trials—such as the Renal Denervation for Hypertension (DENERHTN)14 and Prague-15 trials15—reported conflicting results. Notably, these were not sham-controlled trials.

Newer studies with robust trial designs are ongoing. A quick search of www.clinicaltrials.gov reveals that at least 89 active clinical trials of renal denervation are registered as of the date of this writing. Excluding those with unknown status, there are 63 trials open or ongoing.

Clinical trials are also ongoing to determine the effects of renal denervation in patients with heart failure, atrial fibrillation, sleep apnea, and chronic kidney disease, all of which are known to involve heightened sympathetic nervous system activity.

NOT READY FOR CLINICAL USE

Although nonpharmacologic treatments of hypertension continue to be studied and are supported by an avalanche of trials in animals and small, mostly nonrandomized trials in humans, one should not forget that the SYMPLICITY HTN-3 trial simply did not meet its primary efficacy end points. We need definitive clinical evidence showing that renal denervation reduces either blood pressure or clinical events before it becomes a mainstream therapy in humans.

Additional trials are being conducted that were designed in accordance with the recommendations of the European Clinical Consensus Conference for Renal Denervation16 in terms of study population, design, and end points. Well-designed studies that conform to those recommendations are critical.

Finally, although our enthusiasm for renal denervation as a treatment of hypertension is tempered, there have been no noteworthy safety concerns related to the procedure, which certainly helps maintain the research momentum in this field.              

When renal sympathetic denervation, an endovascular procedure designed to treat resistant hypertension, failed to meet its efficacy goal in the SYMPLICITY HTN-3 trial,1 the news was disappointing.

See related article

In this issue of the Cleveland Clinic Journal of Medicine, Shishehbor et al2 provide a critical review of the findings of that trial and summarize its intricacies, as well as the results of other important trials of renal denervation therapy for hypertension. To their excellent observations, we would like to add some of our own.

HYPERTENSION: COMMON, OFTEN RESISTANT

The worldwide prevalence of hypertension is increasing. In the year 2000, about 26% of the adult world population had hypertension; by the year 2025, the number is projected to rise to 29%—1.56 billion people.3

Only about 50% of patients with hypertension are treated for it and, of those, about half have it adequately controlled. In one report, about 30% of US patients with hypertension had adequate blood pressure control.4

Patients who have uncontrolled hypertension are usually older and more obese, have higher baseline blood pressure and excessive salt intake, and are more likely to have chronic kidney disease, diabetes, obstructive sleep apnea, and aldosterone excess.5 Many of these conditions are also associated with increased sympathetic nervous system activity.6

Resistance and pseudoresistance

But lack of control of blood pressure is not the same as resistant hypertension. It is important to differentiate resistant hypertension from pseudoresistant hypertension, ie, hypertension that only seems to be resistant.7 Resistant hypertension affects 12.8% of all drug-treated hypertensive patients in the United States, according to data from the National Health and Nutrition Examination Survey.8

Factors that can cause pseudoresistant hypertension include:

Suboptimal antihypertensive regimens (truly resistant hypertension means blood pressure that remains high despite concurrent treatment with 3 antihypertensive drugs of different classes, 1 of which is a diuretic, in maximal doses)

The white coat effect (higher blood pressure in the office than at home, presumably due to the stress of an office visit)

  • Suboptimal blood pressure measurement techniques (eg, use of a cuff that is too small, causing falsely high readings)
  • Physician inertia (eg, failure to change a regimen that is not working)
  • Lifestyle factors (eg, excessive sodium intake)
  • Medications that interfere with blood pressure control (eg, nonsteroidal anti-inflammatory drugs)
  • Poor adherence to prescribed medications.

Causes of secondary hypertension such as obstructive sleep apnea, primary aldosteronism, and renal artery stenosis should also be ruled out before concluding that a patient has resistant hypertension.

 

 

Treatment prevents complications

Hypertension causes a myriad of medical diseases, including accelerated atherosclerosis, myocardial ischemia and infarction, both systolic and diastolic heart failure, rhythm problems (eg, atrial fibrillation), and stroke.

Most patients with resistant hypertension have no identifiable reversible causes of it, exhibit increased sympathetic nervous system activity, and have increased risk of cardiovascular events. The risk can be reduced by treatment.9,10

Adequate and sustained treatment of hypertension prevents and mitigates its complications. The classic Veterans Administration Cooperative Study in the 1960s demonstrated a 96% reduction in cardiovascular events over 18 months with the use of 3 antihypertensive medications in patients with severe hypertension.11 A reduction of as little as 2 mm Hg in the mean blood pressure has been associated with a 10% reduction in the risk of stroke mortality and a 7% decrease in ischemic heart disease mortality.12 This is an important consideration when evaluating the clinical end points of hypertension trials.

SYMPLICITY HTN-3 TRIAL: WHAT DID WE LEARN?

As controlling blood pressure is paramount in reducing cardiovascular complications, it is only natural to look for innovative strategies to supplement the medical treatments of hypertension.

The multicenter SYMPLICITY HTN-3 trial1 was undertaken to establish the efficacy of renal-artery denervation using radiofrequency energy delivered by a catheter-based system (Symplicity RDN, Medtronic, Dublin, Ireland). This randomized, sham-controlled, blinded study did not show a benefit from this procedure with respect to either of its efficacy end points—at 6 months, a reduction in office systolic blood pressure of at least 5 mm Hg more than with medical therapy alone, or a reduction in mean ambulatory systolic pressure of at least 2 mm Hg more than with medical therapy alone.

Despite the negative results, this medium-size (N = 535) randomized clinical trial still represents the highest-level evidence in the field, and we ought to learn something from it.

Limitations of SYMPLICITY HTN-3

Several factors may have contributed to the negative results of the trial. 

Patient selection. For the most part, patients enrolled in renal denervation trials, including SYMPLICITY HTN-3, were not selected on the basis of heightened sympathetic nervous system activity. Assessment of sympathetic nervous system activity may identify the population most likely to achieve an adequate response.

Of note, the baseline blood pressure readings of patients in this trial were higher in the office than on ambulatory monitoring. Patients with white coat hypertension have increased sympathetic nervous system activity and thus might actually be good candidates for renal denervation therapy.

Adequacy of ablation was not measured. Many argue that an objective measure of the adequacy of the denervation procedure (qualitative or quantitative) should have been implemented and, if it had been, the results might have been different. For example, when ablation is performed in the main renal artery as well as the branches, the efficacy in reducing levels of norepinephrine is improved.13

Blood pressure fell in both groups. In SYMPLICITY HTN-3 and many other renal denervation trials, patients were assessed using both office and ambulatory blood pressure measurements. The primary end point was the office blood pressure measurement, with a 5-mm Hg difference in reduction chosen to define the superiority margin. This margin was chosen because even small reductions in blood pressure are known to decrease adverse events caused by hypertension. Notably, blood pressure fell significantly in both the control and intervention groups, with an intergroup difference of 2.39 mm Hg (not statistically significant) in favor of denervation.

Medication questions. The SYMPLICITY HTN-3 patients were supposed to be on stable medical regimens with maximal tolerated doses before the procedure. However, it was difficult to assess patients’ adherence to and tolerance of medical therapies. Many (about 40%) of the patients had their medications changed during the study.1

Therefore, a critical look at the study enrollment criteria may shed more light on the reasons for the negative findings. Did these patients truly have resistant hypertension? Before they underwent the treatment, was their prestudy pharmacologic regimen adequately intensified?

 

 

ONGOING STUDIES

After the findings of the SYMPLICITY HTN-3 study were released, several other trials—such as the Renal Denervation for Hypertension (DENERHTN)14 and Prague-15 trials15—reported conflicting results. Notably, these were not sham-controlled trials.

Newer studies with robust trial designs are ongoing. A quick search of www.clinicaltrials.gov reveals that at least 89 active clinical trials of renal denervation are registered as of the date of this writing. Excluding those with unknown status, there are 63 trials open or ongoing.

Clinical trials are also ongoing to determine the effects of renal denervation in patients with heart failure, atrial fibrillation, sleep apnea, and chronic kidney disease, all of which are known to involve heightened sympathetic nervous system activity.

NOT READY FOR CLINICAL USE

Although nonpharmacologic treatments of hypertension continue to be studied and are supported by an avalanche of trials in animals and small, mostly nonrandomized trials in humans, one should not forget that the SYMPLICITY HTN-3 trial simply did not meet its primary efficacy end points. We need definitive clinical evidence showing that renal denervation reduces either blood pressure or clinical events before it becomes a mainstream therapy in humans.

Additional trials are being conducted that were designed in accordance with the recommendations of the European Clinical Consensus Conference for Renal Denervation16 in terms of study population, design, and end points. Well-designed studies that conform to those recommendations are critical.

Finally, although our enthusiasm for renal denervation as a treatment of hypertension is tempered, there have been no noteworthy safety concerns related to the procedure, which certainly helps maintain the research momentum in this field.              

References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al; SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Shishehbor MH, Hammad TA, Thomas G. Renal denervation: what happened, and why? Cleve Clin J Med 2017; 84:681–686.
  3. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet 2005; 365:217–223.
  4. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Worldwide prevalence of hypertension: a systematic review. J Hypertens 2004; 22:11–19.
  5. Calhoun DA, Jones D, Textor S, et al; American Heart Association Professional Education Committee. Resistant hypertension: diagnosis, evaluation, and treatment: a scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research. Circulation 2008; 117:e510–e526.
  6. Tsioufis C, Papademetriou V, Thomopoulos C, Stefanadis C. Renal denervation for sleep apnea and resistant hypertension: alternative or complementary to effective continuous positive airway pressure treatment? Hypertension 2011; 58:e191–e192.
  7. Calhoun DA, Jones D, Textor S, et al. Resistant hypertension: diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research.Hypertension 2008; 51:1403–1419.
  8. Persell SD. Prevalence of resistant hypertension in the United States, 2003–2008. Hypertension 2011; 57:1076–1080.
  9. Papademetriou V, Doumas M, Tsioufis K. Renal sympathetic denervation for the treatment of difficult-to-control or resistant hypertension. Int J Hypertens 2011; 2011:196518.
  10. Doumas M, Faselis C, Papademetriou V. Renal sympathetic denervation in hypertension. Curr Opin Nephrol Hypertens 2011; 20:647–653.
  11. Veterans Administration Cooperative Study Group on Antihypertensive Agents. Effect of treatment on morbidity in hypertension: results in patients with diastolic blood pressures averaging 115 through 129 mm Hg. JAMA 1967; 202:1028–1034.
  12. Lewington S, Clarke R, Qizilbash N, Peto R, Collins R; Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002; 360:1903–1913.
  13. Henegar JR, Zhang Y, Hata C, Narciso I, Hall ME, Hall JE. Catheter-based radiofrequency renal denervation: location effects on renal norepinephrine. Am J Hypertens 2015; 28:909–914.
  14. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  15. Rosa J, Widimsky P, Waldauf P, et al. Role of adding spironolactone and renal denervation in true resistant hypertension: one-year outcomes of randomized PRAGUE-15 study. Hypertension 2016; 67:397–403.
  16. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European Clinical Consensus Conference for Renal Denervation: Considerations on Future Clinical Trial Design. Eur Heart J 2015; 6:2219–2227.
References
  1. Bhatt DL, Kandzari DE, O’Neill WW, et al; SYMPLICITY HTN-3 Investigators. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370:1393–1401.
  2. Shishehbor MH, Hammad TA, Thomas G. Renal denervation: what happened, and why? Cleve Clin J Med 2017; 84:681–686.
  3. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet 2005; 365:217–223.
  4. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Worldwide prevalence of hypertension: a systematic review. J Hypertens 2004; 22:11–19.
  5. Calhoun DA, Jones D, Textor S, et al; American Heart Association Professional Education Committee. Resistant hypertension: diagnosis, evaluation, and treatment: a scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research. Circulation 2008; 117:e510–e526.
  6. Tsioufis C, Papademetriou V, Thomopoulos C, Stefanadis C. Renal denervation for sleep apnea and resistant hypertension: alternative or complementary to effective continuous positive airway pressure treatment? Hypertension 2011; 58:e191–e192.
  7. Calhoun DA, Jones D, Textor S, et al. Resistant hypertension: diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research.Hypertension 2008; 51:1403–1419.
  8. Persell SD. Prevalence of resistant hypertension in the United States, 2003–2008. Hypertension 2011; 57:1076–1080.
  9. Papademetriou V, Doumas M, Tsioufis K. Renal sympathetic denervation for the treatment of difficult-to-control or resistant hypertension. Int J Hypertens 2011; 2011:196518.
  10. Doumas M, Faselis C, Papademetriou V. Renal sympathetic denervation in hypertension. Curr Opin Nephrol Hypertens 2011; 20:647–653.
  11. Veterans Administration Cooperative Study Group on Antihypertensive Agents. Effect of treatment on morbidity in hypertension: results in patients with diastolic blood pressures averaging 115 through 129 mm Hg. JAMA 1967; 202:1028–1034.
  12. Lewington S, Clarke R, Qizilbash N, Peto R, Collins R; Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002; 360:1903–1913.
  13. Henegar JR, Zhang Y, Hata C, Narciso I, Hall ME, Hall JE. Catheter-based radiofrequency renal denervation: location effects on renal norepinephrine. Am J Hypertens 2015; 28:909–914.
  14. Azizi M, Sapoval M, Gosse P, et al; Renal Denervation for Hypertension (DENERHTN) investigators. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet 2015; 385:1957–1965.
  15. Rosa J, Widimsky P, Waldauf P, et al. Role of adding spironolactone and renal denervation in true resistant hypertension: one-year outcomes of randomized PRAGUE-15 study. Hypertension 2016; 67:397–403.
  16. Mahfoud F, Bohm M, Azizi M, et al. Proceedings from the European Clinical Consensus Conference for Renal Denervation: Considerations on Future Clinical Trial Design. Eur Heart J 2015; 6:2219–2227.
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Page Number
687-689
Page Number
687-689
Publications
Publications
Topics
Article Type
Display Headline
Renal denervation: Are we on the right path?
Display Headline
Renal denervation: Are we on the right path?
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Ali Denktas, David Paniagua, Hani Jneid
Legacy Keywords
renal denervation, renal arteries, high blood pressure, hypertension, Symplicity, Symplicity HTN-3, sympathetic nervous system, ablation, catheter ablation, Ali Denktas, David Paniagua, Hani Jneid
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Create an effective social media campaign to market your practice: Here’s how

Article Type
Changed
Thu, 03/28/2019 - 14:48
Display Headline
Create an effective social media campaign to market your practice: Here’s how

Developing an effective social media marketing campaign can expand your practice to bring you more of the type of patient you want to treat. Although ObGyns are often not trained in marketing, we can bring our practices to the attention of women who need our services with a few simple processes.

The American Marketing Association defines marketing as “the activity, set of institutions, and processes for creating, communicating, delivering, and exchanging offerings that have value for customers, clients, partners, and society at large.”1 Social media is described as various forms of online and mobile electronic communication with user-generated content.2 Social media marketing is the application of traditional marketing strategies to a social media platform. Delivering an effective social media marketing campaign requires focused targeting of a particular community to match the needs of those patients with the value of services and products your practice provides.

By communicating and connecting with the spoken and unspoken needs and desires of potential patients, you will generate greater enthusiasm for your medical services. Social media marketing benefits include: accessibility, low cost, the ability to build brand recognition and social capital, and the availability of analytics that provide large amounts of data to measure the effectiveness of the campaign.3

Though social media is pervasive, the medical community has not rapidly embraced it for marketing.4,5 Creating a social media strategy, rather than randomly or impulsively posting on social media, allows for more effective marketing. The discussion here focuses on Facebook, which has 2 billion monthly users,6 but these strategies and tactics can be applied to any social media platform, including YouTube, Instagram, and Twitter.7

Use Facebook to create a business page

Your medical practice needs to have a Facebook account and a Facebook page, separate from your personal account. A business-related Facebook page is similar to a personal Facebook profile except that pages are designed for organizations, brands, businesses, and public figures to share photos, stories, and events with the public.

If you do not have a Facebook account, you can create a new account and profile at http://www.facebook.com. After creating a profile, click on the “create a Facebook page” link. Follow the instructions and select the page category you would like to create; most physicians would select the “Company,” “Organization,” or “Institution” category. Next, follow the instructions to complete the registration.8 Once your Facebook page is created, build an audience asking others to “like” your page. Start posting content and use hashtags in your posts to make them discoverable to others (ie, #fibroids #noscar #singlesitesurgery).9

 

Related article:
Using the Internet in your practice. Part 2: Generating new patients using social media

 

One benefit to having a practice-based Facebook page is the automated visible analytics that come with the page, which are not available for personal profiles. When you write a post or upload a photo or video, Facebook provides the demographics of those engaged with your posts plus analytics on that post, including the number of people who viewed the post, clicked on a photo, and viewed the video for more than 3 seconds.

 

Read how to get patients interested in your practice

 

 

Develop a social media marketing strategy

There are several key factors to consider when planning a strategy. First, know the mission of your organization and the specific service, value, or benefit you would like to provide to the targeted community.8

Segment, target, and position (STP)

It is tempting to try to reach out to all women because your ObGyn practice entails pre‑natal care, family planning, and gynecologic surgery, but by narrowing your target audience, your campaign will be better focused. A very specific target audience can reduce the costs for “boosting” (paid promotion of your posts on Facebook to a chosen audience based on demographics, interests, and behaviors) your posts and improve your return on investment (ROI).

Create different marketing campaigns, but focus on one at a time. Decide on the ideal patient you want to serve in your practice. The more detailed and focused you are about the demographics and type of medical needs to be served, the better you can target this patient.10

Segment. Divide the communities you are considering into different segments. For instance, even though you may do obstetrics and gynecologic surgery, consider breaking up the campaign to focus on 1 specific group, such as those interested in fibroid management.

Target. Identify the kinds of communities where you might find this patient. For example, if you want to focus on laparoscopic hysterectomies or myomectomies, start looking on Facebook for groups, pages, or website discussion boards or blogs that discuss abnormal uterine bleeding or fibroids and follow those pages.

Also, think about what other characteristics are associated with these ideal patients. For example, you might narrow it down to perimenopausal women with fibroids. A potential targeted group could be 40- to 50-year-old women who participate in yoga or running who have concerns about fibroids interfering in their active lifestyle. Perhaps this type of patient would want a minimally invasive surgical approach. A holistic health activist might be interested in nonsurgical management of fibroids.

Position. Once you have identified the specific community to target, position your practice within the community with the value proposition you are offering. For example, as an ObGyn who is focused on surgery, your position might be that your practice will provide the best experience for those medical services, with specific counseling to patients about resuming their active lifestyle.

 

Related article:
Four pillars of a successful practice: 2. Attract new patients

 

Get your potential patient to “raise her hand.” In the campaign, you are not trying to convince everyone up front to schedule an appointment from one post. First, try to get people who may be interested in your service(s) to “raise their hands.” Once your target market has expressed interest, either by their likes of your post, likes of your page, or other engagement, reach out to them with links for more information, such as free fibroid surgery education materials located on your website. On your website, create an opt-in page asking them to register their email address; once you have a compiled email list, send out monthly newsletters on your practice.11

 

Read how to guide patients to your office

 

 

Understand that marketing is a process

Think of marketing as an overall process in which you are guiding potential patients to come to your office. Your campaign has several steps; recognize that just one post will not make a huge difference. Use Facebook analytics to measure cost per engagement to calculate your return on investment and the campaign’s effectiveness, and revise as necessary.

Rather than just considering social media as a soap box to advertise your practice, break up the marketing process into 3 units: the before unit, the during unit, and the after unit.11 The word “unit” denotes the service, benefit, or product you are providing.

The before unit refers to the initial marketing that identifies potential patients—initially getting them to raise their hands and ultimately building an audience. (Once a potential patient provides her email address, you can send her a monthly newsletter or updates about your practice to continue the engagement.) Statistics show that an ObGyn needs to have 7 contacts, on average, with a patient over 18 months to “penetrate” her consciousness in a given market.12 Of course if there is an urgent or emergent need to see a physician, that timeline would be much shorter.

The during unit occurs when the patient comes to your practice and service is being provided. Since you know what she is coming for, you can create informational packets focused on her particular needs, perhaps about different management options for fibroids.

The after unit includes following up with the patient in some automated way. For those being treated for fibroids, it may be a reminder email that discusses the value of follow-up ultrasonography or the various kinds of surgical interventions for fibroids.

In order to continue your campaign, it is helpful to have a designated social media manager who will continue the social media posts and engagement.

When creating the posts, consider developing prescheduled assets (posts that are already produced with photos or links to articles), which can be done through Facebook or Hootsuite (http://www.hootsuite.com).

Manage the risks of social media interaction

There are risks associated with social media. Some things to consider are:

  • Policy. Develop a policy for your practice; if you work for an institution, align your policy with the institution’s.
  • Postings. Supervise content being posted. Never allow social media to be placed by someone without supervision. Either you should do this or assign a manager to be accountable to check on social media interactions so that any inappropriate comments can be addressed immediately.
  • Privacy. Never mention patients’ private health information or use the platform to publicly engage with a patient or future patient about their care. Do not post any references to patients or their photos without written consent.
  • Images. Use photographs and other images properly: obtain releases and obey copyright laws.

 

Related article:
Your patients are talking: Isn’t it time you take responsibility for your online reputation?

 

Bottom line

Social media is a powerful platform. Combined with good marketing strategies, social media campaigns can have a significant impact on expanding your practice to offer the kind of medical services you want to provide.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Definition of Marketing. American Marketing Association website. https://www.ama.org/AboutAMA/Pages/Definition-of-Marketing.aspx. Published July 2013. Accessed August 8, 2017.
  2. Kaplan AH, Haenlein M. Users of the world, unite! The challenges and opportunities of social media. Business Horiz. 2010;53(1):59–68.
  3. Lin KY, Lu HP. Intention to continue using Facebook fan pages from the perspective of social capital theory. Cyberpsychol Behav Soc Netw. 2011;14(10):565–570.
  4. Hawn C. Take two aspirin and tweet me in the morning: how Twitter, Facebook, and other social media are reshaping health care. Health Aff (Millwood). 2009;28(2):361–368.
  5. Wheeler CK, Said H, Prucz R, Rodrich RJ, Mathes DW. Social media in plastic surgery practices: emerging trends in North America. Aesthet Surg J. 2011;31(4):435–441.
  6. Nowak M, Spiller G. Two billion people coming together on Facebook. Facebook Newsroom. https://newsroom.fb.com/news/2017/06/two-billion-people-coming-together-on-facebook/. Published June 27, 2017. Accessed August 8, 2017.
  7. Adamson A. No contest: Twitter and Facebook can both play a role in branding. Forbes. http://www.forbes.com/2009/05/06/twitter-facebook-branding-leadership-cmo-network-adamson.html. Published May 6, 2009. Accessed August 8, 2017.
  8. Kim DS. Harness social media, enhance your practice. Contemp Obstet Gynecol. 2012;57(7):40–42,44–46.
  9. Wolf J. Social Media: Master, Manipulate, And Dominate Social Media Marketing Facebook, Twitter, YouTube, Instagram And LinkedIn. Createspace Independent Publishing Platform; 2015:129–143.
  10. Kotler PT, Keller KL. Marketing Management. 12th ed. Upper Saddle River, NJ: Prentice Hall; 2006:239–268.
  11. Jackson DP. Sunday marketing matinee: I love marketing live–Before, during, and after unit thinking. http://ilovemarketing.com/sunday-marketing-matineei-love-marketing-live-before-during-and-after-unit-thinking/. Accessed July 24, 2017.
  12. Payne D. How many contacts does it take before someone buys your product? Business Insider website. http://www.businessinsider.com/how-many-contacts-does-it-take-before-someone-buys-your-product-2011-7. Published July 12, 2011. Accessed August 8, 2017.
Author and Disclosure Information

Dr. Kim is Associate Clinical Professor, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, and Associate Clinical Professor, David Geffen School of Medicine, University of California–Los Angeles.

The author reports no financial relationships relevant to this article.

Issue
OBG Management - 29(9)
Publications
Topics
Page Number
42-45
Sections
Author and Disclosure Information

Dr. Kim is Associate Clinical Professor, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, and Associate Clinical Professor, David Geffen School of Medicine, University of California–Los Angeles.

The author reports no financial relationships relevant to this article.

Author and Disclosure Information

Dr. Kim is Associate Clinical Professor, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, and Associate Clinical Professor, David Geffen School of Medicine, University of California–Los Angeles.

The author reports no financial relationships relevant to this article.

Developing an effective social media marketing campaign can expand your practice to bring you more of the type of patient you want to treat. Although ObGyns are often not trained in marketing, we can bring our practices to the attention of women who need our services with a few simple processes.

The American Marketing Association defines marketing as “the activity, set of institutions, and processes for creating, communicating, delivering, and exchanging offerings that have value for customers, clients, partners, and society at large.”1 Social media is described as various forms of online and mobile electronic communication with user-generated content.2 Social media marketing is the application of traditional marketing strategies to a social media platform. Delivering an effective social media marketing campaign requires focused targeting of a particular community to match the needs of those patients with the value of services and products your practice provides.

By communicating and connecting with the spoken and unspoken needs and desires of potential patients, you will generate greater enthusiasm for your medical services. Social media marketing benefits include: accessibility, low cost, the ability to build brand recognition and social capital, and the availability of analytics that provide large amounts of data to measure the effectiveness of the campaign.3

Though social media is pervasive, the medical community has not rapidly embraced it for marketing.4,5 Creating a social media strategy, rather than randomly or impulsively posting on social media, allows for more effective marketing. The discussion here focuses on Facebook, which has 2 billion monthly users,6 but these strategies and tactics can be applied to any social media platform, including YouTube, Instagram, and Twitter.7

Use Facebook to create a business page

Your medical practice needs to have a Facebook account and a Facebook page, separate from your personal account. A business-related Facebook page is similar to a personal Facebook profile except that pages are designed for organizations, brands, businesses, and public figures to share photos, stories, and events with the public.

If you do not have a Facebook account, you can create a new account and profile at http://www.facebook.com. After creating a profile, click on the “create a Facebook page” link. Follow the instructions and select the page category you would like to create; most physicians would select the “Company,” “Organization,” or “Institution” category. Next, follow the instructions to complete the registration.8 Once your Facebook page is created, build an audience asking others to “like” your page. Start posting content and use hashtags in your posts to make them discoverable to others (ie, #fibroids #noscar #singlesitesurgery).9

 

Related article:
Using the Internet in your practice. Part 2: Generating new patients using social media

 

One benefit to having a practice-based Facebook page is the automated visible analytics that come with the page, which are not available for personal profiles. When you write a post or upload a photo or video, Facebook provides the demographics of those engaged with your posts plus analytics on that post, including the number of people who viewed the post, clicked on a photo, and viewed the video for more than 3 seconds.

 

Read how to get patients interested in your practice

 

 

Develop a social media marketing strategy

There are several key factors to consider when planning a strategy. First, know the mission of your organization and the specific service, value, or benefit you would like to provide to the targeted community.8

Segment, target, and position (STP)

It is tempting to try to reach out to all women because your ObGyn practice entails pre‑natal care, family planning, and gynecologic surgery, but by narrowing your target audience, your campaign will be better focused. A very specific target audience can reduce the costs for “boosting” (paid promotion of your posts on Facebook to a chosen audience based on demographics, interests, and behaviors) your posts and improve your return on investment (ROI).

Create different marketing campaigns, but focus on one at a time. Decide on the ideal patient you want to serve in your practice. The more detailed and focused you are about the demographics and type of medical needs to be served, the better you can target this patient.10

Segment. Divide the communities you are considering into different segments. For instance, even though you may do obstetrics and gynecologic surgery, consider breaking up the campaign to focus on 1 specific group, such as those interested in fibroid management.

Target. Identify the kinds of communities where you might find this patient. For example, if you want to focus on laparoscopic hysterectomies or myomectomies, start looking on Facebook for groups, pages, or website discussion boards or blogs that discuss abnormal uterine bleeding or fibroids and follow those pages.

Also, think about what other characteristics are associated with these ideal patients. For example, you might narrow it down to perimenopausal women with fibroids. A potential targeted group could be 40- to 50-year-old women who participate in yoga or running who have concerns about fibroids interfering in their active lifestyle. Perhaps this type of patient would want a minimally invasive surgical approach. A holistic health activist might be interested in nonsurgical management of fibroids.

Position. Once you have identified the specific community to target, position your practice within the community with the value proposition you are offering. For example, as an ObGyn who is focused on surgery, your position might be that your practice will provide the best experience for those medical services, with specific counseling to patients about resuming their active lifestyle.

 

Related article:
Four pillars of a successful practice: 2. Attract new patients

 

Get your potential patient to “raise her hand.” In the campaign, you are not trying to convince everyone up front to schedule an appointment from one post. First, try to get people who may be interested in your service(s) to “raise their hands.” Once your target market has expressed interest, either by their likes of your post, likes of your page, or other engagement, reach out to them with links for more information, such as free fibroid surgery education materials located on your website. On your website, create an opt-in page asking them to register their email address; once you have a compiled email list, send out monthly newsletters on your practice.11

 

Read how to guide patients to your office

 

 

Understand that marketing is a process

Think of marketing as an overall process in which you are guiding potential patients to come to your office. Your campaign has several steps; recognize that just one post will not make a huge difference. Use Facebook analytics to measure cost per engagement to calculate your return on investment and the campaign’s effectiveness, and revise as necessary.

Rather than just considering social media as a soap box to advertise your practice, break up the marketing process into 3 units: the before unit, the during unit, and the after unit.11 The word “unit” denotes the service, benefit, or product you are providing.

The before unit refers to the initial marketing that identifies potential patients—initially getting them to raise their hands and ultimately building an audience. (Once a potential patient provides her email address, you can send her a monthly newsletter or updates about your practice to continue the engagement.) Statistics show that an ObGyn needs to have 7 contacts, on average, with a patient over 18 months to “penetrate” her consciousness in a given market.12 Of course if there is an urgent or emergent need to see a physician, that timeline would be much shorter.

The during unit occurs when the patient comes to your practice and service is being provided. Since you know what she is coming for, you can create informational packets focused on her particular needs, perhaps about different management options for fibroids.

The after unit includes following up with the patient in some automated way. For those being treated for fibroids, it may be a reminder email that discusses the value of follow-up ultrasonography or the various kinds of surgical interventions for fibroids.

In order to continue your campaign, it is helpful to have a designated social media manager who will continue the social media posts and engagement.

When creating the posts, consider developing prescheduled assets (posts that are already produced with photos or links to articles), which can be done through Facebook or Hootsuite (http://www.hootsuite.com).

Manage the risks of social media interaction

There are risks associated with social media. Some things to consider are:

  • Policy. Develop a policy for your practice; if you work for an institution, align your policy with the institution’s.
  • Postings. Supervise content being posted. Never allow social media to be placed by someone without supervision. Either you should do this or assign a manager to be accountable to check on social media interactions so that any inappropriate comments can be addressed immediately.
  • Privacy. Never mention patients’ private health information or use the platform to publicly engage with a patient or future patient about their care. Do not post any references to patients or their photos without written consent.
  • Images. Use photographs and other images properly: obtain releases and obey copyright laws.

 

Related article:
Your patients are talking: Isn’t it time you take responsibility for your online reputation?

 

Bottom line

Social media is a powerful platform. Combined with good marketing strategies, social media campaigns can have a significant impact on expanding your practice to offer the kind of medical services you want to provide.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

Developing an effective social media marketing campaign can expand your practice to bring you more of the type of patient you want to treat. Although ObGyns are often not trained in marketing, we can bring our practices to the attention of women who need our services with a few simple processes.

The American Marketing Association defines marketing as “the activity, set of institutions, and processes for creating, communicating, delivering, and exchanging offerings that have value for customers, clients, partners, and society at large.”1 Social media is described as various forms of online and mobile electronic communication with user-generated content.2 Social media marketing is the application of traditional marketing strategies to a social media platform. Delivering an effective social media marketing campaign requires focused targeting of a particular community to match the needs of those patients with the value of services and products your practice provides.

By communicating and connecting with the spoken and unspoken needs and desires of potential patients, you will generate greater enthusiasm for your medical services. Social media marketing benefits include: accessibility, low cost, the ability to build brand recognition and social capital, and the availability of analytics that provide large amounts of data to measure the effectiveness of the campaign.3

Though social media is pervasive, the medical community has not rapidly embraced it for marketing.4,5 Creating a social media strategy, rather than randomly or impulsively posting on social media, allows for more effective marketing. The discussion here focuses on Facebook, which has 2 billion monthly users,6 but these strategies and tactics can be applied to any social media platform, including YouTube, Instagram, and Twitter.7

Use Facebook to create a business page

Your medical practice needs to have a Facebook account and a Facebook page, separate from your personal account. A business-related Facebook page is similar to a personal Facebook profile except that pages are designed for organizations, brands, businesses, and public figures to share photos, stories, and events with the public.

If you do not have a Facebook account, you can create a new account and profile at http://www.facebook.com. After creating a profile, click on the “create a Facebook page” link. Follow the instructions and select the page category you would like to create; most physicians would select the “Company,” “Organization,” or “Institution” category. Next, follow the instructions to complete the registration.8 Once your Facebook page is created, build an audience asking others to “like” your page. Start posting content and use hashtags in your posts to make them discoverable to others (ie, #fibroids #noscar #singlesitesurgery).9

 

Related article:
Using the Internet in your practice. Part 2: Generating new patients using social media

 

One benefit to having a practice-based Facebook page is the automated visible analytics that come with the page, which are not available for personal profiles. When you write a post or upload a photo or video, Facebook provides the demographics of those engaged with your posts plus analytics on that post, including the number of people who viewed the post, clicked on a photo, and viewed the video for more than 3 seconds.

 

Read how to get patients interested in your practice

 

 

Develop a social media marketing strategy

There are several key factors to consider when planning a strategy. First, know the mission of your organization and the specific service, value, or benefit you would like to provide to the targeted community.8

Segment, target, and position (STP)

It is tempting to try to reach out to all women because your ObGyn practice entails pre‑natal care, family planning, and gynecologic surgery, but by narrowing your target audience, your campaign will be better focused. A very specific target audience can reduce the costs for “boosting” (paid promotion of your posts on Facebook to a chosen audience based on demographics, interests, and behaviors) your posts and improve your return on investment (ROI).

Create different marketing campaigns, but focus on one at a time. Decide on the ideal patient you want to serve in your practice. The more detailed and focused you are about the demographics and type of medical needs to be served, the better you can target this patient.10

Segment. Divide the communities you are considering into different segments. For instance, even though you may do obstetrics and gynecologic surgery, consider breaking up the campaign to focus on 1 specific group, such as those interested in fibroid management.

Target. Identify the kinds of communities where you might find this patient. For example, if you want to focus on laparoscopic hysterectomies or myomectomies, start looking on Facebook for groups, pages, or website discussion boards or blogs that discuss abnormal uterine bleeding or fibroids and follow those pages.

Also, think about what other characteristics are associated with these ideal patients. For example, you might narrow it down to perimenopausal women with fibroids. A potential targeted group could be 40- to 50-year-old women who participate in yoga or running who have concerns about fibroids interfering in their active lifestyle. Perhaps this type of patient would want a minimally invasive surgical approach. A holistic health activist might be interested in nonsurgical management of fibroids.

Position. Once you have identified the specific community to target, position your practice within the community with the value proposition you are offering. For example, as an ObGyn who is focused on surgery, your position might be that your practice will provide the best experience for those medical services, with specific counseling to patients about resuming their active lifestyle.

 

Related article:
Four pillars of a successful practice: 2. Attract new patients

 

Get your potential patient to “raise her hand.” In the campaign, you are not trying to convince everyone up front to schedule an appointment from one post. First, try to get people who may be interested in your service(s) to “raise their hands.” Once your target market has expressed interest, either by their likes of your post, likes of your page, or other engagement, reach out to them with links for more information, such as free fibroid surgery education materials located on your website. On your website, create an opt-in page asking them to register their email address; once you have a compiled email list, send out monthly newsletters on your practice.11

 

Read how to guide patients to your office

 

 

Understand that marketing is a process

Think of marketing as an overall process in which you are guiding potential patients to come to your office. Your campaign has several steps; recognize that just one post will not make a huge difference. Use Facebook analytics to measure cost per engagement to calculate your return on investment and the campaign’s effectiveness, and revise as necessary.

Rather than just considering social media as a soap box to advertise your practice, break up the marketing process into 3 units: the before unit, the during unit, and the after unit.11 The word “unit” denotes the service, benefit, or product you are providing.

The before unit refers to the initial marketing that identifies potential patients—initially getting them to raise their hands and ultimately building an audience. (Once a potential patient provides her email address, you can send her a monthly newsletter or updates about your practice to continue the engagement.) Statistics show that an ObGyn needs to have 7 contacts, on average, with a patient over 18 months to “penetrate” her consciousness in a given market.12 Of course if there is an urgent or emergent need to see a physician, that timeline would be much shorter.

The during unit occurs when the patient comes to your practice and service is being provided. Since you know what she is coming for, you can create informational packets focused on her particular needs, perhaps about different management options for fibroids.

The after unit includes following up with the patient in some automated way. For those being treated for fibroids, it may be a reminder email that discusses the value of follow-up ultrasonography or the various kinds of surgical interventions for fibroids.

In order to continue your campaign, it is helpful to have a designated social media manager who will continue the social media posts and engagement.

When creating the posts, consider developing prescheduled assets (posts that are already produced with photos or links to articles), which can be done through Facebook or Hootsuite (http://www.hootsuite.com).

Manage the risks of social media interaction

There are risks associated with social media. Some things to consider are:

  • Policy. Develop a policy for your practice; if you work for an institution, align your policy with the institution’s.
  • Postings. Supervise content being posted. Never allow social media to be placed by someone without supervision. Either you should do this or assign a manager to be accountable to check on social media interactions so that any inappropriate comments can be addressed immediately.
  • Privacy. Never mention patients’ private health information or use the platform to publicly engage with a patient or future patient about their care. Do not post any references to patients or their photos without written consent.
  • Images. Use photographs and other images properly: obtain releases and obey copyright laws.

 

Related article:
Your patients are talking: Isn’t it time you take responsibility for your online reputation?

 

Bottom line

Social media is a powerful platform. Combined with good marketing strategies, social media campaigns can have a significant impact on expanding your practice to offer the kind of medical services you want to provide.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Definition of Marketing. American Marketing Association website. https://www.ama.org/AboutAMA/Pages/Definition-of-Marketing.aspx. Published July 2013. Accessed August 8, 2017.
  2. Kaplan AH, Haenlein M. Users of the world, unite! The challenges and opportunities of social media. Business Horiz. 2010;53(1):59–68.
  3. Lin KY, Lu HP. Intention to continue using Facebook fan pages from the perspective of social capital theory. Cyberpsychol Behav Soc Netw. 2011;14(10):565–570.
  4. Hawn C. Take two aspirin and tweet me in the morning: how Twitter, Facebook, and other social media are reshaping health care. Health Aff (Millwood). 2009;28(2):361–368.
  5. Wheeler CK, Said H, Prucz R, Rodrich RJ, Mathes DW. Social media in plastic surgery practices: emerging trends in North America. Aesthet Surg J. 2011;31(4):435–441.
  6. Nowak M, Spiller G. Two billion people coming together on Facebook. Facebook Newsroom. https://newsroom.fb.com/news/2017/06/two-billion-people-coming-together-on-facebook/. Published June 27, 2017. Accessed August 8, 2017.
  7. Adamson A. No contest: Twitter and Facebook can both play a role in branding. Forbes. http://www.forbes.com/2009/05/06/twitter-facebook-branding-leadership-cmo-network-adamson.html. Published May 6, 2009. Accessed August 8, 2017.
  8. Kim DS. Harness social media, enhance your practice. Contemp Obstet Gynecol. 2012;57(7):40–42,44–46.
  9. Wolf J. Social Media: Master, Manipulate, And Dominate Social Media Marketing Facebook, Twitter, YouTube, Instagram And LinkedIn. Createspace Independent Publishing Platform; 2015:129–143.
  10. Kotler PT, Keller KL. Marketing Management. 12th ed. Upper Saddle River, NJ: Prentice Hall; 2006:239–268.
  11. Jackson DP. Sunday marketing matinee: I love marketing live–Before, during, and after unit thinking. http://ilovemarketing.com/sunday-marketing-matineei-love-marketing-live-before-during-and-after-unit-thinking/. Accessed July 24, 2017.
  12. Payne D. How many contacts does it take before someone buys your product? Business Insider website. http://www.businessinsider.com/how-many-contacts-does-it-take-before-someone-buys-your-product-2011-7. Published July 12, 2011. Accessed August 8, 2017.
References
  1. Definition of Marketing. American Marketing Association website. https://www.ama.org/AboutAMA/Pages/Definition-of-Marketing.aspx. Published July 2013. Accessed August 8, 2017.
  2. Kaplan AH, Haenlein M. Users of the world, unite! The challenges and opportunities of social media. Business Horiz. 2010;53(1):59–68.
  3. Lin KY, Lu HP. Intention to continue using Facebook fan pages from the perspective of social capital theory. Cyberpsychol Behav Soc Netw. 2011;14(10):565–570.
  4. Hawn C. Take two aspirin and tweet me in the morning: how Twitter, Facebook, and other social media are reshaping health care. Health Aff (Millwood). 2009;28(2):361–368.
  5. Wheeler CK, Said H, Prucz R, Rodrich RJ, Mathes DW. Social media in plastic surgery practices: emerging trends in North America. Aesthet Surg J. 2011;31(4):435–441.
  6. Nowak M, Spiller G. Two billion people coming together on Facebook. Facebook Newsroom. https://newsroom.fb.com/news/2017/06/two-billion-people-coming-together-on-facebook/. Published June 27, 2017. Accessed August 8, 2017.
  7. Adamson A. No contest: Twitter and Facebook can both play a role in branding. Forbes. http://www.forbes.com/2009/05/06/twitter-facebook-branding-leadership-cmo-network-adamson.html. Published May 6, 2009. Accessed August 8, 2017.
  8. Kim DS. Harness social media, enhance your practice. Contemp Obstet Gynecol. 2012;57(7):40–42,44–46.
  9. Wolf J. Social Media: Master, Manipulate, And Dominate Social Media Marketing Facebook, Twitter, YouTube, Instagram And LinkedIn. Createspace Independent Publishing Platform; 2015:129–143.
  10. Kotler PT, Keller KL. Marketing Management. 12th ed. Upper Saddle River, NJ: Prentice Hall; 2006:239–268.
  11. Jackson DP. Sunday marketing matinee: I love marketing live–Before, during, and after unit thinking. http://ilovemarketing.com/sunday-marketing-matineei-love-marketing-live-before-during-and-after-unit-thinking/. Accessed July 24, 2017.
  12. Payne D. How many contacts does it take before someone buys your product? Business Insider website. http://www.businessinsider.com/how-many-contacts-does-it-take-before-someone-buys-your-product-2011-7. Published July 12, 2011. Accessed August 8, 2017.
Issue
OBG Management - 29(9)
Issue
OBG Management - 29(9)
Page Number
42-45
Page Number
42-45
Publications
Publications
Topics
Article Type
Display Headline
Create an effective social media campaign to market your practice: Here’s how
Display Headline
Create an effective social media campaign to market your practice: Here’s how
Sections
Inside the Article

Fast Tracks

  • Open a business Facebook page, compile an email list from those who like your postings, and send out useful information and updates on your practice
  • Develop an office policy for social media, supervise postings, ensure patient privacy, and obey copyright laws
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

Are combination estrogen-progestin oral contraceptives associated with an increased risk of cancer?

Article Type
Changed
Tue, 08/28/2018 - 11:09
Display Headline
Are combination estrogen-progestin oral contraceptives associated with an increased risk of cancer?

There are no large randomized clinical trials exploring the relationship between COCs and the risk of developing cancer. Many epidemiological studies, however, have investigated the possible association between COC use and the risk of cancer. Such prospective and retrospective studies consistently report that the use of COCs significantly decreases the risk of ovarian and endometrial cancer. The epidemiological data are less consistent concerning the possible association between COC use and the risk of breast cancer. Meta-analyses conclude that current use of COCs may be associated with a small increase in breast cancer risk. In addition, prolonged use of COCs may be associated with an increased risk of cervical cancer.

Ovarian cancer

COC use is associated with reduced risk of ovarian cancer, and the risk reduction persists after discontinuing COC use. In an individual data meta-analysis of 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 women without it, COC use was associated with a relative risk (RR) of 0.73 for ovarian cancer. The magnitude of risk reduction increased with increasing duration of COC use. The RR and 99% confidence interval (CI) for ovarian cancer and mean duration of use was1:

  • 0.78 (0.73–0.83) for 2.4 years
  • 0.64 (0.59–0.69) for 6.8 years
  • 0.56 (0.50–0.62) for 11.6 years
  • 0.42 (0.36–0.49) for 18.3 years.

In the Royal College of General Practitioners Oral Contraceptive (RCGPOC) study, about 23,000 womenwho did not use COCs and 23,000 current users of COCs were recruited around 1968 and followed for a median of 41 years. In this study, current and recent use of COCs was associated with a decreased RR for ovarian cancer (0.49) and the risk reduction persisted for at least 35 years following COC discontinuation (RR, 0.50; 99% CI, 0.29–0.84).2

In the prospective Nurses’ Health Study (NHS) I, 121,700 nurses were recruited in 1976 and followed for more than 30 years.3 For nurses who reported using COCs for more than 5 years, the rate ratio for ovarian cancer at 20 years or less and greater than 20 years since last use was 0.58 (95% CI, 0.61–0.87) and 0.92 (95% CI, 0.61–1.39), respectively. These studies show that the association between COC use and a decreased risk of ovarian cancer persists for many years after discontinuing COCs.

Endometrial cancer

COC use is associated with decreased risk of endometrial cancer, and the risk reduction persists for many years after discontinuing COC use. In an individual data meta-analysis of 36 studies that included 27,276 women with endometrial cancer and 115,743 women without it, COC use reduced the risk of endometrial cancer by approximately 25% for every 5 years of use. With 10 years of COC use the absolute risk of endometrial cancer before age 75 was 2.3 and 1.3 per 1,000 women for never and ever users of COC. Risk reduction varied slightly by histopathology, with risk reduction being greatest for type I endometrial cancer (RR, 0.68), slightly less for type II endometrial cancer (RR, 0.75), and lowest for endometrial sarcoma (RR, 0.83).4

In the RCGPOC study of 46,000 women, the RR of endometrial cancer among current and recent users of COCs was 0.61, and the reduced risk (0.83) persisted for more than 35 years after discontinuing the COC.2

 

Related article:
2016 Update on cancer: Endometrial cancer

 

It is thought that the progestin in the COC provides most of the beneficial effect. Progestin-only contraceptives, such as depotmedroxyprogesterone acetate, progestin implants, and levonorgestrel-releasingintrauterine devices (LNG-IUDs) are also thought to reduce endometrial cancer risk. For instance, in a study of 93,842 Finnish women who used the LNG-IUD, the standardized incidence ratio for endometrial cancer was 0.50 among LNG-IUD users compared with the general population.5

 

Read about the effects of COC use in breast and cervical cancer.

 

 

Breast cancer

The relationship between COC use and breast cancer is controversial. However, most oncologists believe that current use of COCs may be associated with a small increase in the risk of breast cancer diagnosis. The risk is attenuated after discontinuing COC use. In an individual data meta-analysis of 54 epidemiologicalstudies including 53,297 women with breast cancer and 100,239 without it, the RR of breast cancer with current COC use was 1.24 (95% CI, 1.15–1.33; P<.0001). The RR of breast cancer 10 years after stopping COCs was 1.01 (95% CI, 0.96–1.05; NS).6

In the prospective NHS study of 116,608 nurses with 1,246,967 years of follow-up, the multivariate relative risk (mRR) of breast cancer with current COC use was 1.33 (95% CI, 1.03–1.73). Past use of COCs was not associated with a significantly increased risk of breast cancer (mRR, 1.12; 95% CI, 0.95–1.33; NS).7

In the RCGPOC study (approximately 46,000 women), current use of COCs was associated with an increased risk of breast cancer (incidence rate ratio [IRR], 1.48; 95% CI,1.10–1.97). Five to 15 years after stopping COCs, there was no significant association between prior COC use and breast cancer (IRR, 1.12; 99% CI, 0.91–1.39; NS).2

 

Related article:
Webcast: Oral contraceptives and breast cancer: What’s the risk?

 

It is important to note that it is not possible to conclude from these data whether the reported association between current use of COCs and breast cancer is due to early and accelerated diagnosis of breast cancer, the biological effects of hormones contained in COCs on breast tissue and nascent tumors, or both. In addition, formulations of COCs prescribed in the 1960s and 1970s contained higher doses of estrogen, raising the possibility that the association between COCs and breast cancer is due to COC formulations that are no longer prescribed. However, in animal models and postmenopausal women certain combinations of estrogen plus progestin clearly influence breast cancer biology and cancer risk.8,9

COC use among BRCA1 and BRCA2 carriers

Women carrying BRCA1 and BRCA2 mutations, which increase the risk of ovarian and breast cancer, are often counseled to consider bilateral salpingectomy between age 35 and 40 years to reduce the risk of developing ovarian cancer. An important clinical question is what is the impact of combination estrogen-progestin oral contraceptives (COC) use on ovarian and breast cancer risk among these women?

Meta-analyses of the association between COC use and ovarian cancer consistently report that COC use reduces the risk of ovarian cancer in women with clinically important BRCA1 and BRCA2 mutations.1,2 For example, a meta-analysis of 6 studies reported that women with BRCA1 and BRCA2 mutations who used COCs had a significantly decreased risk of ovarian cancer (odds ratio [OR], 0.58; 95% CI, 0.46–0.73).1

The association between COC use and breast cancer risk is not clear. One meta-analysis reported no significant association between COC use and breast cancer risk among BRCA mutation carriers (OR, 1.21; 95% CI, 0.93–1.58).1 Another meta-analysis reported a significant association between COC use before 1975 and breast cancer risk (RR, 1.47; 95% CI, 1.06–2.04) but not with recent low-estrogen formulations of COC (RR, 1.17; 95% CI, 0.74–1.86).2

Based on the available data, the Society of Gynecologic Oncologists recommends that women with clinically significant BRCA1 and BRCA2 mutations be offered chemoprevention with COCs because the benefit of ovarian cancer risk reduction outweighs the possible impact on breast cancer risk.3 A contrarian view-point espoused by some oncologists is that since women with BRCA mutations should have their ovaries removed prior to getting ovarian cancer, the clinical utility of recommending COC chemoprevention of ovarian cancer is largely irrelevant.

References

  1. Moorman PG, Havrilesky LJ, Gierisch JM, et al. Oral contraceptives and risk of ovarian cancer and breast cancer among high-risk women: a systematic review and meta-analysis. J Clin Oncol. 2013;31(33):4188–4198.
  2. Iodice S, Barile M, Rotmensz N, et al. Oral contraceptive use and breast or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Canc. 2010;46(12):2275–2284.
  3. Walker JL, Powell CB, Chen LM, et al. Society of Gynecologic Oncology recommendations for the prevention of ovarian cancer. Cancer. 2015;121(13):2108–2120.

Cervical cancer

Prolonged COC use is associated with an increased risk of cervical cancer. The risk is no longer observed 10 years after stopping COC use. In an individual data meta-analysis of 24 epidemiological studies including 16,573 women with cervical cancer and 35,509 women without it, the relative risk of cervical cancer with less than 5 years or 5 or more years of COC use was 1.09 and 1.90, respectively. Analyses of potential confounding exposures, including age at first sexual intercourse, condom use, cigarette smoking, and number of sexual partners, did not significantly weaken the observed association between cervical cancer and COC use of 5 or more years.10 In a study of women who were positive for HPV DNA, the odds ratio for cervical cancer among women who had used COCs11:

  • less than 5 years, 0.73 (95% CI, 0.52–1.03)
  • 5 to 9 years, 2.82 (95% CI, 1.46–5.42)
  • ≥10 years, 4.03 (95% CI, 2.09–8.02).

It is not possible to conclude from these data whether the association between COC use and cervical cancer is due to the biological effects of hormones on the initiation and progression of HPV disease or confounding factors that have yet to be identified. It is known that estrogens and progestins influence the immune defense system of the lower genital tract, and this may be a pathway that influences the acquisition and progression of viral disease.12 From a clinical perspective, cervical cancer is largely preventable with HPV vaccination and screening. Therefore, the risk between COC use and cervical cancer is likely limited to women who have not been vaccinated and who are not actively participating in cervical cancer screening.

The bottom line

COC use markedly reduces the risk of ovarian and endometrial cancers, and slightly increases the risk of breast cancer. Prolonged COC use may be associated with an increased risk of cervical cancer. Using available epidemiological data, investigators attempted to project the impact of these competing risks on the approximate 12,300,000 females who live in Australia. Based on the pattern of COC use and the cancer incidence in Australia in 2010, the investigators calculated that COC use would cause about 105 breast and 52 cervical cancers and prevent 1,032 endometrial and 308 ovarian cancers.13 This analysis indicates that the balance of risks and benefits related to COC use and cancer generally favors COC use.

Prevention of unintended pregnancy is a major public health goal. Many women choose COCs as their preferred approach to preventing unintended pregnancy. Evaluated from a whole-life perspective the health benefits of COCs are substantial and represent a great advance in women’s health.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Beral V, Doll R, Hermon C, Peto R, Reeves G; Collaborative Group on Epidemiological Studies of Ovarian Cancer. Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 controls. Lancet. 2008;371(9609):303–314.
  2. Iversen L, Sivasubramaniam S, Lee AJ, Fielding S, Hannaford PC. Lifetime cancer risk and combined oral contraceptives: the Royal College of General Practitioners’ Oral Contraception Study. Am J Obstet Gynecol. 2017;216(6):580.e1–e9.
  3. Tworoger SS, Fairfield KM, Colditz GA, Rosner BA, Hankinson SE. Association of oral contraceptive use, other contraceptive methods, and infertility with ovarian cancer risk. Am J Epidemiol. 2007;166(8):894–901.
  4. Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol. 2015;16(9):1061–1070.
  5. Soini T, Hurskainen R, Grénman S, Mäenpää J, Paavonen J, Pukkala E. Cancer risk in women using the levonorgestrel-releasing intrauterine system in Finland. Obstet Gynecol. 2014;124(2 pt 1):292–299.
  6. Collaborative Group on Hormonal Factors in Breast Cancer. Breast cancer and hormonal contraceptives: collaborative reanalysis of individual data on 53,297 women with breast cancer and 100,239 women without breast cancer from 54 epidemiological studies. Lancet. 1996;347(9017):1713–1727.
  7. Hunter DJ, Colditz GA, Hankinson SE, et al. Oral contraceptive use and breast cancer: a prospective study of young women. Cancer Epidemiol Biomarkers Prev. 2010;19(10):2496–2502.
  8. Simões BM, Alferez DG, Howell SJ, Clarke RB. The role of steroid hormones in breast cancer stem cells. Endocr Relat Cancer. 2015;22(6):T177–T186.
  9. Chlebowski RT, Manson JE, Anderson GL, et al. Estrogen plus progestin and breast cancer incidence and mortality in the Women’s Health Initiative Observational Study. J Natl Cancer Inst. 2013;105(8):526–535.
  10. International Collaboration of Epidemiological Studies of Cervical Cancer. Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet. 2007;370(9599):1609–1621.
  11. Moreno V, Bosch FX, Muñoz N, et al. Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet. 2002;359(9312):1085–1092.
  12. Fichorova RN, Chen PL, Morrison CS, et al. The contribution of cervicovaginal infections to the immunomodulatory effects of hormonal contraception. MBio. 2015;6(5):e00221–e002215.
  13. Jordan SJ, Wilson LF, Nagle CM, et al. Cancers in Australia in 2010 attributable to and prevented by the use of combined oral contraceptives. Aust N Z J Public Health. 2015;39(5):441–445.
Article PDF
Author and Disclosure Information

Dr. Barbieri is Editor in Chief, OBG Management; Chair, Obstetrics and Gynecology, Brigham and Women’s Hospital; and Kate Macy Ladd Professor of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts.

Dr. Barbieri reports no financial relationships relevant to this article.

Issue
OBG Management - 29(9)
Publications
Topics
Page Number
7, 13-14, 16
Sections
Author and Disclosure Information

Dr. Barbieri is Editor in Chief, OBG Management; Chair, Obstetrics and Gynecology, Brigham and Women’s Hospital; and Kate Macy Ladd Professor of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts.

Dr. Barbieri reports no financial relationships relevant to this article.

Author and Disclosure Information

Dr. Barbieri is Editor in Chief, OBG Management; Chair, Obstetrics and Gynecology, Brigham and Women’s Hospital; and Kate Macy Ladd Professor of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts.

Dr. Barbieri reports no financial relationships relevant to this article.

Article PDF
Article PDF

There are no large randomized clinical trials exploring the relationship between COCs and the risk of developing cancer. Many epidemiological studies, however, have investigated the possible association between COC use and the risk of cancer. Such prospective and retrospective studies consistently report that the use of COCs significantly decreases the risk of ovarian and endometrial cancer. The epidemiological data are less consistent concerning the possible association between COC use and the risk of breast cancer. Meta-analyses conclude that current use of COCs may be associated with a small increase in breast cancer risk. In addition, prolonged use of COCs may be associated with an increased risk of cervical cancer.

Ovarian cancer

COC use is associated with reduced risk of ovarian cancer, and the risk reduction persists after discontinuing COC use. In an individual data meta-analysis of 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 women without it, COC use was associated with a relative risk (RR) of 0.73 for ovarian cancer. The magnitude of risk reduction increased with increasing duration of COC use. The RR and 99% confidence interval (CI) for ovarian cancer and mean duration of use was1:

  • 0.78 (0.73–0.83) for 2.4 years
  • 0.64 (0.59–0.69) for 6.8 years
  • 0.56 (0.50–0.62) for 11.6 years
  • 0.42 (0.36–0.49) for 18.3 years.

In the Royal College of General Practitioners Oral Contraceptive (RCGPOC) study, about 23,000 womenwho did not use COCs and 23,000 current users of COCs were recruited around 1968 and followed for a median of 41 years. In this study, current and recent use of COCs was associated with a decreased RR for ovarian cancer (0.49) and the risk reduction persisted for at least 35 years following COC discontinuation (RR, 0.50; 99% CI, 0.29–0.84).2

In the prospective Nurses’ Health Study (NHS) I, 121,700 nurses were recruited in 1976 and followed for more than 30 years.3 For nurses who reported using COCs for more than 5 years, the rate ratio for ovarian cancer at 20 years or less and greater than 20 years since last use was 0.58 (95% CI, 0.61–0.87) and 0.92 (95% CI, 0.61–1.39), respectively. These studies show that the association between COC use and a decreased risk of ovarian cancer persists for many years after discontinuing COCs.

Endometrial cancer

COC use is associated with decreased risk of endometrial cancer, and the risk reduction persists for many years after discontinuing COC use. In an individual data meta-analysis of 36 studies that included 27,276 women with endometrial cancer and 115,743 women without it, COC use reduced the risk of endometrial cancer by approximately 25% for every 5 years of use. With 10 years of COC use the absolute risk of endometrial cancer before age 75 was 2.3 and 1.3 per 1,000 women for never and ever users of COC. Risk reduction varied slightly by histopathology, with risk reduction being greatest for type I endometrial cancer (RR, 0.68), slightly less for type II endometrial cancer (RR, 0.75), and lowest for endometrial sarcoma (RR, 0.83).4

In the RCGPOC study of 46,000 women, the RR of endometrial cancer among current and recent users of COCs was 0.61, and the reduced risk (0.83) persisted for more than 35 years after discontinuing the COC.2

 

Related article:
2016 Update on cancer: Endometrial cancer

 

It is thought that the progestin in the COC provides most of the beneficial effect. Progestin-only contraceptives, such as depotmedroxyprogesterone acetate, progestin implants, and levonorgestrel-releasingintrauterine devices (LNG-IUDs) are also thought to reduce endometrial cancer risk. For instance, in a study of 93,842 Finnish women who used the LNG-IUD, the standardized incidence ratio for endometrial cancer was 0.50 among LNG-IUD users compared with the general population.5

 

Read about the effects of COC use in breast and cervical cancer.

 

 

Breast cancer

The relationship between COC use and breast cancer is controversial. However, most oncologists believe that current use of COCs may be associated with a small increase in the risk of breast cancer diagnosis. The risk is attenuated after discontinuing COC use. In an individual data meta-analysis of 54 epidemiologicalstudies including 53,297 women with breast cancer and 100,239 without it, the RR of breast cancer with current COC use was 1.24 (95% CI, 1.15–1.33; P<.0001). The RR of breast cancer 10 years after stopping COCs was 1.01 (95% CI, 0.96–1.05; NS).6

In the prospective NHS study of 116,608 nurses with 1,246,967 years of follow-up, the multivariate relative risk (mRR) of breast cancer with current COC use was 1.33 (95% CI, 1.03–1.73). Past use of COCs was not associated with a significantly increased risk of breast cancer (mRR, 1.12; 95% CI, 0.95–1.33; NS).7

In the RCGPOC study (approximately 46,000 women), current use of COCs was associated with an increased risk of breast cancer (incidence rate ratio [IRR], 1.48; 95% CI,1.10–1.97). Five to 15 years after stopping COCs, there was no significant association between prior COC use and breast cancer (IRR, 1.12; 99% CI, 0.91–1.39; NS).2

 

Related article:
Webcast: Oral contraceptives and breast cancer: What’s the risk?

 

It is important to note that it is not possible to conclude from these data whether the reported association between current use of COCs and breast cancer is due to early and accelerated diagnosis of breast cancer, the biological effects of hormones contained in COCs on breast tissue and nascent tumors, or both. In addition, formulations of COCs prescribed in the 1960s and 1970s contained higher doses of estrogen, raising the possibility that the association between COCs and breast cancer is due to COC formulations that are no longer prescribed. However, in animal models and postmenopausal women certain combinations of estrogen plus progestin clearly influence breast cancer biology and cancer risk.8,9

COC use among BRCA1 and BRCA2 carriers

Women carrying BRCA1 and BRCA2 mutations, which increase the risk of ovarian and breast cancer, are often counseled to consider bilateral salpingectomy between age 35 and 40 years to reduce the risk of developing ovarian cancer. An important clinical question is what is the impact of combination estrogen-progestin oral contraceptives (COC) use on ovarian and breast cancer risk among these women?

Meta-analyses of the association between COC use and ovarian cancer consistently report that COC use reduces the risk of ovarian cancer in women with clinically important BRCA1 and BRCA2 mutations.1,2 For example, a meta-analysis of 6 studies reported that women with BRCA1 and BRCA2 mutations who used COCs had a significantly decreased risk of ovarian cancer (odds ratio [OR], 0.58; 95% CI, 0.46–0.73).1

The association between COC use and breast cancer risk is not clear. One meta-analysis reported no significant association between COC use and breast cancer risk among BRCA mutation carriers (OR, 1.21; 95% CI, 0.93–1.58).1 Another meta-analysis reported a significant association between COC use before 1975 and breast cancer risk (RR, 1.47; 95% CI, 1.06–2.04) but not with recent low-estrogen formulations of COC (RR, 1.17; 95% CI, 0.74–1.86).2

Based on the available data, the Society of Gynecologic Oncologists recommends that women with clinically significant BRCA1 and BRCA2 mutations be offered chemoprevention with COCs because the benefit of ovarian cancer risk reduction outweighs the possible impact on breast cancer risk.3 A contrarian view-point espoused by some oncologists is that since women with BRCA mutations should have their ovaries removed prior to getting ovarian cancer, the clinical utility of recommending COC chemoprevention of ovarian cancer is largely irrelevant.

References

  1. Moorman PG, Havrilesky LJ, Gierisch JM, et al. Oral contraceptives and risk of ovarian cancer and breast cancer among high-risk women: a systematic review and meta-analysis. J Clin Oncol. 2013;31(33):4188–4198.
  2. Iodice S, Barile M, Rotmensz N, et al. Oral contraceptive use and breast or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Canc. 2010;46(12):2275–2284.
  3. Walker JL, Powell CB, Chen LM, et al. Society of Gynecologic Oncology recommendations for the prevention of ovarian cancer. Cancer. 2015;121(13):2108–2120.

Cervical cancer

Prolonged COC use is associated with an increased risk of cervical cancer. The risk is no longer observed 10 years after stopping COC use. In an individual data meta-analysis of 24 epidemiological studies including 16,573 women with cervical cancer and 35,509 women without it, the relative risk of cervical cancer with less than 5 years or 5 or more years of COC use was 1.09 and 1.90, respectively. Analyses of potential confounding exposures, including age at first sexual intercourse, condom use, cigarette smoking, and number of sexual partners, did not significantly weaken the observed association between cervical cancer and COC use of 5 or more years.10 In a study of women who were positive for HPV DNA, the odds ratio for cervical cancer among women who had used COCs11:

  • less than 5 years, 0.73 (95% CI, 0.52–1.03)
  • 5 to 9 years, 2.82 (95% CI, 1.46–5.42)
  • ≥10 years, 4.03 (95% CI, 2.09–8.02).

It is not possible to conclude from these data whether the association between COC use and cervical cancer is due to the biological effects of hormones on the initiation and progression of HPV disease or confounding factors that have yet to be identified. It is known that estrogens and progestins influence the immune defense system of the lower genital tract, and this may be a pathway that influences the acquisition and progression of viral disease.12 From a clinical perspective, cervical cancer is largely preventable with HPV vaccination and screening. Therefore, the risk between COC use and cervical cancer is likely limited to women who have not been vaccinated and who are not actively participating in cervical cancer screening.

The bottom line

COC use markedly reduces the risk of ovarian and endometrial cancers, and slightly increases the risk of breast cancer. Prolonged COC use may be associated with an increased risk of cervical cancer. Using available epidemiological data, investigators attempted to project the impact of these competing risks on the approximate 12,300,000 females who live in Australia. Based on the pattern of COC use and the cancer incidence in Australia in 2010, the investigators calculated that COC use would cause about 105 breast and 52 cervical cancers and prevent 1,032 endometrial and 308 ovarian cancers.13 This analysis indicates that the balance of risks and benefits related to COC use and cancer generally favors COC use.

Prevention of unintended pregnancy is a major public health goal. Many women choose COCs as their preferred approach to preventing unintended pregnancy. Evaluated from a whole-life perspective the health benefits of COCs are substantial and represent a great advance in women’s health.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

There are no large randomized clinical trials exploring the relationship between COCs and the risk of developing cancer. Many epidemiological studies, however, have investigated the possible association between COC use and the risk of cancer. Such prospective and retrospective studies consistently report that the use of COCs significantly decreases the risk of ovarian and endometrial cancer. The epidemiological data are less consistent concerning the possible association between COC use and the risk of breast cancer. Meta-analyses conclude that current use of COCs may be associated with a small increase in breast cancer risk. In addition, prolonged use of COCs may be associated with an increased risk of cervical cancer.

Ovarian cancer

COC use is associated with reduced risk of ovarian cancer, and the risk reduction persists after discontinuing COC use. In an individual data meta-analysis of 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 women without it, COC use was associated with a relative risk (RR) of 0.73 for ovarian cancer. The magnitude of risk reduction increased with increasing duration of COC use. The RR and 99% confidence interval (CI) for ovarian cancer and mean duration of use was1:

  • 0.78 (0.73–0.83) for 2.4 years
  • 0.64 (0.59–0.69) for 6.8 years
  • 0.56 (0.50–0.62) for 11.6 years
  • 0.42 (0.36–0.49) for 18.3 years.

In the Royal College of General Practitioners Oral Contraceptive (RCGPOC) study, about 23,000 womenwho did not use COCs and 23,000 current users of COCs were recruited around 1968 and followed for a median of 41 years. In this study, current and recent use of COCs was associated with a decreased RR for ovarian cancer (0.49) and the risk reduction persisted for at least 35 years following COC discontinuation (RR, 0.50; 99% CI, 0.29–0.84).2

In the prospective Nurses’ Health Study (NHS) I, 121,700 nurses were recruited in 1976 and followed for more than 30 years.3 For nurses who reported using COCs for more than 5 years, the rate ratio for ovarian cancer at 20 years or less and greater than 20 years since last use was 0.58 (95% CI, 0.61–0.87) and 0.92 (95% CI, 0.61–1.39), respectively. These studies show that the association between COC use and a decreased risk of ovarian cancer persists for many years after discontinuing COCs.

Endometrial cancer

COC use is associated with decreased risk of endometrial cancer, and the risk reduction persists for many years after discontinuing COC use. In an individual data meta-analysis of 36 studies that included 27,276 women with endometrial cancer and 115,743 women without it, COC use reduced the risk of endometrial cancer by approximately 25% for every 5 years of use. With 10 years of COC use the absolute risk of endometrial cancer before age 75 was 2.3 and 1.3 per 1,000 women for never and ever users of COC. Risk reduction varied slightly by histopathology, with risk reduction being greatest for type I endometrial cancer (RR, 0.68), slightly less for type II endometrial cancer (RR, 0.75), and lowest for endometrial sarcoma (RR, 0.83).4

In the RCGPOC study of 46,000 women, the RR of endometrial cancer among current and recent users of COCs was 0.61, and the reduced risk (0.83) persisted for more than 35 years after discontinuing the COC.2

 

Related article:
2016 Update on cancer: Endometrial cancer

 

It is thought that the progestin in the COC provides most of the beneficial effect. Progestin-only contraceptives, such as depotmedroxyprogesterone acetate, progestin implants, and levonorgestrel-releasingintrauterine devices (LNG-IUDs) are also thought to reduce endometrial cancer risk. For instance, in a study of 93,842 Finnish women who used the LNG-IUD, the standardized incidence ratio for endometrial cancer was 0.50 among LNG-IUD users compared with the general population.5

 

Read about the effects of COC use in breast and cervical cancer.

 

 

Breast cancer

The relationship between COC use and breast cancer is controversial. However, most oncologists believe that current use of COCs may be associated with a small increase in the risk of breast cancer diagnosis. The risk is attenuated after discontinuing COC use. In an individual data meta-analysis of 54 epidemiologicalstudies including 53,297 women with breast cancer and 100,239 without it, the RR of breast cancer with current COC use was 1.24 (95% CI, 1.15–1.33; P<.0001). The RR of breast cancer 10 years after stopping COCs was 1.01 (95% CI, 0.96–1.05; NS).6

In the prospective NHS study of 116,608 nurses with 1,246,967 years of follow-up, the multivariate relative risk (mRR) of breast cancer with current COC use was 1.33 (95% CI, 1.03–1.73). Past use of COCs was not associated with a significantly increased risk of breast cancer (mRR, 1.12; 95% CI, 0.95–1.33; NS).7

In the RCGPOC study (approximately 46,000 women), current use of COCs was associated with an increased risk of breast cancer (incidence rate ratio [IRR], 1.48; 95% CI,1.10–1.97). Five to 15 years after stopping COCs, there was no significant association between prior COC use and breast cancer (IRR, 1.12; 99% CI, 0.91–1.39; NS).2

 

Related article:
Webcast: Oral contraceptives and breast cancer: What’s the risk?

 

It is important to note that it is not possible to conclude from these data whether the reported association between current use of COCs and breast cancer is due to early and accelerated diagnosis of breast cancer, the biological effects of hormones contained in COCs on breast tissue and nascent tumors, or both. In addition, formulations of COCs prescribed in the 1960s and 1970s contained higher doses of estrogen, raising the possibility that the association between COCs and breast cancer is due to COC formulations that are no longer prescribed. However, in animal models and postmenopausal women certain combinations of estrogen plus progestin clearly influence breast cancer biology and cancer risk.8,9

COC use among BRCA1 and BRCA2 carriers

Women carrying BRCA1 and BRCA2 mutations, which increase the risk of ovarian and breast cancer, are often counseled to consider bilateral salpingectomy between age 35 and 40 years to reduce the risk of developing ovarian cancer. An important clinical question is what is the impact of combination estrogen-progestin oral contraceptives (COC) use on ovarian and breast cancer risk among these women?

Meta-analyses of the association between COC use and ovarian cancer consistently report that COC use reduces the risk of ovarian cancer in women with clinically important BRCA1 and BRCA2 mutations.1,2 For example, a meta-analysis of 6 studies reported that women with BRCA1 and BRCA2 mutations who used COCs had a significantly decreased risk of ovarian cancer (odds ratio [OR], 0.58; 95% CI, 0.46–0.73).1

The association between COC use and breast cancer risk is not clear. One meta-analysis reported no significant association between COC use and breast cancer risk among BRCA mutation carriers (OR, 1.21; 95% CI, 0.93–1.58).1 Another meta-analysis reported a significant association between COC use before 1975 and breast cancer risk (RR, 1.47; 95% CI, 1.06–2.04) but not with recent low-estrogen formulations of COC (RR, 1.17; 95% CI, 0.74–1.86).2

Based on the available data, the Society of Gynecologic Oncologists recommends that women with clinically significant BRCA1 and BRCA2 mutations be offered chemoprevention with COCs because the benefit of ovarian cancer risk reduction outweighs the possible impact on breast cancer risk.3 A contrarian view-point espoused by some oncologists is that since women with BRCA mutations should have their ovaries removed prior to getting ovarian cancer, the clinical utility of recommending COC chemoprevention of ovarian cancer is largely irrelevant.

References

  1. Moorman PG, Havrilesky LJ, Gierisch JM, et al. Oral contraceptives and risk of ovarian cancer and breast cancer among high-risk women: a systematic review and meta-analysis. J Clin Oncol. 2013;31(33):4188–4198.
  2. Iodice S, Barile M, Rotmensz N, et al. Oral contraceptive use and breast or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Canc. 2010;46(12):2275–2284.
  3. Walker JL, Powell CB, Chen LM, et al. Society of Gynecologic Oncology recommendations for the prevention of ovarian cancer. Cancer. 2015;121(13):2108–2120.

Cervical cancer

Prolonged COC use is associated with an increased risk of cervical cancer. The risk is no longer observed 10 years after stopping COC use. In an individual data meta-analysis of 24 epidemiological studies including 16,573 women with cervical cancer and 35,509 women without it, the relative risk of cervical cancer with less than 5 years or 5 or more years of COC use was 1.09 and 1.90, respectively. Analyses of potential confounding exposures, including age at first sexual intercourse, condom use, cigarette smoking, and number of sexual partners, did not significantly weaken the observed association between cervical cancer and COC use of 5 or more years.10 In a study of women who were positive for HPV DNA, the odds ratio for cervical cancer among women who had used COCs11:

  • less than 5 years, 0.73 (95% CI, 0.52–1.03)
  • 5 to 9 years, 2.82 (95% CI, 1.46–5.42)
  • ≥10 years, 4.03 (95% CI, 2.09–8.02).

It is not possible to conclude from these data whether the association between COC use and cervical cancer is due to the biological effects of hormones on the initiation and progression of HPV disease or confounding factors that have yet to be identified. It is known that estrogens and progestins influence the immune defense system of the lower genital tract, and this may be a pathway that influences the acquisition and progression of viral disease.12 From a clinical perspective, cervical cancer is largely preventable with HPV vaccination and screening. Therefore, the risk between COC use and cervical cancer is likely limited to women who have not been vaccinated and who are not actively participating in cervical cancer screening.

The bottom line

COC use markedly reduces the risk of ovarian and endometrial cancers, and slightly increases the risk of breast cancer. Prolonged COC use may be associated with an increased risk of cervical cancer. Using available epidemiological data, investigators attempted to project the impact of these competing risks on the approximate 12,300,000 females who live in Australia. Based on the pattern of COC use and the cancer incidence in Australia in 2010, the investigators calculated that COC use would cause about 105 breast and 52 cervical cancers and prevent 1,032 endometrial and 308 ovarian cancers.13 This analysis indicates that the balance of risks and benefits related to COC use and cancer generally favors COC use.

Prevention of unintended pregnancy is a major public health goal. Many women choose COCs as their preferred approach to preventing unintended pregnancy. Evaluated from a whole-life perspective the health benefits of COCs are substantial and represent a great advance in women’s health.

 

Share your thoughts! Send your Letter to the Editor to [email protected]. Please include your name and the city and state in which you practice.

References
  1. Beral V, Doll R, Hermon C, Peto R, Reeves G; Collaborative Group on Epidemiological Studies of Ovarian Cancer. Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 controls. Lancet. 2008;371(9609):303–314.
  2. Iversen L, Sivasubramaniam S, Lee AJ, Fielding S, Hannaford PC. Lifetime cancer risk and combined oral contraceptives: the Royal College of General Practitioners’ Oral Contraception Study. Am J Obstet Gynecol. 2017;216(6):580.e1–e9.
  3. Tworoger SS, Fairfield KM, Colditz GA, Rosner BA, Hankinson SE. Association of oral contraceptive use, other contraceptive methods, and infertility with ovarian cancer risk. Am J Epidemiol. 2007;166(8):894–901.
  4. Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol. 2015;16(9):1061–1070.
  5. Soini T, Hurskainen R, Grénman S, Mäenpää J, Paavonen J, Pukkala E. Cancer risk in women using the levonorgestrel-releasing intrauterine system in Finland. Obstet Gynecol. 2014;124(2 pt 1):292–299.
  6. Collaborative Group on Hormonal Factors in Breast Cancer. Breast cancer and hormonal contraceptives: collaborative reanalysis of individual data on 53,297 women with breast cancer and 100,239 women without breast cancer from 54 epidemiological studies. Lancet. 1996;347(9017):1713–1727.
  7. Hunter DJ, Colditz GA, Hankinson SE, et al. Oral contraceptive use and breast cancer: a prospective study of young women. Cancer Epidemiol Biomarkers Prev. 2010;19(10):2496–2502.
  8. Simões BM, Alferez DG, Howell SJ, Clarke RB. The role of steroid hormones in breast cancer stem cells. Endocr Relat Cancer. 2015;22(6):T177–T186.
  9. Chlebowski RT, Manson JE, Anderson GL, et al. Estrogen plus progestin and breast cancer incidence and mortality in the Women’s Health Initiative Observational Study. J Natl Cancer Inst. 2013;105(8):526–535.
  10. International Collaboration of Epidemiological Studies of Cervical Cancer. Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet. 2007;370(9599):1609–1621.
  11. Moreno V, Bosch FX, Muñoz N, et al. Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet. 2002;359(9312):1085–1092.
  12. Fichorova RN, Chen PL, Morrison CS, et al. The contribution of cervicovaginal infections to the immunomodulatory effects of hormonal contraception. MBio. 2015;6(5):e00221–e002215.
  13. Jordan SJ, Wilson LF, Nagle CM, et al. Cancers in Australia in 2010 attributable to and prevented by the use of combined oral contraceptives. Aust N Z J Public Health. 2015;39(5):441–445.
References
  1. Beral V, Doll R, Hermon C, Peto R, Reeves G; Collaborative Group on Epidemiological Studies of Ovarian Cancer. Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 controls. Lancet. 2008;371(9609):303–314.
  2. Iversen L, Sivasubramaniam S, Lee AJ, Fielding S, Hannaford PC. Lifetime cancer risk and combined oral contraceptives: the Royal College of General Practitioners’ Oral Contraception Study. Am J Obstet Gynecol. 2017;216(6):580.e1–e9.
  3. Tworoger SS, Fairfield KM, Colditz GA, Rosner BA, Hankinson SE. Association of oral contraceptive use, other contraceptive methods, and infertility with ovarian cancer risk. Am J Epidemiol. 2007;166(8):894–901.
  4. Collaborative Group on Epidemiological Studies on Endometrial Cancer. Endometrial cancer and oral contraceptives: an individual participant meta-analysis of 27,276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol. 2015;16(9):1061–1070.
  5. Soini T, Hurskainen R, Grénman S, Mäenpää J, Paavonen J, Pukkala E. Cancer risk in women using the levonorgestrel-releasing intrauterine system in Finland. Obstet Gynecol. 2014;124(2 pt 1):292–299.
  6. Collaborative Group on Hormonal Factors in Breast Cancer. Breast cancer and hormonal contraceptives: collaborative reanalysis of individual data on 53,297 women with breast cancer and 100,239 women without breast cancer from 54 epidemiological studies. Lancet. 1996;347(9017):1713–1727.
  7. Hunter DJ, Colditz GA, Hankinson SE, et al. Oral contraceptive use and breast cancer: a prospective study of young women. Cancer Epidemiol Biomarkers Prev. 2010;19(10):2496–2502.
  8. Simões BM, Alferez DG, Howell SJ, Clarke RB. The role of steroid hormones in breast cancer stem cells. Endocr Relat Cancer. 2015;22(6):T177–T186.
  9. Chlebowski RT, Manson JE, Anderson GL, et al. Estrogen plus progestin and breast cancer incidence and mortality in the Women’s Health Initiative Observational Study. J Natl Cancer Inst. 2013;105(8):526–535.
  10. International Collaboration of Epidemiological Studies of Cervical Cancer. Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet. 2007;370(9599):1609–1621.
  11. Moreno V, Bosch FX, Muñoz N, et al. Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet. 2002;359(9312):1085–1092.
  12. Fichorova RN, Chen PL, Morrison CS, et al. The contribution of cervicovaginal infections to the immunomodulatory effects of hormonal contraception. MBio. 2015;6(5):e00221–e002215.
  13. Jordan SJ, Wilson LF, Nagle CM, et al. Cancers in Australia in 2010 attributable to and prevented by the use of combined oral contraceptives. Aust N Z J Public Health. 2015;39(5):441–445.
Issue
OBG Management - 29(9)
Issue
OBG Management - 29(9)
Page Number
7, 13-14, 16
Page Number
7, 13-14, 16
Publications
Publications
Topics
Article Type
Display Headline
Are combination estrogen-progestin oral contraceptives associated with an increased risk of cancer?
Display Headline
Are combination estrogen-progestin oral contraceptives associated with an increased risk of cancer?
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Biostatistics and epidemiology lecture series, part 1

Article Type
Changed
Wed, 04/10/2019 - 11:08
Display Headline
Biostatistics and epidemiology lecture series, part 1
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Publications
Topics
Page Number
e1-e25
Sections
Article PDF
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Issue
Cleveland Clinic Journal of Medicine - 84(9)
Page Number
e1-e25
Page Number
e1-e25
Publications
Publications
Topics
Article Type
Display Headline
Biostatistics and epidemiology lecture series, part 1
Display Headline
Biostatistics and epidemiology lecture series, part 1
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2017 September;84(9 suppl 2):e1-e25
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Introduction: Biostatistics and epidemiology lecture series, part 1

Article Type
Changed
Mon, 10/01/2018 - 14:34
Display Headline
Introduction: Biostatistics and epidemiology lecture series, part 1

Physicians are inundated with clinical research findings that potentially impact patient care. Evaluating the strength and clinical application of research results requires an understanding of the underlying biostatistics and epidemiological principles.

The articles in this supplement are based on a series of lectures originally developed for fellows in pulmonary and critical care medicine to provide them with the tools to transform a scientific or clinical question into research projects, and then pursue the answer to their question with the appropriate methods. The same skills also enable them to appraise the published literature in a systematic and rigorous manner. 

Each topic in the series began with a presentation and discussion of statistical principles and methods, then moved to a practical module using the principles to appraise a specific publication. Participants in the course had an immediate opportunity to try the techniques, both to demonstrate understanding and to reinforce the concepts to each learner. The articles of this series follow the same outline, providing clinicians of all specialties the basic statistical tools to conduct and appraise clinical research, along with a sample article for practicing each statistical method presented.

This Cleveland Clinic Journal of Medicine supplement includes 3 lectures from the “Biostatistics and Epidemiology Lecture Series.” Dr. Stoller’s presentation, The Architecture of Clinical Research, describes the basic structure of clinical research and the nomenclature to understand trial design and sources of bias.

Building on those concepts, Dr. Chatburn’s lecture, Basics of Study Design: Practical Considerations, outlines the structured approach to develop a formal research protocol. How to identify a problem, expand the scope of it through a literature review, create a hypothesis, design a study, and an introduction to basic statistical methods are discussed.

And in Chi-square and Fisher’s Exact Tests, Dr. Nowacki introduces the statistical methodology of these 2 tests to assess associations between 2 independent categorical variables. The sample article illustrates step-by-step calculation of both the large sample approximation (chi-square) and exact (Fisher’s) methodologies providing insight into how these tests are conducted.

My hope is that these articles, and future installments based on forthcoming lectures, are helpful to physicians both in conducting their own research and in evaluating the research of others

Article PDF
Author and Disclosure Information

Aanchal Kapoor, MD
Department of Critical Care Medicine, Cleveland Clinic

Dr. Kapoor reported no financial interests or relationships that pose a potential conflict of interest with this article.

Publications
Page Number
e1
Legacy Keywords
biostatistics, epidemiology, Aanchal Kapoor
Author and Disclosure Information

Aanchal Kapoor, MD
Department of Critical Care Medicine, Cleveland Clinic

Dr. Kapoor reported no financial interests or relationships that pose a potential conflict of interest with this article.

Author and Disclosure Information

Aanchal Kapoor, MD
Department of Critical Care Medicine, Cleveland Clinic

Dr. Kapoor reported no financial interests or relationships that pose a potential conflict of interest with this article.

Article PDF
Article PDF
Related Articles

Physicians are inundated with clinical research findings that potentially impact patient care. Evaluating the strength and clinical application of research results requires an understanding of the underlying biostatistics and epidemiological principles.

The articles in this supplement are based on a series of lectures originally developed for fellows in pulmonary and critical care medicine to provide them with the tools to transform a scientific or clinical question into research projects, and then pursue the answer to their question with the appropriate methods. The same skills also enable them to appraise the published literature in a systematic and rigorous manner. 

Each topic in the series began with a presentation and discussion of statistical principles and methods, then moved to a practical module using the principles to appraise a specific publication. Participants in the course had an immediate opportunity to try the techniques, both to demonstrate understanding and to reinforce the concepts to each learner. The articles of this series follow the same outline, providing clinicians of all specialties the basic statistical tools to conduct and appraise clinical research, along with a sample article for practicing each statistical method presented.

This Cleveland Clinic Journal of Medicine supplement includes 3 lectures from the “Biostatistics and Epidemiology Lecture Series.” Dr. Stoller’s presentation, The Architecture of Clinical Research, describes the basic structure of clinical research and the nomenclature to understand trial design and sources of bias.

Building on those concepts, Dr. Chatburn’s lecture, Basics of Study Design: Practical Considerations, outlines the structured approach to develop a formal research protocol. How to identify a problem, expand the scope of it through a literature review, create a hypothesis, design a study, and an introduction to basic statistical methods are discussed.

And in Chi-square and Fisher’s Exact Tests, Dr. Nowacki introduces the statistical methodology of these 2 tests to assess associations between 2 independent categorical variables. The sample article illustrates step-by-step calculation of both the large sample approximation (chi-square) and exact (Fisher’s) methodologies providing insight into how these tests are conducted.

My hope is that these articles, and future installments based on forthcoming lectures, are helpful to physicians both in conducting their own research and in evaluating the research of others

Physicians are inundated with clinical research findings that potentially impact patient care. Evaluating the strength and clinical application of research results requires an understanding of the underlying biostatistics and epidemiological principles.

The articles in this supplement are based on a series of lectures originally developed for fellows in pulmonary and critical care medicine to provide them with the tools to transform a scientific or clinical question into research projects, and then pursue the answer to their question with the appropriate methods. The same skills also enable them to appraise the published literature in a systematic and rigorous manner. 

Each topic in the series began with a presentation and discussion of statistical principles and methods, then moved to a practical module using the principles to appraise a specific publication. Participants in the course had an immediate opportunity to try the techniques, both to demonstrate understanding and to reinforce the concepts to each learner. The articles of this series follow the same outline, providing clinicians of all specialties the basic statistical tools to conduct and appraise clinical research, along with a sample article for practicing each statistical method presented.

This Cleveland Clinic Journal of Medicine supplement includes 3 lectures from the “Biostatistics and Epidemiology Lecture Series.” Dr. Stoller’s presentation, The Architecture of Clinical Research, describes the basic structure of clinical research and the nomenclature to understand trial design and sources of bias.

Building on those concepts, Dr. Chatburn’s lecture, Basics of Study Design: Practical Considerations, outlines the structured approach to develop a formal research protocol. How to identify a problem, expand the scope of it through a literature review, create a hypothesis, design a study, and an introduction to basic statistical methods are discussed.

And in Chi-square and Fisher’s Exact Tests, Dr. Nowacki introduces the statistical methodology of these 2 tests to assess associations between 2 independent categorical variables. The sample article illustrates step-by-step calculation of both the large sample approximation (chi-square) and exact (Fisher’s) methodologies providing insight into how these tests are conducted.

My hope is that these articles, and future installments based on forthcoming lectures, are helpful to physicians both in conducting their own research and in evaluating the research of others

Page Number
e1
Page Number
e1
Publications
Publications
Article Type
Display Headline
Introduction: Biostatistics and epidemiology lecture series, part 1
Display Headline
Introduction: Biostatistics and epidemiology lecture series, part 1
Legacy Keywords
biostatistics, epidemiology, Aanchal Kapoor
Legacy Keywords
biostatistics, epidemiology, Aanchal Kapoor
Citation Override
Cleveland Clinic Journal of Medicine 2017 September;84(suppl 2):e1
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

The architecture of clinical research

Article Type
Changed
Mon, 10/01/2018 - 14:34
Display Headline
The architecture of clinical research
From the “Biostatistics and Epidemiology Lecture Series, Part 1”

I am flattered to present the inaugural talk in the biostatistics and clinical research design series on the architecture of clinical research. This content is based on the teachings of my mentor, Dr. Alvan Feinstein, who together with Dr. David Sackett, is credited with pioneering clinical epidemiology. Dr. Feinstein was a Sterling Professor at the Yale School of Medicine. His main opus of work is a book called, Clinical Epidemiology: The Architecture of Clinical Research.1 This paper is named in credit to Dr. Feinstein’s enormous contribution. I will review some important terms defined by Dr. Feinstein to provide the background necessary for the remainder of the talks in this series.

To start, I will frame this topic by asking the following question: Why do we do research? I’ll talk about the basic structure of research studies and provide a taxonomy, as Dr. Feinstein would say, a nomenclature with which to understand trial design and the sources of bias in those trials. Then, I will discuss these sources of bias in detail using the taxonomy that Dr. Feinstein described in his aforementioned book. Finally, I will share with you some examples of bias in clinical trials to help you better understand these concepts.

Now, the answer to the basic question posed above is: basically, we do cause-and-effect research to establish the causality of a risk factor or the efficacy of a therapy. Does cigarette smoking cause lung cancer? Does taking hydrochlorothiazide help systemic hypertension? Does air pollution worsen asthma? Does supplemental oxygen help patients with chronic obstructive pulmonary disease (COPD)?

Cause-and-effect research can be subsumed under 2 broad issues: causal risk factors and therapeutic efficacy. In his review of early false understandings in medicine that were based on anecdotal observation alone, Thomas cites many examples—“the undue longevity of useless and even harmful drugs can be laid at the door of authority,” ie, empiricism, lack of rigorous research.2 The field is full of these: yellow fever causality, the value of cupping, and even intermittent mandatory ventilation when it was described by John Downs in 1973 and touted as a superior mode for weaning patients from mechanical ventilation.3 Twenty-five years later, randomized controlled trials by Brochard et al4 indicated not only that intermittent mandatory ventilation was not the best mode to wean but was, in fact, the worst mode for weaning patients from mechanical ventilation compared with either pressure support or spontaneous breathing trials. Many more examples exist to demonstrate the false understandings that can be ascribed to lack of rigorous study or evidence in medicine.

Design of a controlled trial according to Feinstein.
Figure 1. Design of a controlled trial according to Feinstein.1

Before systematically exploring the sources of bias in Feinstein’s construct, let us define some very basic terms from his book. Dr. Feinstein talks about the baseline state, which refers to the group of patients under study who are culled from a larger population to whom the results are intended to be applied (Figure 1).1 This baseline group is hopefully representative of this larger target population. As a nod to the later discussion, Dr. Feinstein would call bias introduced by unusual assembly of the study population from the larger intended population as “assembly bias.” So, if the group under study is not representative of either the patients you see or the world of patients with this condition or if there is something special or distinctively nonrepresentative about the study population, then the results may be subject to “assembly bias.” Assembly bias can compromise the so-called “external” validity of the study—its ability to be applied to populations beyond the study group.

Having assembled a baseline group for study, that group is classically allocated to 1 of 2 (or sometimes more than 2) compared therapies. In a controlled trial, patients can be allocated using a variety of strategies, including randomization. Using the paradigm diagram (Figure 1, which considers a 2-arm trial), patients are allocated to 1 of 2 compared groups—group A and group B. Then, in a treatment trial, 1 group receives the principal maneuver, which is the drug or intervention under study—for example, supplemental oxygen for patients with COPD. The comparative maneuver is allocated to group B, which also receives all the other treatments (called “co-maneuvers”) that are used to treat the condition under study. In a trial of supplemental oxygen for COPD evaluating lung function and exacerbation frequency as outcome measures, such co-maneuvers might include inhaled bronchodilators, inhaled corticosteroids, pulmonary rehabilitation, and Pneumovax vaccine. Ideally, these co-maneuvers are equally distributed between the compared groups (A and B).

So, in summary, we have a comparative maneuver, which is the nonadministration of supplemental oxygen in this proposed trial of supplemental oxygen in COPD, the principal maneuver—administration of oxygen—and all the co-maneuvers that are ideally equally distributed between both groups. This balanced distribution of co-maneuvers between the compared groups helps to ensure that any differences in the study outcome measures (ie, what is counted as the main impact of the intervention under study) can be solely attributed to the principal maneuver. When this condition—that the difference in outcomes can be reliably ascribed to the study intervention—is satisfied, the study is felt to be “internally” valid. As we will see, ensuring internal validity requires freedom from the many sources of what Dr. Feinstein calls “internal bias.”

Back to basic terms: “cohort” in Dr. Feinstein’s language is a group that shares common traits and is followed forward in a longitudinal study. The “outcome measure” is self-evident—it is what is being measured, with the “primary outcome” being the pre-defined measure that is considered the most important (and ideally most clinically relevant) impact of the study intervention. Later in this series of lectures, there will be discussions of power calculations and the so-called “effect size”—the magnitude of effect that the intervention is expected to produce and that is ideally deemed clinically important.

 

 

An important consideration in designing a trial is to define and declare the primary outcome measure carefully because defining the primary outcome measure has important implications for the study. I will provide an example from the alpha-1 antitrypsin deficiency literature. Some of you have probably read what has been called the RAPID trial.5 RAPID was a trial of augmentation therapy vs placebo in patients with severe alpha-1 antitrypsin deficiency. The primary outcome measure (which was pre-negotiated with the US Food and Drug Administration [FDA]) was computer tomography (CT) lung density determined at functional residual capacity (FRC) and total lung capacity (TLC). The trial failed to achieve statistical significance in regard to CT lung density, although the study authors argued that CT density measurements made at TLC were more reproducible than those made at FRC. When the results were analyzed by TLC alone, the results were statistically significant, but when they were analyzed with FRC and TLC combined, they were not. In the end, based on the pre-negotiated primary outcome measure of CT density based on both FRC and TLC, the FDA rejected the proposal for a label change to say that augmentation therapy slowed the loss of lung density even though the weight of evidence was clearly in its favor. This case exemplifies just how critical the choice of primary outcome measure can be.

Design of a randomized crossover trial of terbutaline for diaphragmatic function.
Figure 2. Design of a randomized crossover trial of terbutaline for diaphragmatic function. The wash-out period separates the first and the second interventions (begins at the star in the diagram).

The wash-out period refers to an interval in a subset of randomized trials called “crossover trials” in which the primary intervention is discontinued and the patient returns to his baseline state before the comparative maneuver is then implemented (Figure 2).6 In order to perform a crossover trial, it is important that the effects of the initial intervention can “wash out” or be fully extinguished. So, for example, in trials of radiation therapy vs surgery, it is impossible to do a crossover trial because the effects of radiation can never completely wash out nor can those of surgery, which are similarly permanent. For example, we cannot replace the colon once it is resected for cancer or replace the appendix once removed. Therefore, producing a wash-out requires some very specific pharmacokinetic and pharmacodynamic features in order for a crossover trial to be considered. Later talks in this series will discuss the enhanced statistical power of a crossover trial, where one is comparing every patient to him or herself rather than to another patient.

So, there is always an appetite to do a crossover trial as long as the criteria for wash-out can be met, namely again that the primary intervention can dissipate completely to the baseline state before the alternative intervention is implemented.

“Placebo” is a fairly self-evident and well-understood term; placebo refers to the administration of a maneuver in a way that is identical to the principal maneuver except that the placebo is not expected to exert any clinical effect.

“Blinding” is the unawareness of either the investigator or of the patient to which the intervention is being administered. “Single-blinding” refers to the condition in which either the study or the investigator (but not both) is unaware, and “double-blinding” refers to the condition in which both the subjects and the investigators are unaware. There can be some subtle issues that compromise whether the patient is aware of the intervention that he or she is receiving and that can potentially condition the patient’s response, particularly if there is any subjective component of the assessment of the outcome. So, blinding is important.

With these terms describing the elements of a clinical study now described, let us turn to the types of studies that comprise clinical research. The first group of study types is what Dr. Feinstein called descriptive studies—studies that simply describe phenomena without comparison to a control group. As an example of a descriptive study, Sehgal et al7 recently described the workup of a focal, segmental pneumonia in a patient taking pembrolizumab for lung cancer. In this paper, there were four other cases of focal pneumonia accompanying pembrolizumab use that were assembled from the literature, making this descriptive paper a so-called case series. A “case series” differs from a “single case report,” which reports a single patient experience. Though limited in their ability to establish cause and effect, case reports and case series can help researchers develop proof of principle, so I would not discount the value of case reports.8

I can cite a case report from of my own experience that demonstrates this point. In 1987, I saw a patient from Buffalo who had primary biliary cirrhosis and the hepatopulmonary syndrome (HPS). She was so debilitated by her HPS that she could not stand up without desaturating severely. Although she had normal liver synthetic function, she was severely debilitated by her HPS and the decision was made to offer her a liver transplant, which, at that time, was considered to be relatively contraindicated. Much to everyone’s amazement and satisfaction, her HPS completely resolved after the transplant surgery. Her oxygenation and alveolar-arterial oxygen gradient normalized, and her clubbing resolved. We reported this in a case report, which began to affect the way people thought about the feasibility of liver transplant for the HPS.8 The lesson is: do not underestimate the power of a thoughtful case report.

The second group of research study types is called “cohort studies,” in which one actually compares outcomes between 2 groups in the study. Cohort studies fall into the bucket of either “observational cohort studies,” in which allocation to the compared maneuvers is not performed by randomization but by any other strategy, and “randomized trials.” In observational studies, allocation could occur through physician choice, as when the physician prescribes a treatment to 1 group but not another, or by patient choice or circumstance. For example, an observational cohort study of the risk of cigarette smoking would compare outcomes between smokers and non-smokers where the patient choses to smoke under his/her own volition. Alternatively, the circumstances of an exposure could allocate someone to the principal maneuver, as when we are studying the effect of exposure to World Trade Center dust in the firefighters who responded or of exposure to nuclear radiation in Hiroshima survivors. These are examples of observational cohort studies that compare exposed individuals to unexposed individuals, where the exposure did not occur by randomization but by choice or unfortunate circumstance.

In contrast to observational studies, allocation in randomized trials occurs through a formal process. Randomization has the specific purpose of attempting to ensure that patients are allocated to 2 comparative groups from the baseline group with comparable risk for developing the outcome measure. When randomization is effective, differences in study outcomes can be reliably ascribed to the intervention rather than to differences in the baseline susceptibility of the compared groups.

 

 

While randomization is an excellent strategy to ensure baseline similarity between compared groups, randomization can fail, and its effectiveness must be checked. Specifically, in a randomized trial, it is customary to examine the compared groups at baseline on all features that can affect the likelihood of developing the outcome measure. If the groups turn out to be dissimilar at baseline in an important way, then the study is at risk for bias, which is specifically called “susceptibility bias” in Feinstein’s construct. Obviously, the larger number of baseline clinical and demographic features that can condition the likelihood of developing the outcome measure, the more difficult it is to achieve baseline similarity between compared groups and the more important it becomes to ensure that randomization has been effective. In this circumstance, larger numbers of participants in both compared groups are generally needed. More about susceptibility bias later.

There are generally 2 types of randomized trials: the so-called “parallel controlled trials” in which each group receives either the principal or the comparative maneuver and is followed and “crossover trials” in which each compared group receives both the principal maneuver and the co-maneuver at different times after an effective wash-out period. Wash-out was discussed above. Figure 2 shows an example of a crossover trial examining the effects of terbutaline on diaphragmatic function.6 The investigators administered terbutaline for a week, measured transdiaphragmatic pressures, gave the patient a terbutaline vacation (the “wash-out period”), and then crossed over those patients who were initially receiving terbutaline to placebo and initial placebo recipients to terbutaline, having remeasured diaphragmatic function after the wash-out period to assure that the patient’s diaphragmatic function prior to the second crossover was identical to his/her baseline state. If this return to baseline is accomplished, then the criteria from effective wash-out are satisfied.

Types of bias in a clinical trial according to Feinstein
Now, with these basic structural terms of clinical research defined, bias will occupy the remainder of the discussion. By definition, bias in a clinical trial is any factor in the design or conduct of the trial, either external to the trial or internal to the trial, that can alter the results in a way that either threatens the reliability of attributing the differences in outcomes between the compared groups with the principal maneuver (“internal validity”) or limits the ability of the results, however internally valid, to be applied to a specific population beyond the study group (“external validity”) (Table 1).1 This again is because the main goal of cause-and-effect research is to make sure that you can attribute differences between the 2 compared groups at the end of the trial to the intervention under study and nothing else.

A comparison of surgery vs nonsurgical therapy for advanced lung cancer.
Figure 3. A comparison of surgery vs nonsurgical therapy for advanced lung cancer. An example of possible susceptibility bias.1

As we begin to talk about sources of bias, consider a study in which we compare survival of patients allocated to surgery vs nonsurgical therapy for lung cancer (Figure 3).1 This study is subject to the first type of so-called “internal bias” in the Feinsteinian construct—so-called “selection bias.” For example, all patients treated surgically were considered healthy enough by their doctors to undergo surgery, whereas patients treated without surgery may have been deemed inoperable because of comorbidities, lung dysfunction, cardiac dysfunction, and so on. If the results of such a comparison show that the mortality rate among surgical patients in this study was lower, the question then becomes: is the improved survival in surgical candidates due to the superior efficacy of surgery vs other therapy or was the enhanced survival due to the surgical patients being healthier to begin with? You can intuitively sense that the answer to this question is that the enhanced survival may be due to the better health of patients treated surgically rather than to the surgery itself because of how the patients were selected to receive it. So, this is a simple example of what Dr. Feinstein would call “susceptibility bias.” Susceptibility bias occurs when the 2 baseline groups are not comparably at risk or susceptible to developing the outcome measure, leading the naïve investigator in this specific example to attribute the difference in outcomes to the superiority of surgery when in fact it may have nothing to do with the surgery vs. the other maneuver. When susceptibility bias is in play, the difference between the outcomes in the compared groups could be attributed to the baseline imbalance of the groups rather than to the principal maneuver itself.

Turning back to the taxonomy of bias, there are four types that can threaten internal validity—“susceptibility,” “performance,” “detection,” and “transfer” bias—and 1 type of bias (called “external bias”) that can affect the generalizability of the study called “assembly bias” (Table 1).

Potential sources of bias in a randomized, controlled trial according to Feinstein.
Figure 4. Potential sources of bias in a randomized, controlled trial according to Feinstein.1

Figure 4 shows where these various sources of bias appear in the architecture of a clinical trial. As just discussed, susceptibility bias affects the baseline state and the comparability of the groups. Performance bias relates to how effective and how comparably the co-maneuvers are given and whether the primary intervention is potent enough to affect an outcome. Both transfer and detection bias operate in detecting the outcome, especially regarding the rigor and frequency with which they are investigated. Transfer bias has to do with selective loss to follow-up of those included in the trial. If there is a systematic reason for loss to follow-up that is related to the impact of the intervention, then the study is at risk for transfer bias. For example, in a randomized trial of drug A vs placebo for pneumonia, if drug A is effective but all the drug A recipients fail to follow-up because they feel too good to return for follow-up, then transfer bias could be causing the study to show nonefficacy even though the drug works. So, if those who respond favorably are systematically lost to follow-up, and if all the patients who felt lousy wanted to see the doctor and came back for follow-up, such transfer bias would bias towards nonefficacy. Specifically, only patients remaining in the trial would be those who failed to respond and that would dilute any difference between the 2 groups despite the active efficacy of drug A.

Hopefully, you are already beginning to get a sense that one has to be extremely disciplined in thinking about each of these sources of bias because they can have some very subtle nuances in randomized trials that can easily escape attention.

Returning to sources of bias, let’s consider the second type of bias, “performance bias.” Performance bias relates to the administration of the compared maneuvers—the primary or principal maneuver, compared with the comparative maneuver. Performance bias can occur when the main maneuver is not administered adequately or when the co-maneuvers are administered in an imbalanced way between the compared groups. Consider the example of the Long-Term Oxygen Treatment Trial (LOTT) trial, which compared use of supplemental oxygen with no supplemental oxygen in patients with stable COPD and resting or exercise-induced moderate desaturation.9 The principal outcome measure of LOTT was all-cause hospitalization or death. In such a study, many potential sources of performance bias exist. For example, performance bias might exist if none of the patients allocated to oxygen actually used supplemental oxygen. Alternately, to the extent that use of inhaled corticosteroids or antimuscarinic agents lessens the risk of COPD exacerbation, performance bias could occur if use of these co-maneuvers was imbalanced between the compared groups. As a specific extreme circumstance, if all patients in the nonoxygen group used these inhalers but none of the patients in the oxygen group did, then a lack of difference between exacerbation frequency could be related to this imbalance in co-maneuvers (a form of performance bias) rather than to the lack of efficacy of supplemental oxygen.

 

 

“Compliance bias” is a subset of performance bias which occurs when 2 conditions are satisfied: (1) the main maneuver is not administered adequately, and (2) the investigator is unaware of that nonreceipt so that this cannot be accounted for in interpreting the study results. For example, if a drug has efficacy but if no one in the treatment arm of the trial takes the drug, the absence of a difference in outcomes between the compared groups will be ascribed to nonefficacy, whereas “compliance bias” (ie, no one actually took the drug) could actually be the cause. Ideally, randomized studies should be evaluated on an “intention to treat” basis irrespective of compliance, but there is an analytic approach called “per protocol” analysis in which you can analyze the results according to whether the patient actually used the intervention in an effective way. “Per protocol” analysis is a secondary analysis of the primary results but it can nonetheless help determine whether the negative result is likely related to noncompliance or not.

A third type of internal bias, “detection bias,” is fairly straightforward. Detection bias is related to how avidly and how comparably the outcomes are measured between the 2 compared groups. Let’s say that you are conducting a trial of a new antibiotic and the primary outcome is colony counts on petri dishes of plated collected specimens. If the technicians who read the petri dish counts are unblinded, they may look at the colony counts with a biased eye, seeing fewer colonies on plates collected from patients receiving the antibiotic.

Overall, detection bias occurs when outcomes are ascertained or detected unequally between the compared groups, and detection bias can involve any of the following: is there comparable surveillance of the 2 groups for analysis of the outcome measure? Are the diagnostic tests comparably performed in both groups and is the interpretation comparably unbiased with equipoise? Investigators who know which patients are receiving an active drug and those who are not could experience subliminal bias that renders them more likely to find that the drug under study is efficacious.

Depending on the principal study maneuver, ensuring blinding can be challenging. To demonstrate this point, let’s consider the example of conducting a randomized control trial of Vicks VapoRub. Vicks VapoRub is an old product that smells like wintergreen and that mothers used to rub on the chests of their infants in the hope of speeding recovery from colds and bronchitis episodes. It was felt that the distinctive smell of the product was materially related to wintergreen, which gives rise to the odor. So, imagine a randomized trial of Vicks VaporRub. A trial is designed in which sick children receive Vicks VapoRub on their chest and others receive a placebo rub that lacks the distinctive wintergreen odor. But, the odor itself is felt to be related to how Vicks VapoRub actually works. Thus, it is the odor itself that creates the blinding challenge here.

The primary outcomes in this study are the duration of the child’s cold symptoms, as ascertained by pediatricians actually examining the children. So, pediatricians would come and listen to the infants’ chests: “Yeah, this chest is clear, but this other infant is still full of rhonchi,” and they would ascertain the outcome measure in this way. So, my blinding question to you is: how do you blind a trial of Vicks VapoRub given the conditions described? Namely, you put the VapoRub on the chest, it smells and the smell is the intervention—how do you blind such a trial? 

The clever answer is that you should put Vicks VapoRub on the upper lips of all the examiners, so what they smell is Vicks VapoRub independent of whether the child they are examining also has the Vicks VapoRub or placebo on their chest. In this way, single blinding of the examiners is preserved and detection bias is averted. It is important to point out that double blinding could also be achieved by placing Vicks VapoRub on the child’s upper lip, but there is little reason to suspect that the infants being studied have a bias related to whether they smell the Vicks VapoRub.

The fourth potential source of internal bias is called “transfer bias.” Transfer bias is the selective loss to follow-up of patients from 1 of the 2 compared groups in the trial for a systematic reason. By systematic, I mean that that the drop-out is associated with the development of the outcome event or some impact of the intervention regarding the likelihood to develop the outcome event. As an example, if all patients respond favorably to a drug and everybody fails to follow up because they feel too good to come back, then that would bias the study towards nonefficacy even in the face of an efficacious intervention.

Finally, let’s consider a source of bias that can affect the “external validity,” or the generalizability of the study results to populations other than that included in the study itself. Dr. Feinstein calls this 5th type of bias “assembly bias” (Table 1).1 Assembly bias occurs when the results of the study cannot be reliably applied to populations outside the study itself.

For example, if I screen patients during a study of digoxin for heart rate control in atrial fibrillation, I could establish whether the subject was compliant or not by checking his/her serum digoxin levels. Serum levels of 0 indicate that the patient has not taken the digoxin. If I include a run-in period for the trial—an interval before the actual study when I am assessing potential subjects’ eligibility to participate—and check serum digoxin levels to include only patients who are shown to be taking the drug, then I am screening for study inclusion on compliance. In this way, I will have assembled a population that is highly compliant so that I can truly assess whether digoxin has efficacy in controlling the heart rate in patients with atrial fibrillation. At the same time, this study population is not highly representative of the population of patients with atrial fibrillation at large, because we know that rates of drug noncompliances may be as high as 30% to 40%. So, culling a population with run-in periods on demonstrated compliance criteria may be very important to assess efficacy (ie, whether the drug works), but this design will trade off on the effectiveness of the drug (ie, which asks the question “does the drug work in actual practice?”). This is because, in the yin-yang between assessing efficacy and assessing effectiveness, the focus on assessing efficacy naturally undermines the ability to assess whether the drug works in real-world conditions.

As another example of potential assembly bias, let’s say you are studying an antihypertensive drug at a Veterans Administration (VA) hospital, where most veterans are men. But you are treating women in your practice and wonder whether the drug, which works in a predominately male population, will work in your female patients. So, there could be assembly bias in applying the results of a VA study to a non-VA predominantly female population.

Having now described the design of clinical trials and the major sources of bias, let’s apply this thinking to the earliest clinical trial. James Lind, a British Naval officer, was credited with conducting the first clinical trial of citrus fruits for scurvy while sailing on the ship Salisbury in 1747.2 The question that Lind addressed was “does citrus fruit treat and prevent scurvy?” In describing this trial, Lind stated “I took 12 patients with scurvy, these patients were as similar as I could have them, had one diet common to all.” As you read this through your new Feinsteinian bias lens, Lind is addressing 2 potential sources of bias, namely, susceptibility bias and performance bias. In trying to make the “cases as similar as I could have them,” he is trying to avoid susceptibility bias and in “providing one diet common to all,” he is trying to avoid performance bias.

In terms of the intervention in this trial, these 12 patients were allocated in pairs to several interventions: a quart of cider a day, 25 drops of elixir of vitriol 3 times a day on an empty stomach, 2 spoonsful of vinegar 3 times a day on an empty stomach, ½ pint a day of sea water, 2 oranges and 1 lemon given every day, and a “bigness of nutmeg” 3 times per day. In describing the outcome of the trial, Lind states “the consequence was that the most sudden and visible good effects were perceived from the use of oranges and lemons; one of those who had taken them, being at the end of 6 days fit for duty. The spots were not indeed at that time quite off his body, nor his gums sound, but without any other medicine then a gargarism of elixir vitriol, he became quite healthy before we came into Plymouth which was on the 16th of June. The other was the best recovered of any in his condition; and being now deemed pretty well, was appointed nurse to the rest of the sick.”

 

 

In analyzing this trial, we could characterize it as a parallel controlled trial. Whether the allocation was done by randomization is not clear, but it was certainly an observational cohort study in that there were concurrent controls who were treated as similarly as possible except for the principal maneuver, which was the administration of citrus fruit. Already mentioned was the attention to averting susceptibility and performance bias. There was no evidence of compliance bias as the interventions were enforced, nor was there evidence of transfer bias because all subjects who were enrolled in the study completed the study because they were a captive group on a sailing ship. Finally, the likelihood of assembly bias seems small, as these sailors seemed to be representative of victims of scurvy in general, namely in being otherwise deprived of access to citrus fruits.

In terms of the statistical results of this study, subsequent analysis of the research showed that the impact of lemons and oranges was dramatic and showed a trend (P = .09) towards statistical significance. Notwithstanding the lack of a P < .05, Dr. Feinstein would likely say that this study satisfied the “intra-ocular test” in that the efficacy of the citrus fruit was so dramatic that it “hit you between the eyes.” He often argued that the widespread practice of prescribing penicillin for pneumococcal pneumonia was not based on the results of a convincing randomized controlled trial because the efficacy of penicillin in that setting was so dramatic that a randomized trial was not necessary (and potentially even unethical if the condition of “intra-ocular” efficacy was satisfied).

The final question to address in this lecture is whether randomized controlled trials, for all their rigor, always produce more reliable results than observational studies. This issue has been addressed by several authors.10–12 Sacks et al10 contended in 1983 that observational studies systematically overestimate the magnitude of association between exposure and outcome and therefore argued that randomized trials were more reliable than observational studies. Subsequent analyses tended to challenge this view.11,12 Specifically, Benson and Hartz11 compared the results of 136 reports regarding 19 different therapies that were studied between 1985 and 1998. In only 2 of the 19 analyses did the treatment effects in the observational studies fall outside the 95% confidence interval for the randomized controlled trial results. In this way, these authors argued that observational studies generally are concordant with the results of randomized trials. They stated that “our finding that observational studies and randomized controlled trials usually produce similar results differs from the conclusions of previous authors. The fundamental criticism of observational studies is that unrecognized confounding factors may distort the results. According to the conventional wisdom, this distortion is sufficiently common and unpredictable that observational studies are not liable and should not be funded. Our results suggested observational studies usually do provide valid information.”11

An additional analysis of this issue was performed by Concato et al,12 who identified 99 articles regarding 5 clinical topics. Again, the results from randomized trials were compared with those of observational cohort or case-controlled studies regarding the same intervention. The authors reported that “contrary to prevailing belief, the average results from well-designed observational studies did not systematically overestimate the magnitude of the associations between exposure and outcome as compared with the results of randomized, controlled trials on the same topic. Rather, the summary results of randomized, controlled trials and observational studies were remarkably similar.”12

On the basis of these studies, it appears that randomized control trials continue to serve as the gold standard in clinical research, but we must also recognize that circumstances often preclude the conduct of a randomized trial. As an example, consider a randomized trial of whether cigarette smoking is harmful, which, given the strong suspicion of harm, would be unethical in that patients cannot be randomized to smoke. Similarly, from the example before, a randomized trial of penicillin for pneumococcal pneumonia would be unethical because denying patients in the placebo group access to penicillin would exclude them from access to a drug that has “intra-ocular” efficacy. In circumstances like these, well-performed observational studies that are attentive to sources of bias can likely produce comparably reliable results to randomized trials.

In the end, of course, the interpretation of the study results requires the reader’s careful attention to potential sources of bias that can compromise study validity. The hope is that with Dr. Feinstein’s framework, you can be better equipped to think critically about study results that you review and to keenly ascertain whether there is any threat to internal or to external validity. Similarly, as you go on to design clinical trials yourselves, you can pay attention to these potential sources of bias that, if present, can compromise the reliability of the study conclusions internally or their applicability to patients outside of the study.

References
  1. Feinstein AR. Clinical Epidemiology: The Architecture of Clinical Research. Philadelphia, PA: WB Saunders; 1985.
  2. Thomas DP. Experiment versus authority: James Lind and Benjamin Rush. N Engl J Med 1969; 281:932–934.
  3. Downs JB, Klein EF Jr, Desautels D, Modell JH, Kirby RR. Intermittent mandatory ventilation: a new approach to weaning patients from mechanical ventilators. Chest 1973; 64:331–335.
  4. Brochard L, Rauss A, Benito S, et al. Comparison of three methods of gradual withdrawal from ventilatory support during weaning from mechanical ventilation. Am J Respir Crit Care Med 1994; 150:896–903.
  5. Chapman KR, Burdon JGW, Piitulainen E, et al; on behalf of the RAPID Trial Study Group. Intravenous augmentation treatment and lung density in severe 1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 386:360–368.
  6. Stoller JK, Wiedemann HP, Loke J, Snyder P, Virgulto J, Matthay RA. Terbutaline and diaphragm function in chronic obstructive pulmonary disease: a double-blind randomized clinical trial. Br J Dis Chest 1988; 82:242–250.
  7. Sehgal S, Velcheti V, Mukhopadhyay S, Stoller JK. Focal lung infiltrate complicating PD-1 inhibitor use: a new pattern of drug-associated lung toxicity? Respir Med Case Rep 2016; 19:118–120.
  8. Stoller JK, Moodie D, Schiavone WA, et al. Reduction of intrapulmonary shunt and resolution of digital clubbing associated with primary biliary cirrhosis after liver transplantation. Hepatology 1990; 11:54–58.
  9. Albert RK, Au DH, Blackford AL, et al; for the Long-Term Oxygen Treatment Trial Group. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med 2016; 375:1617–1627.
  10. Sacks HS, Chalmers TC, Smith H Jr. Sensitivity and specificity of clinical trials: randomized v historical controls. Arch Intern Med 1983; 143:753–755.
  11. Benson K, Hartz AJ. A comparison of observational studies and randomized, controlled trials. N Engl J Med 2000; 342:1878–1886.
  12. Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med 2000; 342:1887–1892.
Article PDF
Author and Disclosure Information

James K. Stoller, MD, MS
Chairman, Education Institute; Head, Cleveland Clinic Respiratory Therapy, Department of Pulmonary Medicine; and the Department of Critical Care Medicine, Cleveland Clinic, Cleveland, OH

Correspondence: James K. Stoller, MD, MS, Education Institute, NA22, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Dr. Stoller’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Dr. Stoller presented his lecture on August 2, 2016, at Cleveland Clinic.

Dr. Stoller reported research grant support from CSL Behring and consulting for Grifols, Shire, CSL Behring, and Arrowhead Pharmaceuticals.

Publications
Page Number
e2-e9
Legacy Keywords
Alvan Feinstein, David Sackett, trial design, clinical research, bias, outcome measure, wash-out period, James Lind, scurvy, James Stoller
Author and Disclosure Information

James K. Stoller, MD, MS
Chairman, Education Institute; Head, Cleveland Clinic Respiratory Therapy, Department of Pulmonary Medicine; and the Department of Critical Care Medicine, Cleveland Clinic, Cleveland, OH

Correspondence: James K. Stoller, MD, MS, Education Institute, NA22, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Dr. Stoller’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Dr. Stoller presented his lecture on August 2, 2016, at Cleveland Clinic.

Dr. Stoller reported research grant support from CSL Behring and consulting for Grifols, Shire, CSL Behring, and Arrowhead Pharmaceuticals.

Author and Disclosure Information

James K. Stoller, MD, MS
Chairman, Education Institute; Head, Cleveland Clinic Respiratory Therapy, Department of Pulmonary Medicine; and the Department of Critical Care Medicine, Cleveland Clinic, Cleveland, OH

Correspondence: James K. Stoller, MD, MS, Education Institute, NA22, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Dr. Stoller’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Dr. Stoller presented his lecture on August 2, 2016, at Cleveland Clinic.

Dr. Stoller reported research grant support from CSL Behring and consulting for Grifols, Shire, CSL Behring, and Arrowhead Pharmaceuticals.

Article PDF
Article PDF
Related Articles
From the “Biostatistics and Epidemiology Lecture Series, Part 1”
From the “Biostatistics and Epidemiology Lecture Series, Part 1”

I am flattered to present the inaugural talk in the biostatistics and clinical research design series on the architecture of clinical research. This content is based on the teachings of my mentor, Dr. Alvan Feinstein, who together with Dr. David Sackett, is credited with pioneering clinical epidemiology. Dr. Feinstein was a Sterling Professor at the Yale School of Medicine. His main opus of work is a book called, Clinical Epidemiology: The Architecture of Clinical Research.1 This paper is named in credit to Dr. Feinstein’s enormous contribution. I will review some important terms defined by Dr. Feinstein to provide the background necessary for the remainder of the talks in this series.

To start, I will frame this topic by asking the following question: Why do we do research? I’ll talk about the basic structure of research studies and provide a taxonomy, as Dr. Feinstein would say, a nomenclature with which to understand trial design and the sources of bias in those trials. Then, I will discuss these sources of bias in detail using the taxonomy that Dr. Feinstein described in his aforementioned book. Finally, I will share with you some examples of bias in clinical trials to help you better understand these concepts.

Now, the answer to the basic question posed above is: basically, we do cause-and-effect research to establish the causality of a risk factor or the efficacy of a therapy. Does cigarette smoking cause lung cancer? Does taking hydrochlorothiazide help systemic hypertension? Does air pollution worsen asthma? Does supplemental oxygen help patients with chronic obstructive pulmonary disease (COPD)?

Cause-and-effect research can be subsumed under 2 broad issues: causal risk factors and therapeutic efficacy. In his review of early false understandings in medicine that were based on anecdotal observation alone, Thomas cites many examples—“the undue longevity of useless and even harmful drugs can be laid at the door of authority,” ie, empiricism, lack of rigorous research.2 The field is full of these: yellow fever causality, the value of cupping, and even intermittent mandatory ventilation when it was described by John Downs in 1973 and touted as a superior mode for weaning patients from mechanical ventilation.3 Twenty-five years later, randomized controlled trials by Brochard et al4 indicated not only that intermittent mandatory ventilation was not the best mode to wean but was, in fact, the worst mode for weaning patients from mechanical ventilation compared with either pressure support or spontaneous breathing trials. Many more examples exist to demonstrate the false understandings that can be ascribed to lack of rigorous study or evidence in medicine.

Design of a controlled trial according to Feinstein.
Figure 1. Design of a controlled trial according to Feinstein.1

Before systematically exploring the sources of bias in Feinstein’s construct, let us define some very basic terms from his book. Dr. Feinstein talks about the baseline state, which refers to the group of patients under study who are culled from a larger population to whom the results are intended to be applied (Figure 1).1 This baseline group is hopefully representative of this larger target population. As a nod to the later discussion, Dr. Feinstein would call bias introduced by unusual assembly of the study population from the larger intended population as “assembly bias.” So, if the group under study is not representative of either the patients you see or the world of patients with this condition or if there is something special or distinctively nonrepresentative about the study population, then the results may be subject to “assembly bias.” Assembly bias can compromise the so-called “external” validity of the study—its ability to be applied to populations beyond the study group.

Having assembled a baseline group for study, that group is classically allocated to 1 of 2 (or sometimes more than 2) compared therapies. In a controlled trial, patients can be allocated using a variety of strategies, including randomization. Using the paradigm diagram (Figure 1, which considers a 2-arm trial), patients are allocated to 1 of 2 compared groups—group A and group B. Then, in a treatment trial, 1 group receives the principal maneuver, which is the drug or intervention under study—for example, supplemental oxygen for patients with COPD. The comparative maneuver is allocated to group B, which also receives all the other treatments (called “co-maneuvers”) that are used to treat the condition under study. In a trial of supplemental oxygen for COPD evaluating lung function and exacerbation frequency as outcome measures, such co-maneuvers might include inhaled bronchodilators, inhaled corticosteroids, pulmonary rehabilitation, and Pneumovax vaccine. Ideally, these co-maneuvers are equally distributed between the compared groups (A and B).

So, in summary, we have a comparative maneuver, which is the nonadministration of supplemental oxygen in this proposed trial of supplemental oxygen in COPD, the principal maneuver—administration of oxygen—and all the co-maneuvers that are ideally equally distributed between both groups. This balanced distribution of co-maneuvers between the compared groups helps to ensure that any differences in the study outcome measures (ie, what is counted as the main impact of the intervention under study) can be solely attributed to the principal maneuver. When this condition—that the difference in outcomes can be reliably ascribed to the study intervention—is satisfied, the study is felt to be “internally” valid. As we will see, ensuring internal validity requires freedom from the many sources of what Dr. Feinstein calls “internal bias.”

Back to basic terms: “cohort” in Dr. Feinstein’s language is a group that shares common traits and is followed forward in a longitudinal study. The “outcome measure” is self-evident—it is what is being measured, with the “primary outcome” being the pre-defined measure that is considered the most important (and ideally most clinically relevant) impact of the study intervention. Later in this series of lectures, there will be discussions of power calculations and the so-called “effect size”—the magnitude of effect that the intervention is expected to produce and that is ideally deemed clinically important.

 

 

An important consideration in designing a trial is to define and declare the primary outcome measure carefully because defining the primary outcome measure has important implications for the study. I will provide an example from the alpha-1 antitrypsin deficiency literature. Some of you have probably read what has been called the RAPID trial.5 RAPID was a trial of augmentation therapy vs placebo in patients with severe alpha-1 antitrypsin deficiency. The primary outcome measure (which was pre-negotiated with the US Food and Drug Administration [FDA]) was computer tomography (CT) lung density determined at functional residual capacity (FRC) and total lung capacity (TLC). The trial failed to achieve statistical significance in regard to CT lung density, although the study authors argued that CT density measurements made at TLC were more reproducible than those made at FRC. When the results were analyzed by TLC alone, the results were statistically significant, but when they were analyzed with FRC and TLC combined, they were not. In the end, based on the pre-negotiated primary outcome measure of CT density based on both FRC and TLC, the FDA rejected the proposal for a label change to say that augmentation therapy slowed the loss of lung density even though the weight of evidence was clearly in its favor. This case exemplifies just how critical the choice of primary outcome measure can be.

Design of a randomized crossover trial of terbutaline for diaphragmatic function.
Figure 2. Design of a randomized crossover trial of terbutaline for diaphragmatic function. The wash-out period separates the first and the second interventions (begins at the star in the diagram).

The wash-out period refers to an interval in a subset of randomized trials called “crossover trials” in which the primary intervention is discontinued and the patient returns to his baseline state before the comparative maneuver is then implemented (Figure 2).6 In order to perform a crossover trial, it is important that the effects of the initial intervention can “wash out” or be fully extinguished. So, for example, in trials of radiation therapy vs surgery, it is impossible to do a crossover trial because the effects of radiation can never completely wash out nor can those of surgery, which are similarly permanent. For example, we cannot replace the colon once it is resected for cancer or replace the appendix once removed. Therefore, producing a wash-out requires some very specific pharmacokinetic and pharmacodynamic features in order for a crossover trial to be considered. Later talks in this series will discuss the enhanced statistical power of a crossover trial, where one is comparing every patient to him or herself rather than to another patient.

So, there is always an appetite to do a crossover trial as long as the criteria for wash-out can be met, namely again that the primary intervention can dissipate completely to the baseline state before the alternative intervention is implemented.

“Placebo” is a fairly self-evident and well-understood term; placebo refers to the administration of a maneuver in a way that is identical to the principal maneuver except that the placebo is not expected to exert any clinical effect.

“Blinding” is the unawareness of either the investigator or of the patient to which the intervention is being administered. “Single-blinding” refers to the condition in which either the study or the investigator (but not both) is unaware, and “double-blinding” refers to the condition in which both the subjects and the investigators are unaware. There can be some subtle issues that compromise whether the patient is aware of the intervention that he or she is receiving and that can potentially condition the patient’s response, particularly if there is any subjective component of the assessment of the outcome. So, blinding is important.

With these terms describing the elements of a clinical study now described, let us turn to the types of studies that comprise clinical research. The first group of study types is what Dr. Feinstein called descriptive studies—studies that simply describe phenomena without comparison to a control group. As an example of a descriptive study, Sehgal et al7 recently described the workup of a focal, segmental pneumonia in a patient taking pembrolizumab for lung cancer. In this paper, there were four other cases of focal pneumonia accompanying pembrolizumab use that were assembled from the literature, making this descriptive paper a so-called case series. A “case series” differs from a “single case report,” which reports a single patient experience. Though limited in their ability to establish cause and effect, case reports and case series can help researchers develop proof of principle, so I would not discount the value of case reports.8

I can cite a case report from of my own experience that demonstrates this point. In 1987, I saw a patient from Buffalo who had primary biliary cirrhosis and the hepatopulmonary syndrome (HPS). She was so debilitated by her HPS that she could not stand up without desaturating severely. Although she had normal liver synthetic function, she was severely debilitated by her HPS and the decision was made to offer her a liver transplant, which, at that time, was considered to be relatively contraindicated. Much to everyone’s amazement and satisfaction, her HPS completely resolved after the transplant surgery. Her oxygenation and alveolar-arterial oxygen gradient normalized, and her clubbing resolved. We reported this in a case report, which began to affect the way people thought about the feasibility of liver transplant for the HPS.8 The lesson is: do not underestimate the power of a thoughtful case report.

The second group of research study types is called “cohort studies,” in which one actually compares outcomes between 2 groups in the study. Cohort studies fall into the bucket of either “observational cohort studies,” in which allocation to the compared maneuvers is not performed by randomization but by any other strategy, and “randomized trials.” In observational studies, allocation could occur through physician choice, as when the physician prescribes a treatment to 1 group but not another, or by patient choice or circumstance. For example, an observational cohort study of the risk of cigarette smoking would compare outcomes between smokers and non-smokers where the patient choses to smoke under his/her own volition. Alternatively, the circumstances of an exposure could allocate someone to the principal maneuver, as when we are studying the effect of exposure to World Trade Center dust in the firefighters who responded or of exposure to nuclear radiation in Hiroshima survivors. These are examples of observational cohort studies that compare exposed individuals to unexposed individuals, where the exposure did not occur by randomization but by choice or unfortunate circumstance.

In contrast to observational studies, allocation in randomized trials occurs through a formal process. Randomization has the specific purpose of attempting to ensure that patients are allocated to 2 comparative groups from the baseline group with comparable risk for developing the outcome measure. When randomization is effective, differences in study outcomes can be reliably ascribed to the intervention rather than to differences in the baseline susceptibility of the compared groups.

 

 

While randomization is an excellent strategy to ensure baseline similarity between compared groups, randomization can fail, and its effectiveness must be checked. Specifically, in a randomized trial, it is customary to examine the compared groups at baseline on all features that can affect the likelihood of developing the outcome measure. If the groups turn out to be dissimilar at baseline in an important way, then the study is at risk for bias, which is specifically called “susceptibility bias” in Feinstein’s construct. Obviously, the larger number of baseline clinical and demographic features that can condition the likelihood of developing the outcome measure, the more difficult it is to achieve baseline similarity between compared groups and the more important it becomes to ensure that randomization has been effective. In this circumstance, larger numbers of participants in both compared groups are generally needed. More about susceptibility bias later.

There are generally 2 types of randomized trials: the so-called “parallel controlled trials” in which each group receives either the principal or the comparative maneuver and is followed and “crossover trials” in which each compared group receives both the principal maneuver and the co-maneuver at different times after an effective wash-out period. Wash-out was discussed above. Figure 2 shows an example of a crossover trial examining the effects of terbutaline on diaphragmatic function.6 The investigators administered terbutaline for a week, measured transdiaphragmatic pressures, gave the patient a terbutaline vacation (the “wash-out period”), and then crossed over those patients who were initially receiving terbutaline to placebo and initial placebo recipients to terbutaline, having remeasured diaphragmatic function after the wash-out period to assure that the patient’s diaphragmatic function prior to the second crossover was identical to his/her baseline state. If this return to baseline is accomplished, then the criteria from effective wash-out are satisfied.

Types of bias in a clinical trial according to Feinstein
Now, with these basic structural terms of clinical research defined, bias will occupy the remainder of the discussion. By definition, bias in a clinical trial is any factor in the design or conduct of the trial, either external to the trial or internal to the trial, that can alter the results in a way that either threatens the reliability of attributing the differences in outcomes between the compared groups with the principal maneuver (“internal validity”) or limits the ability of the results, however internally valid, to be applied to a specific population beyond the study group (“external validity”) (Table 1).1 This again is because the main goal of cause-and-effect research is to make sure that you can attribute differences between the 2 compared groups at the end of the trial to the intervention under study and nothing else.

A comparison of surgery vs nonsurgical therapy for advanced lung cancer.
Figure 3. A comparison of surgery vs nonsurgical therapy for advanced lung cancer. An example of possible susceptibility bias.1

As we begin to talk about sources of bias, consider a study in which we compare survival of patients allocated to surgery vs nonsurgical therapy for lung cancer (Figure 3).1 This study is subject to the first type of so-called “internal bias” in the Feinsteinian construct—so-called “selection bias.” For example, all patients treated surgically were considered healthy enough by their doctors to undergo surgery, whereas patients treated without surgery may have been deemed inoperable because of comorbidities, lung dysfunction, cardiac dysfunction, and so on. If the results of such a comparison show that the mortality rate among surgical patients in this study was lower, the question then becomes: is the improved survival in surgical candidates due to the superior efficacy of surgery vs other therapy or was the enhanced survival due to the surgical patients being healthier to begin with? You can intuitively sense that the answer to this question is that the enhanced survival may be due to the better health of patients treated surgically rather than to the surgery itself because of how the patients were selected to receive it. So, this is a simple example of what Dr. Feinstein would call “susceptibility bias.” Susceptibility bias occurs when the 2 baseline groups are not comparably at risk or susceptible to developing the outcome measure, leading the naïve investigator in this specific example to attribute the difference in outcomes to the superiority of surgery when in fact it may have nothing to do with the surgery vs. the other maneuver. When susceptibility bias is in play, the difference between the outcomes in the compared groups could be attributed to the baseline imbalance of the groups rather than to the principal maneuver itself.

Turning back to the taxonomy of bias, there are four types that can threaten internal validity—“susceptibility,” “performance,” “detection,” and “transfer” bias—and 1 type of bias (called “external bias”) that can affect the generalizability of the study called “assembly bias” (Table 1).

Potential sources of bias in a randomized, controlled trial according to Feinstein.
Figure 4. Potential sources of bias in a randomized, controlled trial according to Feinstein.1

Figure 4 shows where these various sources of bias appear in the architecture of a clinical trial. As just discussed, susceptibility bias affects the baseline state and the comparability of the groups. Performance bias relates to how effective and how comparably the co-maneuvers are given and whether the primary intervention is potent enough to affect an outcome. Both transfer and detection bias operate in detecting the outcome, especially regarding the rigor and frequency with which they are investigated. Transfer bias has to do with selective loss to follow-up of those included in the trial. If there is a systematic reason for loss to follow-up that is related to the impact of the intervention, then the study is at risk for transfer bias. For example, in a randomized trial of drug A vs placebo for pneumonia, if drug A is effective but all the drug A recipients fail to follow-up because they feel too good to return for follow-up, then transfer bias could be causing the study to show nonefficacy even though the drug works. So, if those who respond favorably are systematically lost to follow-up, and if all the patients who felt lousy wanted to see the doctor and came back for follow-up, such transfer bias would bias towards nonefficacy. Specifically, only patients remaining in the trial would be those who failed to respond and that would dilute any difference between the 2 groups despite the active efficacy of drug A.

Hopefully, you are already beginning to get a sense that one has to be extremely disciplined in thinking about each of these sources of bias because they can have some very subtle nuances in randomized trials that can easily escape attention.

Returning to sources of bias, let’s consider the second type of bias, “performance bias.” Performance bias relates to the administration of the compared maneuvers—the primary or principal maneuver, compared with the comparative maneuver. Performance bias can occur when the main maneuver is not administered adequately or when the co-maneuvers are administered in an imbalanced way between the compared groups. Consider the example of the Long-Term Oxygen Treatment Trial (LOTT) trial, which compared use of supplemental oxygen with no supplemental oxygen in patients with stable COPD and resting or exercise-induced moderate desaturation.9 The principal outcome measure of LOTT was all-cause hospitalization or death. In such a study, many potential sources of performance bias exist. For example, performance bias might exist if none of the patients allocated to oxygen actually used supplemental oxygen. Alternately, to the extent that use of inhaled corticosteroids or antimuscarinic agents lessens the risk of COPD exacerbation, performance bias could occur if use of these co-maneuvers was imbalanced between the compared groups. As a specific extreme circumstance, if all patients in the nonoxygen group used these inhalers but none of the patients in the oxygen group did, then a lack of difference between exacerbation frequency could be related to this imbalance in co-maneuvers (a form of performance bias) rather than to the lack of efficacy of supplemental oxygen.

 

 

“Compliance bias” is a subset of performance bias which occurs when 2 conditions are satisfied: (1) the main maneuver is not administered adequately, and (2) the investigator is unaware of that nonreceipt so that this cannot be accounted for in interpreting the study results. For example, if a drug has efficacy but if no one in the treatment arm of the trial takes the drug, the absence of a difference in outcomes between the compared groups will be ascribed to nonefficacy, whereas “compliance bias” (ie, no one actually took the drug) could actually be the cause. Ideally, randomized studies should be evaluated on an “intention to treat” basis irrespective of compliance, but there is an analytic approach called “per protocol” analysis in which you can analyze the results according to whether the patient actually used the intervention in an effective way. “Per protocol” analysis is a secondary analysis of the primary results but it can nonetheless help determine whether the negative result is likely related to noncompliance or not.

A third type of internal bias, “detection bias,” is fairly straightforward. Detection bias is related to how avidly and how comparably the outcomes are measured between the 2 compared groups. Let’s say that you are conducting a trial of a new antibiotic and the primary outcome is colony counts on petri dishes of plated collected specimens. If the technicians who read the petri dish counts are unblinded, they may look at the colony counts with a biased eye, seeing fewer colonies on plates collected from patients receiving the antibiotic.

Overall, detection bias occurs when outcomes are ascertained or detected unequally between the compared groups, and detection bias can involve any of the following: is there comparable surveillance of the 2 groups for analysis of the outcome measure? Are the diagnostic tests comparably performed in both groups and is the interpretation comparably unbiased with equipoise? Investigators who know which patients are receiving an active drug and those who are not could experience subliminal bias that renders them more likely to find that the drug under study is efficacious.

Depending on the principal study maneuver, ensuring blinding can be challenging. To demonstrate this point, let’s consider the example of conducting a randomized control trial of Vicks VapoRub. Vicks VapoRub is an old product that smells like wintergreen and that mothers used to rub on the chests of their infants in the hope of speeding recovery from colds and bronchitis episodes. It was felt that the distinctive smell of the product was materially related to wintergreen, which gives rise to the odor. So, imagine a randomized trial of Vicks VaporRub. A trial is designed in which sick children receive Vicks VapoRub on their chest and others receive a placebo rub that lacks the distinctive wintergreen odor. But, the odor itself is felt to be related to how Vicks VapoRub actually works. Thus, it is the odor itself that creates the blinding challenge here.

The primary outcomes in this study are the duration of the child’s cold symptoms, as ascertained by pediatricians actually examining the children. So, pediatricians would come and listen to the infants’ chests: “Yeah, this chest is clear, but this other infant is still full of rhonchi,” and they would ascertain the outcome measure in this way. So, my blinding question to you is: how do you blind a trial of Vicks VapoRub given the conditions described? Namely, you put the VapoRub on the chest, it smells and the smell is the intervention—how do you blind such a trial? 

The clever answer is that you should put Vicks VapoRub on the upper lips of all the examiners, so what they smell is Vicks VapoRub independent of whether the child they are examining also has the Vicks VapoRub or placebo on their chest. In this way, single blinding of the examiners is preserved and detection bias is averted. It is important to point out that double blinding could also be achieved by placing Vicks VapoRub on the child’s upper lip, but there is little reason to suspect that the infants being studied have a bias related to whether they smell the Vicks VapoRub.

The fourth potential source of internal bias is called “transfer bias.” Transfer bias is the selective loss to follow-up of patients from 1 of the 2 compared groups in the trial for a systematic reason. By systematic, I mean that that the drop-out is associated with the development of the outcome event or some impact of the intervention regarding the likelihood to develop the outcome event. As an example, if all patients respond favorably to a drug and everybody fails to follow up because they feel too good to come back, then that would bias the study towards nonefficacy even in the face of an efficacious intervention.

Finally, let’s consider a source of bias that can affect the “external validity,” or the generalizability of the study results to populations other than that included in the study itself. Dr. Feinstein calls this 5th type of bias “assembly bias” (Table 1).1 Assembly bias occurs when the results of the study cannot be reliably applied to populations outside the study itself.

For example, if I screen patients during a study of digoxin for heart rate control in atrial fibrillation, I could establish whether the subject was compliant or not by checking his/her serum digoxin levels. Serum levels of 0 indicate that the patient has not taken the digoxin. If I include a run-in period for the trial—an interval before the actual study when I am assessing potential subjects’ eligibility to participate—and check serum digoxin levels to include only patients who are shown to be taking the drug, then I am screening for study inclusion on compliance. In this way, I will have assembled a population that is highly compliant so that I can truly assess whether digoxin has efficacy in controlling the heart rate in patients with atrial fibrillation. At the same time, this study population is not highly representative of the population of patients with atrial fibrillation at large, because we know that rates of drug noncompliances may be as high as 30% to 40%. So, culling a population with run-in periods on demonstrated compliance criteria may be very important to assess efficacy (ie, whether the drug works), but this design will trade off on the effectiveness of the drug (ie, which asks the question “does the drug work in actual practice?”). This is because, in the yin-yang between assessing efficacy and assessing effectiveness, the focus on assessing efficacy naturally undermines the ability to assess whether the drug works in real-world conditions.

As another example of potential assembly bias, let’s say you are studying an antihypertensive drug at a Veterans Administration (VA) hospital, where most veterans are men. But you are treating women in your practice and wonder whether the drug, which works in a predominately male population, will work in your female patients. So, there could be assembly bias in applying the results of a VA study to a non-VA predominantly female population.

Having now described the design of clinical trials and the major sources of bias, let’s apply this thinking to the earliest clinical trial. James Lind, a British Naval officer, was credited with conducting the first clinical trial of citrus fruits for scurvy while sailing on the ship Salisbury in 1747.2 The question that Lind addressed was “does citrus fruit treat and prevent scurvy?” In describing this trial, Lind stated “I took 12 patients with scurvy, these patients were as similar as I could have them, had one diet common to all.” As you read this through your new Feinsteinian bias lens, Lind is addressing 2 potential sources of bias, namely, susceptibility bias and performance bias. In trying to make the “cases as similar as I could have them,” he is trying to avoid susceptibility bias and in “providing one diet common to all,” he is trying to avoid performance bias.

In terms of the intervention in this trial, these 12 patients were allocated in pairs to several interventions: a quart of cider a day, 25 drops of elixir of vitriol 3 times a day on an empty stomach, 2 spoonsful of vinegar 3 times a day on an empty stomach, ½ pint a day of sea water, 2 oranges and 1 lemon given every day, and a “bigness of nutmeg” 3 times per day. In describing the outcome of the trial, Lind states “the consequence was that the most sudden and visible good effects were perceived from the use of oranges and lemons; one of those who had taken them, being at the end of 6 days fit for duty. The spots were not indeed at that time quite off his body, nor his gums sound, but without any other medicine then a gargarism of elixir vitriol, he became quite healthy before we came into Plymouth which was on the 16th of June. The other was the best recovered of any in his condition; and being now deemed pretty well, was appointed nurse to the rest of the sick.”

 

 

In analyzing this trial, we could characterize it as a parallel controlled trial. Whether the allocation was done by randomization is not clear, but it was certainly an observational cohort study in that there were concurrent controls who were treated as similarly as possible except for the principal maneuver, which was the administration of citrus fruit. Already mentioned was the attention to averting susceptibility and performance bias. There was no evidence of compliance bias as the interventions were enforced, nor was there evidence of transfer bias because all subjects who were enrolled in the study completed the study because they were a captive group on a sailing ship. Finally, the likelihood of assembly bias seems small, as these sailors seemed to be representative of victims of scurvy in general, namely in being otherwise deprived of access to citrus fruits.

In terms of the statistical results of this study, subsequent analysis of the research showed that the impact of lemons and oranges was dramatic and showed a trend (P = .09) towards statistical significance. Notwithstanding the lack of a P < .05, Dr. Feinstein would likely say that this study satisfied the “intra-ocular test” in that the efficacy of the citrus fruit was so dramatic that it “hit you between the eyes.” He often argued that the widespread practice of prescribing penicillin for pneumococcal pneumonia was not based on the results of a convincing randomized controlled trial because the efficacy of penicillin in that setting was so dramatic that a randomized trial was not necessary (and potentially even unethical if the condition of “intra-ocular” efficacy was satisfied).

The final question to address in this lecture is whether randomized controlled trials, for all their rigor, always produce more reliable results than observational studies. This issue has been addressed by several authors.10–12 Sacks et al10 contended in 1983 that observational studies systematically overestimate the magnitude of association between exposure and outcome and therefore argued that randomized trials were more reliable than observational studies. Subsequent analyses tended to challenge this view.11,12 Specifically, Benson and Hartz11 compared the results of 136 reports regarding 19 different therapies that were studied between 1985 and 1998. In only 2 of the 19 analyses did the treatment effects in the observational studies fall outside the 95% confidence interval for the randomized controlled trial results. In this way, these authors argued that observational studies generally are concordant with the results of randomized trials. They stated that “our finding that observational studies and randomized controlled trials usually produce similar results differs from the conclusions of previous authors. The fundamental criticism of observational studies is that unrecognized confounding factors may distort the results. According to the conventional wisdom, this distortion is sufficiently common and unpredictable that observational studies are not liable and should not be funded. Our results suggested observational studies usually do provide valid information.”11

An additional analysis of this issue was performed by Concato et al,12 who identified 99 articles regarding 5 clinical topics. Again, the results from randomized trials were compared with those of observational cohort or case-controlled studies regarding the same intervention. The authors reported that “contrary to prevailing belief, the average results from well-designed observational studies did not systematically overestimate the magnitude of the associations between exposure and outcome as compared with the results of randomized, controlled trials on the same topic. Rather, the summary results of randomized, controlled trials and observational studies were remarkably similar.”12

On the basis of these studies, it appears that randomized control trials continue to serve as the gold standard in clinical research, but we must also recognize that circumstances often preclude the conduct of a randomized trial. As an example, consider a randomized trial of whether cigarette smoking is harmful, which, given the strong suspicion of harm, would be unethical in that patients cannot be randomized to smoke. Similarly, from the example before, a randomized trial of penicillin for pneumococcal pneumonia would be unethical because denying patients in the placebo group access to penicillin would exclude them from access to a drug that has “intra-ocular” efficacy. In circumstances like these, well-performed observational studies that are attentive to sources of bias can likely produce comparably reliable results to randomized trials.

In the end, of course, the interpretation of the study results requires the reader’s careful attention to potential sources of bias that can compromise study validity. The hope is that with Dr. Feinstein’s framework, you can be better equipped to think critically about study results that you review and to keenly ascertain whether there is any threat to internal or to external validity. Similarly, as you go on to design clinical trials yourselves, you can pay attention to these potential sources of bias that, if present, can compromise the reliability of the study conclusions internally or their applicability to patients outside of the study.

I am flattered to present the inaugural talk in the biostatistics and clinical research design series on the architecture of clinical research. This content is based on the teachings of my mentor, Dr. Alvan Feinstein, who together with Dr. David Sackett, is credited with pioneering clinical epidemiology. Dr. Feinstein was a Sterling Professor at the Yale School of Medicine. His main opus of work is a book called, Clinical Epidemiology: The Architecture of Clinical Research.1 This paper is named in credit to Dr. Feinstein’s enormous contribution. I will review some important terms defined by Dr. Feinstein to provide the background necessary for the remainder of the talks in this series.

To start, I will frame this topic by asking the following question: Why do we do research? I’ll talk about the basic structure of research studies and provide a taxonomy, as Dr. Feinstein would say, a nomenclature with which to understand trial design and the sources of bias in those trials. Then, I will discuss these sources of bias in detail using the taxonomy that Dr. Feinstein described in his aforementioned book. Finally, I will share with you some examples of bias in clinical trials to help you better understand these concepts.

Now, the answer to the basic question posed above is: basically, we do cause-and-effect research to establish the causality of a risk factor or the efficacy of a therapy. Does cigarette smoking cause lung cancer? Does taking hydrochlorothiazide help systemic hypertension? Does air pollution worsen asthma? Does supplemental oxygen help patients with chronic obstructive pulmonary disease (COPD)?

Cause-and-effect research can be subsumed under 2 broad issues: causal risk factors and therapeutic efficacy. In his review of early false understandings in medicine that were based on anecdotal observation alone, Thomas cites many examples—“the undue longevity of useless and even harmful drugs can be laid at the door of authority,” ie, empiricism, lack of rigorous research.2 The field is full of these: yellow fever causality, the value of cupping, and even intermittent mandatory ventilation when it was described by John Downs in 1973 and touted as a superior mode for weaning patients from mechanical ventilation.3 Twenty-five years later, randomized controlled trials by Brochard et al4 indicated not only that intermittent mandatory ventilation was not the best mode to wean but was, in fact, the worst mode for weaning patients from mechanical ventilation compared with either pressure support or spontaneous breathing trials. Many more examples exist to demonstrate the false understandings that can be ascribed to lack of rigorous study or evidence in medicine.

Design of a controlled trial according to Feinstein.
Figure 1. Design of a controlled trial according to Feinstein.1

Before systematically exploring the sources of bias in Feinstein’s construct, let us define some very basic terms from his book. Dr. Feinstein talks about the baseline state, which refers to the group of patients under study who are culled from a larger population to whom the results are intended to be applied (Figure 1).1 This baseline group is hopefully representative of this larger target population. As a nod to the later discussion, Dr. Feinstein would call bias introduced by unusual assembly of the study population from the larger intended population as “assembly bias.” So, if the group under study is not representative of either the patients you see or the world of patients with this condition or if there is something special or distinctively nonrepresentative about the study population, then the results may be subject to “assembly bias.” Assembly bias can compromise the so-called “external” validity of the study—its ability to be applied to populations beyond the study group.

Having assembled a baseline group for study, that group is classically allocated to 1 of 2 (or sometimes more than 2) compared therapies. In a controlled trial, patients can be allocated using a variety of strategies, including randomization. Using the paradigm diagram (Figure 1, which considers a 2-arm trial), patients are allocated to 1 of 2 compared groups—group A and group B. Then, in a treatment trial, 1 group receives the principal maneuver, which is the drug or intervention under study—for example, supplemental oxygen for patients with COPD. The comparative maneuver is allocated to group B, which also receives all the other treatments (called “co-maneuvers”) that are used to treat the condition under study. In a trial of supplemental oxygen for COPD evaluating lung function and exacerbation frequency as outcome measures, such co-maneuvers might include inhaled bronchodilators, inhaled corticosteroids, pulmonary rehabilitation, and Pneumovax vaccine. Ideally, these co-maneuvers are equally distributed between the compared groups (A and B).

So, in summary, we have a comparative maneuver, which is the nonadministration of supplemental oxygen in this proposed trial of supplemental oxygen in COPD, the principal maneuver—administration of oxygen—and all the co-maneuvers that are ideally equally distributed between both groups. This balanced distribution of co-maneuvers between the compared groups helps to ensure that any differences in the study outcome measures (ie, what is counted as the main impact of the intervention under study) can be solely attributed to the principal maneuver. When this condition—that the difference in outcomes can be reliably ascribed to the study intervention—is satisfied, the study is felt to be “internally” valid. As we will see, ensuring internal validity requires freedom from the many sources of what Dr. Feinstein calls “internal bias.”

Back to basic terms: “cohort” in Dr. Feinstein’s language is a group that shares common traits and is followed forward in a longitudinal study. The “outcome measure” is self-evident—it is what is being measured, with the “primary outcome” being the pre-defined measure that is considered the most important (and ideally most clinically relevant) impact of the study intervention. Later in this series of lectures, there will be discussions of power calculations and the so-called “effect size”—the magnitude of effect that the intervention is expected to produce and that is ideally deemed clinically important.

 

 

An important consideration in designing a trial is to define and declare the primary outcome measure carefully because defining the primary outcome measure has important implications for the study. I will provide an example from the alpha-1 antitrypsin deficiency literature. Some of you have probably read what has been called the RAPID trial.5 RAPID was a trial of augmentation therapy vs placebo in patients with severe alpha-1 antitrypsin deficiency. The primary outcome measure (which was pre-negotiated with the US Food and Drug Administration [FDA]) was computer tomography (CT) lung density determined at functional residual capacity (FRC) and total lung capacity (TLC). The trial failed to achieve statistical significance in regard to CT lung density, although the study authors argued that CT density measurements made at TLC were more reproducible than those made at FRC. When the results were analyzed by TLC alone, the results were statistically significant, but when they were analyzed with FRC and TLC combined, they were not. In the end, based on the pre-negotiated primary outcome measure of CT density based on both FRC and TLC, the FDA rejected the proposal for a label change to say that augmentation therapy slowed the loss of lung density even though the weight of evidence was clearly in its favor. This case exemplifies just how critical the choice of primary outcome measure can be.

Design of a randomized crossover trial of terbutaline for diaphragmatic function.
Figure 2. Design of a randomized crossover trial of terbutaline for diaphragmatic function. The wash-out period separates the first and the second interventions (begins at the star in the diagram).

The wash-out period refers to an interval in a subset of randomized trials called “crossover trials” in which the primary intervention is discontinued and the patient returns to his baseline state before the comparative maneuver is then implemented (Figure 2).6 In order to perform a crossover trial, it is important that the effects of the initial intervention can “wash out” or be fully extinguished. So, for example, in trials of radiation therapy vs surgery, it is impossible to do a crossover trial because the effects of radiation can never completely wash out nor can those of surgery, which are similarly permanent. For example, we cannot replace the colon once it is resected for cancer or replace the appendix once removed. Therefore, producing a wash-out requires some very specific pharmacokinetic and pharmacodynamic features in order for a crossover trial to be considered. Later talks in this series will discuss the enhanced statistical power of a crossover trial, where one is comparing every patient to him or herself rather than to another patient.

So, there is always an appetite to do a crossover trial as long as the criteria for wash-out can be met, namely again that the primary intervention can dissipate completely to the baseline state before the alternative intervention is implemented.

“Placebo” is a fairly self-evident and well-understood term; placebo refers to the administration of a maneuver in a way that is identical to the principal maneuver except that the placebo is not expected to exert any clinical effect.

“Blinding” is the unawareness of either the investigator or of the patient to which the intervention is being administered. “Single-blinding” refers to the condition in which either the study or the investigator (but not both) is unaware, and “double-blinding” refers to the condition in which both the subjects and the investigators are unaware. There can be some subtle issues that compromise whether the patient is aware of the intervention that he or she is receiving and that can potentially condition the patient’s response, particularly if there is any subjective component of the assessment of the outcome. So, blinding is important.

With these terms describing the elements of a clinical study now described, let us turn to the types of studies that comprise clinical research. The first group of study types is what Dr. Feinstein called descriptive studies—studies that simply describe phenomena without comparison to a control group. As an example of a descriptive study, Sehgal et al7 recently described the workup of a focal, segmental pneumonia in a patient taking pembrolizumab for lung cancer. In this paper, there were four other cases of focal pneumonia accompanying pembrolizumab use that were assembled from the literature, making this descriptive paper a so-called case series. A “case series” differs from a “single case report,” which reports a single patient experience. Though limited in their ability to establish cause and effect, case reports and case series can help researchers develop proof of principle, so I would not discount the value of case reports.8

I can cite a case report from of my own experience that demonstrates this point. In 1987, I saw a patient from Buffalo who had primary biliary cirrhosis and the hepatopulmonary syndrome (HPS). She was so debilitated by her HPS that she could not stand up without desaturating severely. Although she had normal liver synthetic function, she was severely debilitated by her HPS and the decision was made to offer her a liver transplant, which, at that time, was considered to be relatively contraindicated. Much to everyone’s amazement and satisfaction, her HPS completely resolved after the transplant surgery. Her oxygenation and alveolar-arterial oxygen gradient normalized, and her clubbing resolved. We reported this in a case report, which began to affect the way people thought about the feasibility of liver transplant for the HPS.8 The lesson is: do not underestimate the power of a thoughtful case report.

The second group of research study types is called “cohort studies,” in which one actually compares outcomes between 2 groups in the study. Cohort studies fall into the bucket of either “observational cohort studies,” in which allocation to the compared maneuvers is not performed by randomization but by any other strategy, and “randomized trials.” In observational studies, allocation could occur through physician choice, as when the physician prescribes a treatment to 1 group but not another, or by patient choice or circumstance. For example, an observational cohort study of the risk of cigarette smoking would compare outcomes between smokers and non-smokers where the patient choses to smoke under his/her own volition. Alternatively, the circumstances of an exposure could allocate someone to the principal maneuver, as when we are studying the effect of exposure to World Trade Center dust in the firefighters who responded or of exposure to nuclear radiation in Hiroshima survivors. These are examples of observational cohort studies that compare exposed individuals to unexposed individuals, where the exposure did not occur by randomization but by choice or unfortunate circumstance.

In contrast to observational studies, allocation in randomized trials occurs through a formal process. Randomization has the specific purpose of attempting to ensure that patients are allocated to 2 comparative groups from the baseline group with comparable risk for developing the outcome measure. When randomization is effective, differences in study outcomes can be reliably ascribed to the intervention rather than to differences in the baseline susceptibility of the compared groups.

 

 

While randomization is an excellent strategy to ensure baseline similarity between compared groups, randomization can fail, and its effectiveness must be checked. Specifically, in a randomized trial, it is customary to examine the compared groups at baseline on all features that can affect the likelihood of developing the outcome measure. If the groups turn out to be dissimilar at baseline in an important way, then the study is at risk for bias, which is specifically called “susceptibility bias” in Feinstein’s construct. Obviously, the larger number of baseline clinical and demographic features that can condition the likelihood of developing the outcome measure, the more difficult it is to achieve baseline similarity between compared groups and the more important it becomes to ensure that randomization has been effective. In this circumstance, larger numbers of participants in both compared groups are generally needed. More about susceptibility bias later.

There are generally 2 types of randomized trials: the so-called “parallel controlled trials” in which each group receives either the principal or the comparative maneuver and is followed and “crossover trials” in which each compared group receives both the principal maneuver and the co-maneuver at different times after an effective wash-out period. Wash-out was discussed above. Figure 2 shows an example of a crossover trial examining the effects of terbutaline on diaphragmatic function.6 The investigators administered terbutaline for a week, measured transdiaphragmatic pressures, gave the patient a terbutaline vacation (the “wash-out period”), and then crossed over those patients who were initially receiving terbutaline to placebo and initial placebo recipients to terbutaline, having remeasured diaphragmatic function after the wash-out period to assure that the patient’s diaphragmatic function prior to the second crossover was identical to his/her baseline state. If this return to baseline is accomplished, then the criteria from effective wash-out are satisfied.

Types of bias in a clinical trial according to Feinstein
Now, with these basic structural terms of clinical research defined, bias will occupy the remainder of the discussion. By definition, bias in a clinical trial is any factor in the design or conduct of the trial, either external to the trial or internal to the trial, that can alter the results in a way that either threatens the reliability of attributing the differences in outcomes between the compared groups with the principal maneuver (“internal validity”) or limits the ability of the results, however internally valid, to be applied to a specific population beyond the study group (“external validity”) (Table 1).1 This again is because the main goal of cause-and-effect research is to make sure that you can attribute differences between the 2 compared groups at the end of the trial to the intervention under study and nothing else.

A comparison of surgery vs nonsurgical therapy for advanced lung cancer.
Figure 3. A comparison of surgery vs nonsurgical therapy for advanced lung cancer. An example of possible susceptibility bias.1

As we begin to talk about sources of bias, consider a study in which we compare survival of patients allocated to surgery vs nonsurgical therapy for lung cancer (Figure 3).1 This study is subject to the first type of so-called “internal bias” in the Feinsteinian construct—so-called “selection bias.” For example, all patients treated surgically were considered healthy enough by their doctors to undergo surgery, whereas patients treated without surgery may have been deemed inoperable because of comorbidities, lung dysfunction, cardiac dysfunction, and so on. If the results of such a comparison show that the mortality rate among surgical patients in this study was lower, the question then becomes: is the improved survival in surgical candidates due to the superior efficacy of surgery vs other therapy or was the enhanced survival due to the surgical patients being healthier to begin with? You can intuitively sense that the answer to this question is that the enhanced survival may be due to the better health of patients treated surgically rather than to the surgery itself because of how the patients were selected to receive it. So, this is a simple example of what Dr. Feinstein would call “susceptibility bias.” Susceptibility bias occurs when the 2 baseline groups are not comparably at risk or susceptible to developing the outcome measure, leading the naïve investigator in this specific example to attribute the difference in outcomes to the superiority of surgery when in fact it may have nothing to do with the surgery vs. the other maneuver. When susceptibility bias is in play, the difference between the outcomes in the compared groups could be attributed to the baseline imbalance of the groups rather than to the principal maneuver itself.

Turning back to the taxonomy of bias, there are four types that can threaten internal validity—“susceptibility,” “performance,” “detection,” and “transfer” bias—and 1 type of bias (called “external bias”) that can affect the generalizability of the study called “assembly bias” (Table 1).

Potential sources of bias in a randomized, controlled trial according to Feinstein.
Figure 4. Potential sources of bias in a randomized, controlled trial according to Feinstein.1

Figure 4 shows where these various sources of bias appear in the architecture of a clinical trial. As just discussed, susceptibility bias affects the baseline state and the comparability of the groups. Performance bias relates to how effective and how comparably the co-maneuvers are given and whether the primary intervention is potent enough to affect an outcome. Both transfer and detection bias operate in detecting the outcome, especially regarding the rigor and frequency with which they are investigated. Transfer bias has to do with selective loss to follow-up of those included in the trial. If there is a systematic reason for loss to follow-up that is related to the impact of the intervention, then the study is at risk for transfer bias. For example, in a randomized trial of drug A vs placebo for pneumonia, if drug A is effective but all the drug A recipients fail to follow-up because they feel too good to return for follow-up, then transfer bias could be causing the study to show nonefficacy even though the drug works. So, if those who respond favorably are systematically lost to follow-up, and if all the patients who felt lousy wanted to see the doctor and came back for follow-up, such transfer bias would bias towards nonefficacy. Specifically, only patients remaining in the trial would be those who failed to respond and that would dilute any difference between the 2 groups despite the active efficacy of drug A.

Hopefully, you are already beginning to get a sense that one has to be extremely disciplined in thinking about each of these sources of bias because they can have some very subtle nuances in randomized trials that can easily escape attention.

Returning to sources of bias, let’s consider the second type of bias, “performance bias.” Performance bias relates to the administration of the compared maneuvers—the primary or principal maneuver, compared with the comparative maneuver. Performance bias can occur when the main maneuver is not administered adequately or when the co-maneuvers are administered in an imbalanced way between the compared groups. Consider the example of the Long-Term Oxygen Treatment Trial (LOTT) trial, which compared use of supplemental oxygen with no supplemental oxygen in patients with stable COPD and resting or exercise-induced moderate desaturation.9 The principal outcome measure of LOTT was all-cause hospitalization or death. In such a study, many potential sources of performance bias exist. For example, performance bias might exist if none of the patients allocated to oxygen actually used supplemental oxygen. Alternately, to the extent that use of inhaled corticosteroids or antimuscarinic agents lessens the risk of COPD exacerbation, performance bias could occur if use of these co-maneuvers was imbalanced between the compared groups. As a specific extreme circumstance, if all patients in the nonoxygen group used these inhalers but none of the patients in the oxygen group did, then a lack of difference between exacerbation frequency could be related to this imbalance in co-maneuvers (a form of performance bias) rather than to the lack of efficacy of supplemental oxygen.

 

 

“Compliance bias” is a subset of performance bias which occurs when 2 conditions are satisfied: (1) the main maneuver is not administered adequately, and (2) the investigator is unaware of that nonreceipt so that this cannot be accounted for in interpreting the study results. For example, if a drug has efficacy but if no one in the treatment arm of the trial takes the drug, the absence of a difference in outcomes between the compared groups will be ascribed to nonefficacy, whereas “compliance bias” (ie, no one actually took the drug) could actually be the cause. Ideally, randomized studies should be evaluated on an “intention to treat” basis irrespective of compliance, but there is an analytic approach called “per protocol” analysis in which you can analyze the results according to whether the patient actually used the intervention in an effective way. “Per protocol” analysis is a secondary analysis of the primary results but it can nonetheless help determine whether the negative result is likely related to noncompliance or not.

A third type of internal bias, “detection bias,” is fairly straightforward. Detection bias is related to how avidly and how comparably the outcomes are measured between the 2 compared groups. Let’s say that you are conducting a trial of a new antibiotic and the primary outcome is colony counts on petri dishes of plated collected specimens. If the technicians who read the petri dish counts are unblinded, they may look at the colony counts with a biased eye, seeing fewer colonies on plates collected from patients receiving the antibiotic.

Overall, detection bias occurs when outcomes are ascertained or detected unequally between the compared groups, and detection bias can involve any of the following: is there comparable surveillance of the 2 groups for analysis of the outcome measure? Are the diagnostic tests comparably performed in both groups and is the interpretation comparably unbiased with equipoise? Investigators who know which patients are receiving an active drug and those who are not could experience subliminal bias that renders them more likely to find that the drug under study is efficacious.

Depending on the principal study maneuver, ensuring blinding can be challenging. To demonstrate this point, let’s consider the example of conducting a randomized control trial of Vicks VapoRub. Vicks VapoRub is an old product that smells like wintergreen and that mothers used to rub on the chests of their infants in the hope of speeding recovery from colds and bronchitis episodes. It was felt that the distinctive smell of the product was materially related to wintergreen, which gives rise to the odor. So, imagine a randomized trial of Vicks VaporRub. A trial is designed in which sick children receive Vicks VapoRub on their chest and others receive a placebo rub that lacks the distinctive wintergreen odor. But, the odor itself is felt to be related to how Vicks VapoRub actually works. Thus, it is the odor itself that creates the blinding challenge here.

The primary outcomes in this study are the duration of the child’s cold symptoms, as ascertained by pediatricians actually examining the children. So, pediatricians would come and listen to the infants’ chests: “Yeah, this chest is clear, but this other infant is still full of rhonchi,” and they would ascertain the outcome measure in this way. So, my blinding question to you is: how do you blind a trial of Vicks VapoRub given the conditions described? Namely, you put the VapoRub on the chest, it smells and the smell is the intervention—how do you blind such a trial? 

The clever answer is that you should put Vicks VapoRub on the upper lips of all the examiners, so what they smell is Vicks VapoRub independent of whether the child they are examining also has the Vicks VapoRub or placebo on their chest. In this way, single blinding of the examiners is preserved and detection bias is averted. It is important to point out that double blinding could also be achieved by placing Vicks VapoRub on the child’s upper lip, but there is little reason to suspect that the infants being studied have a bias related to whether they smell the Vicks VapoRub.

The fourth potential source of internal bias is called “transfer bias.” Transfer bias is the selective loss to follow-up of patients from 1 of the 2 compared groups in the trial for a systematic reason. By systematic, I mean that that the drop-out is associated with the development of the outcome event or some impact of the intervention regarding the likelihood to develop the outcome event. As an example, if all patients respond favorably to a drug and everybody fails to follow up because they feel too good to come back, then that would bias the study towards nonefficacy even in the face of an efficacious intervention.

Finally, let’s consider a source of bias that can affect the “external validity,” or the generalizability of the study results to populations other than that included in the study itself. Dr. Feinstein calls this 5th type of bias “assembly bias” (Table 1).1 Assembly bias occurs when the results of the study cannot be reliably applied to populations outside the study itself.

For example, if I screen patients during a study of digoxin for heart rate control in atrial fibrillation, I could establish whether the subject was compliant or not by checking his/her serum digoxin levels. Serum levels of 0 indicate that the patient has not taken the digoxin. If I include a run-in period for the trial—an interval before the actual study when I am assessing potential subjects’ eligibility to participate—and check serum digoxin levels to include only patients who are shown to be taking the drug, then I am screening for study inclusion on compliance. In this way, I will have assembled a population that is highly compliant so that I can truly assess whether digoxin has efficacy in controlling the heart rate in patients with atrial fibrillation. At the same time, this study population is not highly representative of the population of patients with atrial fibrillation at large, because we know that rates of drug noncompliances may be as high as 30% to 40%. So, culling a population with run-in periods on demonstrated compliance criteria may be very important to assess efficacy (ie, whether the drug works), but this design will trade off on the effectiveness of the drug (ie, which asks the question “does the drug work in actual practice?”). This is because, in the yin-yang between assessing efficacy and assessing effectiveness, the focus on assessing efficacy naturally undermines the ability to assess whether the drug works in real-world conditions.

As another example of potential assembly bias, let’s say you are studying an antihypertensive drug at a Veterans Administration (VA) hospital, where most veterans are men. But you are treating women in your practice and wonder whether the drug, which works in a predominately male population, will work in your female patients. So, there could be assembly bias in applying the results of a VA study to a non-VA predominantly female population.

Having now described the design of clinical trials and the major sources of bias, let’s apply this thinking to the earliest clinical trial. James Lind, a British Naval officer, was credited with conducting the first clinical trial of citrus fruits for scurvy while sailing on the ship Salisbury in 1747.2 The question that Lind addressed was “does citrus fruit treat and prevent scurvy?” In describing this trial, Lind stated “I took 12 patients with scurvy, these patients were as similar as I could have them, had one diet common to all.” As you read this through your new Feinsteinian bias lens, Lind is addressing 2 potential sources of bias, namely, susceptibility bias and performance bias. In trying to make the “cases as similar as I could have them,” he is trying to avoid susceptibility bias and in “providing one diet common to all,” he is trying to avoid performance bias.

In terms of the intervention in this trial, these 12 patients were allocated in pairs to several interventions: a quart of cider a day, 25 drops of elixir of vitriol 3 times a day on an empty stomach, 2 spoonsful of vinegar 3 times a day on an empty stomach, ½ pint a day of sea water, 2 oranges and 1 lemon given every day, and a “bigness of nutmeg” 3 times per day. In describing the outcome of the trial, Lind states “the consequence was that the most sudden and visible good effects were perceived from the use of oranges and lemons; one of those who had taken them, being at the end of 6 days fit for duty. The spots were not indeed at that time quite off his body, nor his gums sound, but without any other medicine then a gargarism of elixir vitriol, he became quite healthy before we came into Plymouth which was on the 16th of June. The other was the best recovered of any in his condition; and being now deemed pretty well, was appointed nurse to the rest of the sick.”

 

 

In analyzing this trial, we could characterize it as a parallel controlled trial. Whether the allocation was done by randomization is not clear, but it was certainly an observational cohort study in that there were concurrent controls who were treated as similarly as possible except for the principal maneuver, which was the administration of citrus fruit. Already mentioned was the attention to averting susceptibility and performance bias. There was no evidence of compliance bias as the interventions were enforced, nor was there evidence of transfer bias because all subjects who were enrolled in the study completed the study because they were a captive group on a sailing ship. Finally, the likelihood of assembly bias seems small, as these sailors seemed to be representative of victims of scurvy in general, namely in being otherwise deprived of access to citrus fruits.

In terms of the statistical results of this study, subsequent analysis of the research showed that the impact of lemons and oranges was dramatic and showed a trend (P = .09) towards statistical significance. Notwithstanding the lack of a P < .05, Dr. Feinstein would likely say that this study satisfied the “intra-ocular test” in that the efficacy of the citrus fruit was so dramatic that it “hit you between the eyes.” He often argued that the widespread practice of prescribing penicillin for pneumococcal pneumonia was not based on the results of a convincing randomized controlled trial because the efficacy of penicillin in that setting was so dramatic that a randomized trial was not necessary (and potentially even unethical if the condition of “intra-ocular” efficacy was satisfied).

The final question to address in this lecture is whether randomized controlled trials, for all their rigor, always produce more reliable results than observational studies. This issue has been addressed by several authors.10–12 Sacks et al10 contended in 1983 that observational studies systematically overestimate the magnitude of association between exposure and outcome and therefore argued that randomized trials were more reliable than observational studies. Subsequent analyses tended to challenge this view.11,12 Specifically, Benson and Hartz11 compared the results of 136 reports regarding 19 different therapies that were studied between 1985 and 1998. In only 2 of the 19 analyses did the treatment effects in the observational studies fall outside the 95% confidence interval for the randomized controlled trial results. In this way, these authors argued that observational studies generally are concordant with the results of randomized trials. They stated that “our finding that observational studies and randomized controlled trials usually produce similar results differs from the conclusions of previous authors. The fundamental criticism of observational studies is that unrecognized confounding factors may distort the results. According to the conventional wisdom, this distortion is sufficiently common and unpredictable that observational studies are not liable and should not be funded. Our results suggested observational studies usually do provide valid information.”11

An additional analysis of this issue was performed by Concato et al,12 who identified 99 articles regarding 5 clinical topics. Again, the results from randomized trials were compared with those of observational cohort or case-controlled studies regarding the same intervention. The authors reported that “contrary to prevailing belief, the average results from well-designed observational studies did not systematically overestimate the magnitude of the associations between exposure and outcome as compared with the results of randomized, controlled trials on the same topic. Rather, the summary results of randomized, controlled trials and observational studies were remarkably similar.”12

On the basis of these studies, it appears that randomized control trials continue to serve as the gold standard in clinical research, but we must also recognize that circumstances often preclude the conduct of a randomized trial. As an example, consider a randomized trial of whether cigarette smoking is harmful, which, given the strong suspicion of harm, would be unethical in that patients cannot be randomized to smoke. Similarly, from the example before, a randomized trial of penicillin for pneumococcal pneumonia would be unethical because denying patients in the placebo group access to penicillin would exclude them from access to a drug that has “intra-ocular” efficacy. In circumstances like these, well-performed observational studies that are attentive to sources of bias can likely produce comparably reliable results to randomized trials.

In the end, of course, the interpretation of the study results requires the reader’s careful attention to potential sources of bias that can compromise study validity. The hope is that with Dr. Feinstein’s framework, you can be better equipped to think critically about study results that you review and to keenly ascertain whether there is any threat to internal or to external validity. Similarly, as you go on to design clinical trials yourselves, you can pay attention to these potential sources of bias that, if present, can compromise the reliability of the study conclusions internally or their applicability to patients outside of the study.

References
  1. Feinstein AR. Clinical Epidemiology: The Architecture of Clinical Research. Philadelphia, PA: WB Saunders; 1985.
  2. Thomas DP. Experiment versus authority: James Lind and Benjamin Rush. N Engl J Med 1969; 281:932–934.
  3. Downs JB, Klein EF Jr, Desautels D, Modell JH, Kirby RR. Intermittent mandatory ventilation: a new approach to weaning patients from mechanical ventilators. Chest 1973; 64:331–335.
  4. Brochard L, Rauss A, Benito S, et al. Comparison of three methods of gradual withdrawal from ventilatory support during weaning from mechanical ventilation. Am J Respir Crit Care Med 1994; 150:896–903.
  5. Chapman KR, Burdon JGW, Piitulainen E, et al; on behalf of the RAPID Trial Study Group. Intravenous augmentation treatment and lung density in severe 1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 386:360–368.
  6. Stoller JK, Wiedemann HP, Loke J, Snyder P, Virgulto J, Matthay RA. Terbutaline and diaphragm function in chronic obstructive pulmonary disease: a double-blind randomized clinical trial. Br J Dis Chest 1988; 82:242–250.
  7. Sehgal S, Velcheti V, Mukhopadhyay S, Stoller JK. Focal lung infiltrate complicating PD-1 inhibitor use: a new pattern of drug-associated lung toxicity? Respir Med Case Rep 2016; 19:118–120.
  8. Stoller JK, Moodie D, Schiavone WA, et al. Reduction of intrapulmonary shunt and resolution of digital clubbing associated with primary biliary cirrhosis after liver transplantation. Hepatology 1990; 11:54–58.
  9. Albert RK, Au DH, Blackford AL, et al; for the Long-Term Oxygen Treatment Trial Group. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med 2016; 375:1617–1627.
  10. Sacks HS, Chalmers TC, Smith H Jr. Sensitivity and specificity of clinical trials: randomized v historical controls. Arch Intern Med 1983; 143:753–755.
  11. Benson K, Hartz AJ. A comparison of observational studies and randomized, controlled trials. N Engl J Med 2000; 342:1878–1886.
  12. Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med 2000; 342:1887–1892.
References
  1. Feinstein AR. Clinical Epidemiology: The Architecture of Clinical Research. Philadelphia, PA: WB Saunders; 1985.
  2. Thomas DP. Experiment versus authority: James Lind and Benjamin Rush. N Engl J Med 1969; 281:932–934.
  3. Downs JB, Klein EF Jr, Desautels D, Modell JH, Kirby RR. Intermittent mandatory ventilation: a new approach to weaning patients from mechanical ventilators. Chest 1973; 64:331–335.
  4. Brochard L, Rauss A, Benito S, et al. Comparison of three methods of gradual withdrawal from ventilatory support during weaning from mechanical ventilation. Am J Respir Crit Care Med 1994; 150:896–903.
  5. Chapman KR, Burdon JGW, Piitulainen E, et al; on behalf of the RAPID Trial Study Group. Intravenous augmentation treatment and lung density in severe 1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 386:360–368.
  6. Stoller JK, Wiedemann HP, Loke J, Snyder P, Virgulto J, Matthay RA. Terbutaline and diaphragm function in chronic obstructive pulmonary disease: a double-blind randomized clinical trial. Br J Dis Chest 1988; 82:242–250.
  7. Sehgal S, Velcheti V, Mukhopadhyay S, Stoller JK. Focal lung infiltrate complicating PD-1 inhibitor use: a new pattern of drug-associated lung toxicity? Respir Med Case Rep 2016; 19:118–120.
  8. Stoller JK, Moodie D, Schiavone WA, et al. Reduction of intrapulmonary shunt and resolution of digital clubbing associated with primary biliary cirrhosis after liver transplantation. Hepatology 1990; 11:54–58.
  9. Albert RK, Au DH, Blackford AL, et al; for the Long-Term Oxygen Treatment Trial Group. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med 2016; 375:1617–1627.
  10. Sacks HS, Chalmers TC, Smith H Jr. Sensitivity and specificity of clinical trials: randomized v historical controls. Arch Intern Med 1983; 143:753–755.
  11. Benson K, Hartz AJ. A comparison of observational studies and randomized, controlled trials. N Engl J Med 2000; 342:1878–1886.
  12. Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med 2000; 342:1887–1892.
Page Number
e2-e9
Page Number
e2-e9
Publications
Publications
Article Type
Display Headline
The architecture of clinical research
Display Headline
The architecture of clinical research
Legacy Keywords
Alvan Feinstein, David Sackett, trial design, clinical research, bias, outcome measure, wash-out period, James Lind, scurvy, James Stoller
Legacy Keywords
Alvan Feinstein, David Sackett, trial design, clinical research, bias, outcome measure, wash-out period, James Lind, scurvy, James Stoller
Citation Override
Cleveland Clinic Journal of Medicine 2017 September;84(suppl 2):e2-e9
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Basics of study design: Practical considerations

Article Type
Changed
Mon, 10/01/2018 - 14:35
Display Headline
Basics of study design: Practical considerations
From the “Biostatistics and Epidemiology Lecture Series, Part 1”

INTRODUCTION

Basic research skills are not acquired from medical school but from a mentor.1,2 A mentor with experience in study design and technical writing can make a real difference in your career. Most good mentors have more ideas for studies than they have time for research, so they are willing to share and guide your course. Your daily clinical experience provides a wealth of ideas in the form of “why do we do it this way” or “what is the evidence for” or “how can we improve outcomes or cut cost?” Of course, just about every study you read in a medical journal has suggestions for further research in the discussion section. Finally, keep in mind that the creation of study ideas and in particular, hypotheses, is a mysterious process, as this quote indicates: “It is not possible, deliberately, to create ideas or to control their creation. What we can do deliberately is to prepare our minds.” 3 Remember that chance favors the prepared mind.

DEVELOPING THE STUDY IDEA

Often, the most difficult task for someone new to research is developing a practical study idea. This section will explain a detailed process for creating a formal research protocol. We will focus on two common sticking points: (1) finding a good idea, and (2) developing a good idea into a problem statement.

Novice researchers with little experience, no mentors, and short time frames are encouraged not to take on a clinical human study as the principle investigator. Instead, device evaluations are a low-cost, time-efficient alternative. Human studies in the form of a survey are also possible and are often exempt from full Institutional Review Board (IRB) review. Many human-like conditions can be simulated, as was done, for example, in the study of patient-ventilator synchrony.4,5 And if you have the aptitude, whole studies can be based on mathematical models and predictions, particularly with the vast array of computer tools now available.6,7 And don’t forget studies based on surveys.8

A structured approach

A structured approach for developing a formal research protocol.
Figure 1. A structured approach for developing a formal research protocol.

A formal research protocol is required for any human research. However, it is also recommended for all but the simplest investigations. Most of the new researchers I have mentored take a rather lax approach to developing the protocol, and most IRBs are more interested in protecting human rights than validating the study design. As a result, much time is wasted and sometimes an entire study has to be abandoned due to poor planning. Figure 1 illustrates a structured approach that helps to ensure success. It shows a 3-step, iterative process.

The first step is a process of expanding the scope of the project, primarily through literature review. Along the way you learn (or invent) appropriate terminology and become familiar with the current state of the research art on a broad topic. For example, let’s suppose you were interested in the factors that affect the duration of mechanical ventilation. The literature review might include topics such as weaning and patient-ventilator synchrony as well as ventilator-associated pneumonia. During this process, you might discover that the topic of synchrony is currently generating a lot of interest in the literature and generating a lot of questions or confusion. You then focus on expanding your knowledge in this area.

In the second step, you might develop a theoretical framework for understanding patient-ventilator synchrony that could include a mathematical model and, perhaps, an idea to include simulation to study the problem.

In the third step, you need to narrow the scope of the study to a manageable level that includes identifying measurable outcome variables, creating testable hypotheses, considering experimental designs, and evaluating the overall feasibility of the study. At this point, you may discover that you cannot measure the specific outcome variables indicated by your theoretical framework. In that case, you need to create a new framework for supporting your research. Alternatively, you may find that it is not possible to conduct the study you envision given your resources. In that case, it is back to step 1.

Eventually, this process will result in a well-planned research protocol that is ready for review. Keep in mind that many times a protocol needs to be refined after some initial experiments are conducted. For human studies, any changes to the protocol must be approved by the IRB.

The problem statement rubric

The most common problem I have seen novices struggle with is creating a meaningful problem statement and hypothesis. This is crucial because the problem statement sets the stage for the methods, the methods yield the results, and the results are analyzed in light of the original problem statement and hypotheses. To get past any writer’s block, I recommend that you start by just describing what you see happening and why you think it is important. For example, you might say, “Patients with acute lung injury often seem to be fighting the ventilator.” This is important because patient-ventilator asynchrony may lead to increased sedation levels and prolonged intensive care unit stays. Now you can more easily envision a specific purpose and testable hypothesis. For example, you could state that the purpose of this study is to determine the baseline rates of different kinds of patient-ventilator synchrony problems. The hypothesis is that the rate of dyssynchrony is correlated with duration of mechanical ventilation.

Here is an actual example of how a problem statement evolved from a vague notion to a testable hypothesis.

Original: The purpose of this study is to determine whether measures of ineffective cough in patients with stroke recently liberated from mechanical ventilation correlate with risk of extubation failure and reintubation.

Final: The purpose of this study is to test the hypothesis that use of CoughAssist device in the immediate post-extubation period by stroke patients reduces the rate of extubation failure and pneumonia.

The original statement is a run-on sentence that is vague and hard to follow. Once the actual treatment and outcome measures are in focus, then a clear hypothesis statement can be made. Notice that the hypothesis should be clear enough that the reader can anticipate the actual experimental measures and procedures to be described in the methods section of the protocol.

Here is another example:

Original: The purpose of this study is to evaluate a device that allows continuous electronic cuff pressure control.

Final: The purpose of this study is to test the hypothesis that the Pressure Eyes electronic cuff monitor will maintain constant endotracheal tube cuff pressures better than manual cuff inflation during mechanical ventilation.

The problem with the original statement is that “to evaluate” is vague. The final statement makes the outcome variable explicit and suggests what the experimental procedure will be.

This is a final example:

Original: Following cardiac/respiratory arrest, many patients are profoundly acidotic. Ventilator settings based on initial arterial blood gases may result in inappropriate hyperventilation when follow-up is delayed. The purpose of this study is to establish the frequency of this occurrence at a large academic institution and the feasibility of a quality improvement project.

Final: The primary purpose of this study is to evaluate the frequency of hyperventilation occurring post-arrest during the first 24 hours. A secondary purpose is to determine if this hyperventilation is associated with an initial diagnosis of acidosis.

Note that the original statement follows the rubric of telling us what is observed and why it is important. However, the actual problem statement derived from the observation is vague: what is “this occurrence” and is the study really to establish any kind of feasibility? The purpose is simply to evaluate the frequency of hyperventilation and determine if the condition is associated with acidosis.

 

 

EXAMPLES OF RESEARCH PROJECTS BY FELLOWS

The following are examples of well-written statements of study purpose from actual studies conducted by our fellows.

Device evaluation

Defining “Flow Starvation” in volume control mechanical ventilation.

  • The purpose of this study is to evaluate the relationship between the patient and ventilator inspiratory work of breathing to define the term “Flow Starvation.”

Auto-positive end expiratory pressure (auto-PEEP) during airway pressure release ventilation varies with the ventilator model.

  • The purpose of this study was to compare auto-PEEP levels, peak expiratory flows, and flow decay profiles among 4 common intensive care ventilators.

Patient study

Diaphragmatic electrical activity and extubation outcomes in newborn infants: an observational study.

  • The purpose of this study is to describe the electrical activity of the diaphragm before, during, and after extubation in a mixed-age cohort of preterm infants.

Comparison of predicted and measured carbon dioxide production for monitoring dead space fraction during mechanical ventilation.

  • The purpose of this pilot study was to compare dead space with tidal volume ratios calculated from estimated and measured values for carbon dioxide production.

Practice evaluation

Incidence of asynchronies during invasive mechanical ventilation in a medical intensive care unit.

  • The purpose of this study is to conduct a pilot investigation to determine the baseline incidence of various forms of patient-ventilator dyssynchrony during invasive mechanical ventilation.

Simulation training results in improved knowledge about intubation policies and procedures.

  • The purpose of this study was to develop and test a simulation-based rapid-sequence intubation curriculum for fellows in pulmonary and critical care training.

HOW TO SEARCH THE LITERATURE

After creating a problem statement, the next step in planning research is to search the literature. The 10th issue of Respiratory Care journal in 2009 was devoted to research. Here are the articles in that issue related to the literature search:

  • How to find the best evidence (search internet)9
  • How to read a scientific research paper10
  • How to read a case report (or teaching case of the month)11
  • How to read a review paper.12

I recommend that you read these papers.

Literature search resources

My best advice is to befriend your local librarian.13 These people seldom get the recognition they deserve as experts at finding information and even as co-investigators.14 In addition to personal help, some libraries offer training sessions on various useful skills.

PubMed

The Internet resource I use most often is PubMed (www.ncbi.nlm.nih.gov/pubmed). It offers free access to MEDLINE, which is the National Library of Medicine’s database of citations and abstracts in the fields of medicine, nursing, dentistry, veterinary medicine, health care systems, and preclinical sciences. There are links to full-text articles and other resources. The website provides a clinical queries search filters page as well as a special queries page. Using a feature called “My NCBI,” you can have automatic e-mailing of search updates and save records and filters for search results. Access the PubMed Quick Start Guide for frequently asked questions and tutorials.

SearchMedica.com

The SearchMedica website (www.searchmedica.co.uk) is free and intended for medical professionals. It provides answers for clinical questions. Searches return articles, abstracts, and recommended medical websites.

Synthetic databases

There is a class of websites called synthetic databases, which are essentially prefiltered records for particular topics. However, these sites are usually subscription-based, and the cost is relatively high. You should check with your medical library to get access. Their advantage is that often they provide the best evidence without extensive searches of standard, bibliographic databases. Examples include the Cochrane Database of Systematic Reviews (www.cochrane.org/evidence), the National Guideline Clearinghouse (www.guideline.gov), and UpToDate (www.uptodate.com). UpToDate claims to be the largest clinical community in the world dedicated to synthesized knowledge for clinicians and patients. It features the work of more than 6,000 expert clinician authors/reviewers on more than 10,000 topics in 23 medical specialties. The site offers graded recommendations based on the best medical evidence.

Portals

Portals are web pages that act as a starting point for using the web or web-based services. One popular example is ClinicalKey (www.clinicalkey.com/info), formerly called MD Consult, which offers books, journals, patient education materials, and images. Another popular portal is Ovid (ovid.com), offering books, journals, evidence-based medicine databases, and CINAHL (Cumulative Index to Nursing and Allied Health Literature).

Electronic journals

Many medical journals now have online databases of current and archived issues. Such sites may require membership to access the databases, so again, check with your medical library. Popular examples in pulmonary and critical care medicine include the following:

Electronic books

Amazon.com is a great database search engine for books on specific topics. It even finds out-of-print books. And you don’t have to buy the books, because now you can rent them. Sometimes, I find what I wanted by using the “Look Inside” feature for some books. Note that you can look for books at PubMed. Just change the search box from PubMed to Books on the PubMed home page. Of course, Google also has a book search feature. A great (subscription) resource for medical and technical books is Safari (https://www.safaribooksonline.com). Once again, your library may have a subscription.

General Internet resources

You probably already know about Google Scholar (scholar.google.com) and Wikipedia.com. Because of its open source nature, you should use Wikipedia with caution. However, I have found it to be a very good first step in finding technical information, particularly about mathematics, physics, and statistics.

 

 

Using reference management software

One of the most important things you can do to make your research life easier is to use some sort of reference management software. As described in Wikipedia, “Reference management software, citation management software or personal bibliographic management software is software for scholars and authors to use for recording and using bibliographic citations (references). Once a citation has been recorded, it can be used time and again in generating bibliographies, such as lists of references in scholarly books, articles, and essays.” I was late in adopting this technology, but now I am a firm believer. Most Internet reference sources offer the ability to download citations to your reference management software. Downloading automatically places the citation into a searchable database on your computer with backup to the Internet. In addition, you can get the reference manager software to find a PDF version of the manuscript and store it with the citation on your computer (and/or in the Cloud) automatically.

But the most powerful feature of such software is its ability to add or subtract and rearrange the order of references in your manuscripts as you are writing, using seamless integration with Microsoft Word. The references can be automatically formatted using just about any journal’s style. This is a great time saver for resubmitting manuscripts to different journals. If you are still numbering references by hand (God forbid) or even using the Insert Endnote feature in Word (deficient when using multiple occurrences of the same reference), your life will be much easier if you take the time to start using reference management software.

The most popular commercial software is probably EndNote (endnote.com). A really good free software system with about the same functionality as Zotero (zotero.com). Search for “comparison of reference management software” in Wikipedia. You can find tutorials on software packages in YouTube.

STUDY DESIGN

Schematic of pre-experimental research designs.
Figure 2. Schematic of pre-experimental research designs.

When designing the experiment, note that there are many different approaches, each with their advantages and disadvantages. A full treatment of this topic is beyond the scope of this article. Suffice it to say that pre-experimental designs (Figure 2) are considered to generate weak evidence. But they are quick and easy and might be appropriate for pilot studies.

Schematic of a quasi-experimental research design.
Figure 3. Schematic of a quasi-experimental research design.

Quasi-experimental designs (Figure 3) generate a higher level of evidence. Such a design might be appropriate when you are stuck with collecting a convenience sample, rather than being able to use a full randomized assignment of study subjects.

The randomized controlled study design.
Figure 4. The randomized controlled study design.

The fully randomized design (Figure 4) generates the highest level of evidence. This is because if the sample size is large enough, the unknown and uncontrollable sources of bias are evenly distributed between the study groups. 

BASIC MEASUREMENT METHODS

If your research involves physical measurements, you need to be familiar with the devices considered to be the gold standards. In cardiopulmonary research, most measurements involve pressure volume, flow, and gas concentration. You need to know which devices are appropriate for static vs dynamic measurements of these variables. In addition, you need to understand issues related to systematic and random measurement errors and how these errors are managed through calibration and calibration verification. I recommend these two textbooks:

Principles and Practice of Intensive Care Monitoring 1st Edition by Martin J. Tobin MD.

  • This book is out of print, but if you can find a used copy or one in a library, it describes just about every kind of physiologic measurement used in clinical medicine.

Medical Instrumentation: Application and Design 4th Edition by John G. Webster.

  • This book is readily available and reasonably priced. It is a more technical book describing medical instrumentation and measurement principles. It is a standard textbook for biometrical engineers.

STATISTICS FOR THE UNINTERESTED

I know what you are thinking: I hate statistics. Look at the book Essential Biostatistics: A Nonmathematical Approach.15 It is a short, inexpensive paperback book that is easy to read. The author does a great job of explaining why we use statistics rather than getting bogged down explaining how we calculate them. After all, novice researchers usually seek the help of a professional statistician to do the heavy lifting.

My book, Handbook for Health Care Research,16 covers most of the statistical procedures you will encounter in medical research and gives examples of how to use a popular tactical software package called SigmaPlot. By the way, I strongly suggest that you consult a statistician early in your study design phase to avoid the disappointment of finding out later that your results are uninterpretable. For an in-depth treatment of the subject, I recommend How to Report Statistics in Medicine.17

Statistical bare essentials

Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data.
Figure 5. Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data. COPD = chronic obstructive pulmonary disease; PaCOs = partial pressure of carbon dioxide, artery; PS = pressure support; RDS = respiratory distress syndrome; SIMV = synchronized intermittent mandatory ventilation

To do research or even just to understand published research reports, you must have at least a minimal skill set. The necessary skills include understanding some basic terminology, if only to be able to communicate with a statistician consultant. Important terms include levels of measurement (nominal, ordinal, continuous), accuracy, precision, measures of central tendency (mean, median, mode), measures of variability (variance, standard deviation, coefficient of variation), and percentile. The first step in analyzing your results is usually to represent it graphically. That means you should be able to use a spreadsheet to make simple graphs (Figure 5).

Example flowchart for selecting the appropriate statistical test.
Figure 6. Example flowchart for selecting the appropriate statistical test. ANOVA = analysis of variance

You should also know the basics of inferential statistics (ie, hypothesis testing). For example, you need to know the difference between parametric and non-parametric tests. You should be able to explain correlation and regression and know when to use Chi-squared vs a Fisher exact test. You should know that when comparing two mean values, you typically use the Student’s t test (and know when to use paired vs unpaired versions of the test). When comparing more than 2 mean values, you use analysis of variance methods (ANOVA). You can teach yourself these concepts from a book,16 but even an introductory college level course on statistics will be immensely helpful. Most statistics textbooks provide some sort of map to guide your selection of the appropriate statistical test (Figure 6), and there are good articles in medical journals.

You can learn a lot simply by reading the Methods section of research articles. Authors will often describe the statistical tests used and why they were used. But be aware that a certain percentage of papers get published with the wrong statistics.18 

One of the underlying assumptions of most parametric statistical methods is that the data may be adequately described by a normal or Gaussian distribution. This assumption needs to be verified before selecting a statistical test. The common test for data normality is the Kolmogorov-Smirnov test. The following text from a methods section describes 2 very common procedures—the Student’s t test for comparing 2 mean values and the one-way ANOVA for comparing more than 2 mean values.19

“Normal distribution of data was verified using the Kolmogorov-Smirnov test. Body weights between groups were compared using one-way ANOVA for repeated measures to investigate temporal differences. At each time point, all data were analyzed using one-way ANOVA to compare PCV and VCV groups. Tukey’s post hoc analyses were performed when significant time effects were detected within groups, and Student’s t test was used to investigate differences between groups. Data were analyzed using commercial software and values were presented as mean ± SD. A P value < .05 was considered statistically significant.” 

 

 

Estimating sample size and power analysis

One very important consideration in any study is the required number of study subjects for meaningful statistical conclusions. In other words, how big should the sample size be? Sample size is important because it affects the feasibility of the study and the reliability of the conclusions in terms of statistical power. The necessary sample size depends on 2 basic factors. One factor is the variability of the data (often expressed as the standard deviation). The other factor is the effect size, meaning, for example, how big of a difference between mean values you want to detect. In general, the bigger the variability and the smaller the difference, the bigger the sample size required.

As the above equation shows, the effect size is expressed, in general, as a mean difference divided by a standard deviation. In the first case, the numerator represents the difference between the sample mean and the assumed population mean. In the denominator, SD is the standard deviation of the sample (used to estimate the standard deviation of the population). In the second case, the numerator represents the difference between the mean values of 2 samples and the denominator is the pooled standard deviation of the 2 samples.

In order to understand the issues involved with selecting sample size, we need to first understand the types of errors that can be made in any type of decision. Suppose our research goal is to make a decision about whether a new treatment results in a clinical difference (improvement). The results of our statistical test are dichotomous—we decide either yes there is a significant difference or no there isn’t. The truth, which we may never know, is that in reality, the difference exists or it doesn’t.

Types of errors in statistical decision making.
Figure 7. Types of errors in statistical decision making.

As Figure 7 shows, the result of our decision making is that there are 2 ways to be right and 2 ways to be wrong. If we decide there is a difference (eg, our statistical tests yields P ≤ .05) but in realty there is not a difference, then we make what is called a type I error. On the other hand, if we conclude that there is not a difference (ie, our statistical test yields P > .05) but in reality there is a difference that we did not detect, then we have made a type II error.

Probabilities associated with type I and type II errors.
Figure 8. Probabilities associated with type I and type II errors.

The associated math is shown in Figure 8. The probability of making a type I error is called alpha. By convention in medicine, we set our rejection criterion to alpha = 0.05. In other words, we would reject the null hypothesis (that there is no difference) anytime our statistical test yields a P value less than alpha. The probability of making a type II error is called beta. For historical reasons, the probability of not making a type II error is called the statistical power of the test and is equal to 1 minus beta. Power is affected by sample size: the larger the sample the larger the power. Most researchers, by convention, keep the sample size large enough to keep power above 0.80.

Nomogram for calculating power and sample size
Figure 9. Nomogram for calculating power and sample size.

Figure 9 is a nomogram that brings all these ideas together. The red line shows that for your study, given the desired effect size (0.8), if you collected samples from the 30 patients you planned on then the power would be unacceptable at 0.60, indicating a high probability of a false negative decision if the P value comes out greater than .50. The solution is to increase the sample size to about 50 (or more), as indicated by the blue line. From this nomogram we can generalize to say that when you want to detect a small effect with data that have high variability, you need a large sample size to provide acceptable power.

The text below is an example of a power analysis presented in the methods section of a published study.20 Note that the authors give their reasoning for the sample size they selected. This kind of explanation may inform your study design. But what if you don’t know the variability of the data you want to collect? In that case, you need to collect some pilot data and calculate from that an appropriate sample size for a subsequent study.

A prospective power calculation indicated that a sample size of 25 per group was required to achieve 80% power based on an effect size of probability of 0.24 that an observation in the PRVCa group is less than an observation in the ASV group using the Mann-Whitney tests, an alpha of 0.05 (two-tailed) and a 20% dropout.

JUDGING FEASIBILITY

Once you have a draft of your study design, including the estimated sample size, it is time to judge the overall feasibility of the study before committing to it.

Factors to consider when judging the feasibility of a new study
Table 1 shows some of the most important factors in judging feasibility. The first question is whether the outcome will be worth the resources needed to complete the study, implying that you must define costs and benefits. Second, assure yourself that you can both define and measure the outcome variables of interest, which can be a challenge in psychological studies and even in quality improvement projects. Next consider the time constraints, which are affected mainly by the sample size and the time needed to observe all the individuals in that sample. Naturally, if you are studying a rare disorder, the time needed to collect even a modest sample size may make the project impractical.

Every study has associated costs. Those costs and the sources of funding must be identified. Don’t forget costs for consultants, particularly if you need statistical consultation.

Finally, consider your level of experience. If you are contemplating your first study, a human clinical trial might not be the best choice, given the complexity of such a project. Studies such as a meta-analysis or mathematical simulation require special training beyond basic research procedures, and should be avoided.

References
  1. Tobin MJ. Mentoring: seven roles and some specifics. Am J Respir Crit Care Med 2004; 170:114–117.
  2. Chatburn RL. Advancing beyond the average: the importance of mentoring in professional achievement. Respir Care 2004; 49:304–308.
  3. Beveridge WIB. The Art of Scientific Investigation. New York, NY: WW Norton & Company; 1950.
  4. Chatburn RL, Mireles-Cabodevila E, Sasidhar M. Tidal volume measurement error in pressure control modes of mechanical ventilation: a model study. Comput Biol Med 2016; 75:235–242.
  5. Mireles-Cabodevila E, Chatburn RL. Work of breathing in adaptive pressure control continuous mandatory ventilation. Respir Care 2009; 54:1467–1472.
  6. Chatburn RL, Ford RM. Procedure to normalize data for benchmarking. Respir Care 2006; 51:145–157.
  7. Bou-Khalil P, Zeineldine S, Chatburn R, et al. Prediction of inspired oxygen fraction for targeted arterial oxygen tension following open heart surgery in non-smoking and smoking patients. J Clin Monit Comput 2016. https://doi.org/10.1007/s10877-016-9941-6.
  8. Mireles-Cabodevila E, Diaz-Guzman E, Arroliga AC, Chatburn RL. Human versus computer controlled selection of ventilator settings: an evaluation of adaptive support ventilation and mid-frequency ventilation. Crit Care Res Pract 2012; 2012:204314.
  9. Chatburn RL. How to find the best evidence. Respir Care 2009; 54:1360–1365.
  10. Durbin CG Jr. How to read a scientific research paper. Respir Care 2009; 54:1366–1371.
  11. Pierson DJ. How to read a case report (or teaching case of the month). Respir Care 2009; 54:1372–1378.
  12. Callcut RA, Branson RD. How to read a review paper. Respir Care 2009; 54:1379–1385.
  13. Eresuma E, Lake E. How do I find the evidence? Find your librarian—stat! Orthop Nurs 2016; 35:421–423.
  14. Janke R, Rush KL. The academic librarian as co-investigator on an interprofessional primary research team: a case study. Health Info Libr J 2014; 31:116–122.
  15. Motulsky H. Essential Biostatistics: A Nonmathematical Approach. New York, NY: Oxford University Press; 2016.
  16. Chatburn RL. Handbook for Health Care Research. 2nd ed. Sudbury, MA: Jones and Bartlett Publishers; 2011.
  17. Lang TA, Secic M. How to Report Statistics in Medicine. 2nd ed. Philadelphia, PA: American College of Physicians; 2006.
  18. Prescott RJ, Civil I. Lies, damn lies and statistics: errors and omission in papers submitted to INJURY 2010–2012. Injury 2013; 44:6–11.
  19. Fantoni DT, Ida KK, Lopes TF, Otsuki DA, Auler JO Jr, Ambrosio AM. A comparison of the cardiopulmonary effects of pressure controlled ventilation and volume controlled ventilation in healthy anesthetized dogs. J Vet Emerg Crit Care (San Antonio) 2016; 26:524–530.
  20. Gruber PC, Gomersall CD, Leung P, et al. Randomized controlled trial comparing adaptive-support ventilation with pressure-regulated volume-controlled ventilation with automode in weaning patients after cardiac surgery. Anesthesiology 2008; 109:81–87.
Article PDF
Author and Disclosure Information

Robert L. Chatburn, MHHS, RRT-NPS, FAARC
Clinical Research Manager, Respiratory Institute; Director Simulation Fellowship, Education Institute; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Correspondence: Robert L. Chatburn, MHHS, RRT-NPS, FAARC, Clinical Research Manager, Respiratory Institute, M56, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Mr. Chatburn’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Mr. Chatburn presented his lecture on September 6, 2016, at Cleveland Clinic.

Mr. Chatburn reported no financial interests or relationships that pose a potential conflict of interest with this article.

Publications
Page Number
e10-e19
Legacy Keywords
study design, statistics, hypothesis testing, sample size, power, Robert Chatburn
Author and Disclosure Information

Robert L. Chatburn, MHHS, RRT-NPS, FAARC
Clinical Research Manager, Respiratory Institute; Director Simulation Fellowship, Education Institute; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Correspondence: Robert L. Chatburn, MHHS, RRT-NPS, FAARC, Clinical Research Manager, Respiratory Institute, M56, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Mr. Chatburn’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Mr. Chatburn presented his lecture on September 6, 2016, at Cleveland Clinic.

Mr. Chatburn reported no financial interests or relationships that pose a potential conflict of interest with this article.

Author and Disclosure Information

Robert L. Chatburn, MHHS, RRT-NPS, FAARC
Clinical Research Manager, Respiratory Institute; Director Simulation Fellowship, Education Institute; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Correspondence: Robert L. Chatburn, MHHS, RRT-NPS, FAARC, Clinical Research Manager, Respiratory Institute, M56, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

This article is based on Mr. Chatburn’s presentation at the “Biostatistics and Epidemiology” lecture series created by Aanchal Kapoor, MD, Critical Care Medicine, Cleveland Clinic. Mr. Chatburn presented his lecture on September 6, 2016, at Cleveland Clinic.

Mr. Chatburn reported no financial interests or relationships that pose a potential conflict of interest with this article.

Article PDF
Article PDF
Related Articles
From the “Biostatistics and Epidemiology Lecture Series, Part 1”
From the “Biostatistics and Epidemiology Lecture Series, Part 1”

INTRODUCTION

Basic research skills are not acquired from medical school but from a mentor.1,2 A mentor with experience in study design and technical writing can make a real difference in your career. Most good mentors have more ideas for studies than they have time for research, so they are willing to share and guide your course. Your daily clinical experience provides a wealth of ideas in the form of “why do we do it this way” or “what is the evidence for” or “how can we improve outcomes or cut cost?” Of course, just about every study you read in a medical journal has suggestions for further research in the discussion section. Finally, keep in mind that the creation of study ideas and in particular, hypotheses, is a mysterious process, as this quote indicates: “It is not possible, deliberately, to create ideas or to control their creation. What we can do deliberately is to prepare our minds.” 3 Remember that chance favors the prepared mind.

DEVELOPING THE STUDY IDEA

Often, the most difficult task for someone new to research is developing a practical study idea. This section will explain a detailed process for creating a formal research protocol. We will focus on two common sticking points: (1) finding a good idea, and (2) developing a good idea into a problem statement.

Novice researchers with little experience, no mentors, and short time frames are encouraged not to take on a clinical human study as the principle investigator. Instead, device evaluations are a low-cost, time-efficient alternative. Human studies in the form of a survey are also possible and are often exempt from full Institutional Review Board (IRB) review. Many human-like conditions can be simulated, as was done, for example, in the study of patient-ventilator synchrony.4,5 And if you have the aptitude, whole studies can be based on mathematical models and predictions, particularly with the vast array of computer tools now available.6,7 And don’t forget studies based on surveys.8

A structured approach

A structured approach for developing a formal research protocol.
Figure 1. A structured approach for developing a formal research protocol.

A formal research protocol is required for any human research. However, it is also recommended for all but the simplest investigations. Most of the new researchers I have mentored take a rather lax approach to developing the protocol, and most IRBs are more interested in protecting human rights than validating the study design. As a result, much time is wasted and sometimes an entire study has to be abandoned due to poor planning. Figure 1 illustrates a structured approach that helps to ensure success. It shows a 3-step, iterative process.

The first step is a process of expanding the scope of the project, primarily through literature review. Along the way you learn (or invent) appropriate terminology and become familiar with the current state of the research art on a broad topic. For example, let’s suppose you were interested in the factors that affect the duration of mechanical ventilation. The literature review might include topics such as weaning and patient-ventilator synchrony as well as ventilator-associated pneumonia. During this process, you might discover that the topic of synchrony is currently generating a lot of interest in the literature and generating a lot of questions or confusion. You then focus on expanding your knowledge in this area.

In the second step, you might develop a theoretical framework for understanding patient-ventilator synchrony that could include a mathematical model and, perhaps, an idea to include simulation to study the problem.

In the third step, you need to narrow the scope of the study to a manageable level that includes identifying measurable outcome variables, creating testable hypotheses, considering experimental designs, and evaluating the overall feasibility of the study. At this point, you may discover that you cannot measure the specific outcome variables indicated by your theoretical framework. In that case, you need to create a new framework for supporting your research. Alternatively, you may find that it is not possible to conduct the study you envision given your resources. In that case, it is back to step 1.

Eventually, this process will result in a well-planned research protocol that is ready for review. Keep in mind that many times a protocol needs to be refined after some initial experiments are conducted. For human studies, any changes to the protocol must be approved by the IRB.

The problem statement rubric

The most common problem I have seen novices struggle with is creating a meaningful problem statement and hypothesis. This is crucial because the problem statement sets the stage for the methods, the methods yield the results, and the results are analyzed in light of the original problem statement and hypotheses. To get past any writer’s block, I recommend that you start by just describing what you see happening and why you think it is important. For example, you might say, “Patients with acute lung injury often seem to be fighting the ventilator.” This is important because patient-ventilator asynchrony may lead to increased sedation levels and prolonged intensive care unit stays. Now you can more easily envision a specific purpose and testable hypothesis. For example, you could state that the purpose of this study is to determine the baseline rates of different kinds of patient-ventilator synchrony problems. The hypothesis is that the rate of dyssynchrony is correlated with duration of mechanical ventilation.

Here is an actual example of how a problem statement evolved from a vague notion to a testable hypothesis.

Original: The purpose of this study is to determine whether measures of ineffective cough in patients with stroke recently liberated from mechanical ventilation correlate with risk of extubation failure and reintubation.

Final: The purpose of this study is to test the hypothesis that use of CoughAssist device in the immediate post-extubation period by stroke patients reduces the rate of extubation failure and pneumonia.

The original statement is a run-on sentence that is vague and hard to follow. Once the actual treatment and outcome measures are in focus, then a clear hypothesis statement can be made. Notice that the hypothesis should be clear enough that the reader can anticipate the actual experimental measures and procedures to be described in the methods section of the protocol.

Here is another example:

Original: The purpose of this study is to evaluate a device that allows continuous electronic cuff pressure control.

Final: The purpose of this study is to test the hypothesis that the Pressure Eyes electronic cuff monitor will maintain constant endotracheal tube cuff pressures better than manual cuff inflation during mechanical ventilation.

The problem with the original statement is that “to evaluate” is vague. The final statement makes the outcome variable explicit and suggests what the experimental procedure will be.

This is a final example:

Original: Following cardiac/respiratory arrest, many patients are profoundly acidotic. Ventilator settings based on initial arterial blood gases may result in inappropriate hyperventilation when follow-up is delayed. The purpose of this study is to establish the frequency of this occurrence at a large academic institution and the feasibility of a quality improvement project.

Final: The primary purpose of this study is to evaluate the frequency of hyperventilation occurring post-arrest during the first 24 hours. A secondary purpose is to determine if this hyperventilation is associated with an initial diagnosis of acidosis.

Note that the original statement follows the rubric of telling us what is observed and why it is important. However, the actual problem statement derived from the observation is vague: what is “this occurrence” and is the study really to establish any kind of feasibility? The purpose is simply to evaluate the frequency of hyperventilation and determine if the condition is associated with acidosis.

 

 

EXAMPLES OF RESEARCH PROJECTS BY FELLOWS

The following are examples of well-written statements of study purpose from actual studies conducted by our fellows.

Device evaluation

Defining “Flow Starvation” in volume control mechanical ventilation.

  • The purpose of this study is to evaluate the relationship between the patient and ventilator inspiratory work of breathing to define the term “Flow Starvation.”

Auto-positive end expiratory pressure (auto-PEEP) during airway pressure release ventilation varies with the ventilator model.

  • The purpose of this study was to compare auto-PEEP levels, peak expiratory flows, and flow decay profiles among 4 common intensive care ventilators.

Patient study

Diaphragmatic electrical activity and extubation outcomes in newborn infants: an observational study.

  • The purpose of this study is to describe the electrical activity of the diaphragm before, during, and after extubation in a mixed-age cohort of preterm infants.

Comparison of predicted and measured carbon dioxide production for monitoring dead space fraction during mechanical ventilation.

  • The purpose of this pilot study was to compare dead space with tidal volume ratios calculated from estimated and measured values for carbon dioxide production.

Practice evaluation

Incidence of asynchronies during invasive mechanical ventilation in a medical intensive care unit.

  • The purpose of this study is to conduct a pilot investigation to determine the baseline incidence of various forms of patient-ventilator dyssynchrony during invasive mechanical ventilation.

Simulation training results in improved knowledge about intubation policies and procedures.

  • The purpose of this study was to develop and test a simulation-based rapid-sequence intubation curriculum for fellows in pulmonary and critical care training.

HOW TO SEARCH THE LITERATURE

After creating a problem statement, the next step in planning research is to search the literature. The 10th issue of Respiratory Care journal in 2009 was devoted to research. Here are the articles in that issue related to the literature search:

  • How to find the best evidence (search internet)9
  • How to read a scientific research paper10
  • How to read a case report (or teaching case of the month)11
  • How to read a review paper.12

I recommend that you read these papers.

Literature search resources

My best advice is to befriend your local librarian.13 These people seldom get the recognition they deserve as experts at finding information and even as co-investigators.14 In addition to personal help, some libraries offer training sessions on various useful skills.

PubMed

The Internet resource I use most often is PubMed (www.ncbi.nlm.nih.gov/pubmed). It offers free access to MEDLINE, which is the National Library of Medicine’s database of citations and abstracts in the fields of medicine, nursing, dentistry, veterinary medicine, health care systems, and preclinical sciences. There are links to full-text articles and other resources. The website provides a clinical queries search filters page as well as a special queries page. Using a feature called “My NCBI,” you can have automatic e-mailing of search updates and save records and filters for search results. Access the PubMed Quick Start Guide for frequently asked questions and tutorials.

SearchMedica.com

The SearchMedica website (www.searchmedica.co.uk) is free and intended for medical professionals. It provides answers for clinical questions. Searches return articles, abstracts, and recommended medical websites.

Synthetic databases

There is a class of websites called synthetic databases, which are essentially prefiltered records for particular topics. However, these sites are usually subscription-based, and the cost is relatively high. You should check with your medical library to get access. Their advantage is that often they provide the best evidence without extensive searches of standard, bibliographic databases. Examples include the Cochrane Database of Systematic Reviews (www.cochrane.org/evidence), the National Guideline Clearinghouse (www.guideline.gov), and UpToDate (www.uptodate.com). UpToDate claims to be the largest clinical community in the world dedicated to synthesized knowledge for clinicians and patients. It features the work of more than 6,000 expert clinician authors/reviewers on more than 10,000 topics in 23 medical specialties. The site offers graded recommendations based on the best medical evidence.

Portals

Portals are web pages that act as a starting point for using the web or web-based services. One popular example is ClinicalKey (www.clinicalkey.com/info), formerly called MD Consult, which offers books, journals, patient education materials, and images. Another popular portal is Ovid (ovid.com), offering books, journals, evidence-based medicine databases, and CINAHL (Cumulative Index to Nursing and Allied Health Literature).

Electronic journals

Many medical journals now have online databases of current and archived issues. Such sites may require membership to access the databases, so again, check with your medical library. Popular examples in pulmonary and critical care medicine include the following:

Electronic books

Amazon.com is a great database search engine for books on specific topics. It even finds out-of-print books. And you don’t have to buy the books, because now you can rent them. Sometimes, I find what I wanted by using the “Look Inside” feature for some books. Note that you can look for books at PubMed. Just change the search box from PubMed to Books on the PubMed home page. Of course, Google also has a book search feature. A great (subscription) resource for medical and technical books is Safari (https://www.safaribooksonline.com). Once again, your library may have a subscription.

General Internet resources

You probably already know about Google Scholar (scholar.google.com) and Wikipedia.com. Because of its open source nature, you should use Wikipedia with caution. However, I have found it to be a very good first step in finding technical information, particularly about mathematics, physics, and statistics.

 

 

Using reference management software

One of the most important things you can do to make your research life easier is to use some sort of reference management software. As described in Wikipedia, “Reference management software, citation management software or personal bibliographic management software is software for scholars and authors to use for recording and using bibliographic citations (references). Once a citation has been recorded, it can be used time and again in generating bibliographies, such as lists of references in scholarly books, articles, and essays.” I was late in adopting this technology, but now I am a firm believer. Most Internet reference sources offer the ability to download citations to your reference management software. Downloading automatically places the citation into a searchable database on your computer with backup to the Internet. In addition, you can get the reference manager software to find a PDF version of the manuscript and store it with the citation on your computer (and/or in the Cloud) automatically.

But the most powerful feature of such software is its ability to add or subtract and rearrange the order of references in your manuscripts as you are writing, using seamless integration with Microsoft Word. The references can be automatically formatted using just about any journal’s style. This is a great time saver for resubmitting manuscripts to different journals. If you are still numbering references by hand (God forbid) or even using the Insert Endnote feature in Word (deficient when using multiple occurrences of the same reference), your life will be much easier if you take the time to start using reference management software.

The most popular commercial software is probably EndNote (endnote.com). A really good free software system with about the same functionality as Zotero (zotero.com). Search for “comparison of reference management software” in Wikipedia. You can find tutorials on software packages in YouTube.

STUDY DESIGN

Schematic of pre-experimental research designs.
Figure 2. Schematic of pre-experimental research designs.

When designing the experiment, note that there are many different approaches, each with their advantages and disadvantages. A full treatment of this topic is beyond the scope of this article. Suffice it to say that pre-experimental designs (Figure 2) are considered to generate weak evidence. But they are quick and easy and might be appropriate for pilot studies.

Schematic of a quasi-experimental research design.
Figure 3. Schematic of a quasi-experimental research design.

Quasi-experimental designs (Figure 3) generate a higher level of evidence. Such a design might be appropriate when you are stuck with collecting a convenience sample, rather than being able to use a full randomized assignment of study subjects.

The randomized controlled study design.
Figure 4. The randomized controlled study design.

The fully randomized design (Figure 4) generates the highest level of evidence. This is because if the sample size is large enough, the unknown and uncontrollable sources of bias are evenly distributed between the study groups. 

BASIC MEASUREMENT METHODS

If your research involves physical measurements, you need to be familiar with the devices considered to be the gold standards. In cardiopulmonary research, most measurements involve pressure volume, flow, and gas concentration. You need to know which devices are appropriate for static vs dynamic measurements of these variables. In addition, you need to understand issues related to systematic and random measurement errors and how these errors are managed through calibration and calibration verification. I recommend these two textbooks:

Principles and Practice of Intensive Care Monitoring 1st Edition by Martin J. Tobin MD.

  • This book is out of print, but if you can find a used copy or one in a library, it describes just about every kind of physiologic measurement used in clinical medicine.

Medical Instrumentation: Application and Design 4th Edition by John G. Webster.

  • This book is readily available and reasonably priced. It is a more technical book describing medical instrumentation and measurement principles. It is a standard textbook for biometrical engineers.

STATISTICS FOR THE UNINTERESTED

I know what you are thinking: I hate statistics. Look at the book Essential Biostatistics: A Nonmathematical Approach.15 It is a short, inexpensive paperback book that is easy to read. The author does a great job of explaining why we use statistics rather than getting bogged down explaining how we calculate them. After all, novice researchers usually seek the help of a professional statistician to do the heavy lifting.

My book, Handbook for Health Care Research,16 covers most of the statistical procedures you will encounter in medical research and gives examples of how to use a popular tactical software package called SigmaPlot. By the way, I strongly suggest that you consult a statistician early in your study design phase to avoid the disappointment of finding out later that your results are uninterpretable. For an in-depth treatment of the subject, I recommend How to Report Statistics in Medicine.17

Statistical bare essentials

Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data.
Figure 5. Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data. COPD = chronic obstructive pulmonary disease; PaCOs = partial pressure of carbon dioxide, artery; PS = pressure support; RDS = respiratory distress syndrome; SIMV = synchronized intermittent mandatory ventilation

To do research or even just to understand published research reports, you must have at least a minimal skill set. The necessary skills include understanding some basic terminology, if only to be able to communicate with a statistician consultant. Important terms include levels of measurement (nominal, ordinal, continuous), accuracy, precision, measures of central tendency (mean, median, mode), measures of variability (variance, standard deviation, coefficient of variation), and percentile. The first step in analyzing your results is usually to represent it graphically. That means you should be able to use a spreadsheet to make simple graphs (Figure 5).

Example flowchart for selecting the appropriate statistical test.
Figure 6. Example flowchart for selecting the appropriate statistical test. ANOVA = analysis of variance

You should also know the basics of inferential statistics (ie, hypothesis testing). For example, you need to know the difference between parametric and non-parametric tests. You should be able to explain correlation and regression and know when to use Chi-squared vs a Fisher exact test. You should know that when comparing two mean values, you typically use the Student’s t test (and know when to use paired vs unpaired versions of the test). When comparing more than 2 mean values, you use analysis of variance methods (ANOVA). You can teach yourself these concepts from a book,16 but even an introductory college level course on statistics will be immensely helpful. Most statistics textbooks provide some sort of map to guide your selection of the appropriate statistical test (Figure 6), and there are good articles in medical journals.

You can learn a lot simply by reading the Methods section of research articles. Authors will often describe the statistical tests used and why they were used. But be aware that a certain percentage of papers get published with the wrong statistics.18 

One of the underlying assumptions of most parametric statistical methods is that the data may be adequately described by a normal or Gaussian distribution. This assumption needs to be verified before selecting a statistical test. The common test for data normality is the Kolmogorov-Smirnov test. The following text from a methods section describes 2 very common procedures—the Student’s t test for comparing 2 mean values and the one-way ANOVA for comparing more than 2 mean values.19

“Normal distribution of data was verified using the Kolmogorov-Smirnov test. Body weights between groups were compared using one-way ANOVA for repeated measures to investigate temporal differences. At each time point, all data were analyzed using one-way ANOVA to compare PCV and VCV groups. Tukey’s post hoc analyses were performed when significant time effects were detected within groups, and Student’s t test was used to investigate differences between groups. Data were analyzed using commercial software and values were presented as mean ± SD. A P value < .05 was considered statistically significant.” 

 

 

Estimating sample size and power analysis

One very important consideration in any study is the required number of study subjects for meaningful statistical conclusions. In other words, how big should the sample size be? Sample size is important because it affects the feasibility of the study and the reliability of the conclusions in terms of statistical power. The necessary sample size depends on 2 basic factors. One factor is the variability of the data (often expressed as the standard deviation). The other factor is the effect size, meaning, for example, how big of a difference between mean values you want to detect. In general, the bigger the variability and the smaller the difference, the bigger the sample size required.

As the above equation shows, the effect size is expressed, in general, as a mean difference divided by a standard deviation. In the first case, the numerator represents the difference between the sample mean and the assumed population mean. In the denominator, SD is the standard deviation of the sample (used to estimate the standard deviation of the population). In the second case, the numerator represents the difference between the mean values of 2 samples and the denominator is the pooled standard deviation of the 2 samples.

In order to understand the issues involved with selecting sample size, we need to first understand the types of errors that can be made in any type of decision. Suppose our research goal is to make a decision about whether a new treatment results in a clinical difference (improvement). The results of our statistical test are dichotomous—we decide either yes there is a significant difference or no there isn’t. The truth, which we may never know, is that in reality, the difference exists or it doesn’t.

Types of errors in statistical decision making.
Figure 7. Types of errors in statistical decision making.

As Figure 7 shows, the result of our decision making is that there are 2 ways to be right and 2 ways to be wrong. If we decide there is a difference (eg, our statistical tests yields P ≤ .05) but in realty there is not a difference, then we make what is called a type I error. On the other hand, if we conclude that there is not a difference (ie, our statistical test yields P > .05) but in reality there is a difference that we did not detect, then we have made a type II error.

Probabilities associated with type I and type II errors.
Figure 8. Probabilities associated with type I and type II errors.

The associated math is shown in Figure 8. The probability of making a type I error is called alpha. By convention in medicine, we set our rejection criterion to alpha = 0.05. In other words, we would reject the null hypothesis (that there is no difference) anytime our statistical test yields a P value less than alpha. The probability of making a type II error is called beta. For historical reasons, the probability of not making a type II error is called the statistical power of the test and is equal to 1 minus beta. Power is affected by sample size: the larger the sample the larger the power. Most researchers, by convention, keep the sample size large enough to keep power above 0.80.

Nomogram for calculating power and sample size
Figure 9. Nomogram for calculating power and sample size.

Figure 9 is a nomogram that brings all these ideas together. The red line shows that for your study, given the desired effect size (0.8), if you collected samples from the 30 patients you planned on then the power would be unacceptable at 0.60, indicating a high probability of a false negative decision if the P value comes out greater than .50. The solution is to increase the sample size to about 50 (or more), as indicated by the blue line. From this nomogram we can generalize to say that when you want to detect a small effect with data that have high variability, you need a large sample size to provide acceptable power.

The text below is an example of a power analysis presented in the methods section of a published study.20 Note that the authors give their reasoning for the sample size they selected. This kind of explanation may inform your study design. But what if you don’t know the variability of the data you want to collect? In that case, you need to collect some pilot data and calculate from that an appropriate sample size for a subsequent study.

A prospective power calculation indicated that a sample size of 25 per group was required to achieve 80% power based on an effect size of probability of 0.24 that an observation in the PRVCa group is less than an observation in the ASV group using the Mann-Whitney tests, an alpha of 0.05 (two-tailed) and a 20% dropout.

JUDGING FEASIBILITY

Once you have a draft of your study design, including the estimated sample size, it is time to judge the overall feasibility of the study before committing to it.

Factors to consider when judging the feasibility of a new study
Table 1 shows some of the most important factors in judging feasibility. The first question is whether the outcome will be worth the resources needed to complete the study, implying that you must define costs and benefits. Second, assure yourself that you can both define and measure the outcome variables of interest, which can be a challenge in psychological studies and even in quality improvement projects. Next consider the time constraints, which are affected mainly by the sample size and the time needed to observe all the individuals in that sample. Naturally, if you are studying a rare disorder, the time needed to collect even a modest sample size may make the project impractical.

Every study has associated costs. Those costs and the sources of funding must be identified. Don’t forget costs for consultants, particularly if you need statistical consultation.

Finally, consider your level of experience. If you are contemplating your first study, a human clinical trial might not be the best choice, given the complexity of such a project. Studies such as a meta-analysis or mathematical simulation require special training beyond basic research procedures, and should be avoided.

INTRODUCTION

Basic research skills are not acquired from medical school but from a mentor.1,2 A mentor with experience in study design and technical writing can make a real difference in your career. Most good mentors have more ideas for studies than they have time for research, so they are willing to share and guide your course. Your daily clinical experience provides a wealth of ideas in the form of “why do we do it this way” or “what is the evidence for” or “how can we improve outcomes or cut cost?” Of course, just about every study you read in a medical journal has suggestions for further research in the discussion section. Finally, keep in mind that the creation of study ideas and in particular, hypotheses, is a mysterious process, as this quote indicates: “It is not possible, deliberately, to create ideas or to control their creation. What we can do deliberately is to prepare our minds.” 3 Remember that chance favors the prepared mind.

DEVELOPING THE STUDY IDEA

Often, the most difficult task for someone new to research is developing a practical study idea. This section will explain a detailed process for creating a formal research protocol. We will focus on two common sticking points: (1) finding a good idea, and (2) developing a good idea into a problem statement.

Novice researchers with little experience, no mentors, and short time frames are encouraged not to take on a clinical human study as the principle investigator. Instead, device evaluations are a low-cost, time-efficient alternative. Human studies in the form of a survey are also possible and are often exempt from full Institutional Review Board (IRB) review. Many human-like conditions can be simulated, as was done, for example, in the study of patient-ventilator synchrony.4,5 And if you have the aptitude, whole studies can be based on mathematical models and predictions, particularly with the vast array of computer tools now available.6,7 And don’t forget studies based on surveys.8

A structured approach

A structured approach for developing a formal research protocol.
Figure 1. A structured approach for developing a formal research protocol.

A formal research protocol is required for any human research. However, it is also recommended for all but the simplest investigations. Most of the new researchers I have mentored take a rather lax approach to developing the protocol, and most IRBs are more interested in protecting human rights than validating the study design. As a result, much time is wasted and sometimes an entire study has to be abandoned due to poor planning. Figure 1 illustrates a structured approach that helps to ensure success. It shows a 3-step, iterative process.

The first step is a process of expanding the scope of the project, primarily through literature review. Along the way you learn (or invent) appropriate terminology and become familiar with the current state of the research art on a broad topic. For example, let’s suppose you were interested in the factors that affect the duration of mechanical ventilation. The literature review might include topics such as weaning and patient-ventilator synchrony as well as ventilator-associated pneumonia. During this process, you might discover that the topic of synchrony is currently generating a lot of interest in the literature and generating a lot of questions or confusion. You then focus on expanding your knowledge in this area.

In the second step, you might develop a theoretical framework for understanding patient-ventilator synchrony that could include a mathematical model and, perhaps, an idea to include simulation to study the problem.

In the third step, you need to narrow the scope of the study to a manageable level that includes identifying measurable outcome variables, creating testable hypotheses, considering experimental designs, and evaluating the overall feasibility of the study. At this point, you may discover that you cannot measure the specific outcome variables indicated by your theoretical framework. In that case, you need to create a new framework for supporting your research. Alternatively, you may find that it is not possible to conduct the study you envision given your resources. In that case, it is back to step 1.

Eventually, this process will result in a well-planned research protocol that is ready for review. Keep in mind that many times a protocol needs to be refined after some initial experiments are conducted. For human studies, any changes to the protocol must be approved by the IRB.

The problem statement rubric

The most common problem I have seen novices struggle with is creating a meaningful problem statement and hypothesis. This is crucial because the problem statement sets the stage for the methods, the methods yield the results, and the results are analyzed in light of the original problem statement and hypotheses. To get past any writer’s block, I recommend that you start by just describing what you see happening and why you think it is important. For example, you might say, “Patients with acute lung injury often seem to be fighting the ventilator.” This is important because patient-ventilator asynchrony may lead to increased sedation levels and prolonged intensive care unit stays. Now you can more easily envision a specific purpose and testable hypothesis. For example, you could state that the purpose of this study is to determine the baseline rates of different kinds of patient-ventilator synchrony problems. The hypothesis is that the rate of dyssynchrony is correlated with duration of mechanical ventilation.

Here is an actual example of how a problem statement evolved from a vague notion to a testable hypothesis.

Original: The purpose of this study is to determine whether measures of ineffective cough in patients with stroke recently liberated from mechanical ventilation correlate with risk of extubation failure and reintubation.

Final: The purpose of this study is to test the hypothesis that use of CoughAssist device in the immediate post-extubation period by stroke patients reduces the rate of extubation failure and pneumonia.

The original statement is a run-on sentence that is vague and hard to follow. Once the actual treatment and outcome measures are in focus, then a clear hypothesis statement can be made. Notice that the hypothesis should be clear enough that the reader can anticipate the actual experimental measures and procedures to be described in the methods section of the protocol.

Here is another example:

Original: The purpose of this study is to evaluate a device that allows continuous electronic cuff pressure control.

Final: The purpose of this study is to test the hypothesis that the Pressure Eyes electronic cuff monitor will maintain constant endotracheal tube cuff pressures better than manual cuff inflation during mechanical ventilation.

The problem with the original statement is that “to evaluate” is vague. The final statement makes the outcome variable explicit and suggests what the experimental procedure will be.

This is a final example:

Original: Following cardiac/respiratory arrest, many patients are profoundly acidotic. Ventilator settings based on initial arterial blood gases may result in inappropriate hyperventilation when follow-up is delayed. The purpose of this study is to establish the frequency of this occurrence at a large academic institution and the feasibility of a quality improvement project.

Final: The primary purpose of this study is to evaluate the frequency of hyperventilation occurring post-arrest during the first 24 hours. A secondary purpose is to determine if this hyperventilation is associated with an initial diagnosis of acidosis.

Note that the original statement follows the rubric of telling us what is observed and why it is important. However, the actual problem statement derived from the observation is vague: what is “this occurrence” and is the study really to establish any kind of feasibility? The purpose is simply to evaluate the frequency of hyperventilation and determine if the condition is associated with acidosis.

 

 

EXAMPLES OF RESEARCH PROJECTS BY FELLOWS

The following are examples of well-written statements of study purpose from actual studies conducted by our fellows.

Device evaluation

Defining “Flow Starvation” in volume control mechanical ventilation.

  • The purpose of this study is to evaluate the relationship between the patient and ventilator inspiratory work of breathing to define the term “Flow Starvation.”

Auto-positive end expiratory pressure (auto-PEEP) during airway pressure release ventilation varies with the ventilator model.

  • The purpose of this study was to compare auto-PEEP levels, peak expiratory flows, and flow decay profiles among 4 common intensive care ventilators.

Patient study

Diaphragmatic electrical activity and extubation outcomes in newborn infants: an observational study.

  • The purpose of this study is to describe the electrical activity of the diaphragm before, during, and after extubation in a mixed-age cohort of preterm infants.

Comparison of predicted and measured carbon dioxide production for monitoring dead space fraction during mechanical ventilation.

  • The purpose of this pilot study was to compare dead space with tidal volume ratios calculated from estimated and measured values for carbon dioxide production.

Practice evaluation

Incidence of asynchronies during invasive mechanical ventilation in a medical intensive care unit.

  • The purpose of this study is to conduct a pilot investigation to determine the baseline incidence of various forms of patient-ventilator dyssynchrony during invasive mechanical ventilation.

Simulation training results in improved knowledge about intubation policies and procedures.

  • The purpose of this study was to develop and test a simulation-based rapid-sequence intubation curriculum for fellows in pulmonary and critical care training.

HOW TO SEARCH THE LITERATURE

After creating a problem statement, the next step in planning research is to search the literature. The 10th issue of Respiratory Care journal in 2009 was devoted to research. Here are the articles in that issue related to the literature search:

  • How to find the best evidence (search internet)9
  • How to read a scientific research paper10
  • How to read a case report (or teaching case of the month)11
  • How to read a review paper.12

I recommend that you read these papers.

Literature search resources

My best advice is to befriend your local librarian.13 These people seldom get the recognition they deserve as experts at finding information and even as co-investigators.14 In addition to personal help, some libraries offer training sessions on various useful skills.

PubMed

The Internet resource I use most often is PubMed (www.ncbi.nlm.nih.gov/pubmed). It offers free access to MEDLINE, which is the National Library of Medicine’s database of citations and abstracts in the fields of medicine, nursing, dentistry, veterinary medicine, health care systems, and preclinical sciences. There are links to full-text articles and other resources. The website provides a clinical queries search filters page as well as a special queries page. Using a feature called “My NCBI,” you can have automatic e-mailing of search updates and save records and filters for search results. Access the PubMed Quick Start Guide for frequently asked questions and tutorials.

SearchMedica.com

The SearchMedica website (www.searchmedica.co.uk) is free and intended for medical professionals. It provides answers for clinical questions. Searches return articles, abstracts, and recommended medical websites.

Synthetic databases

There is a class of websites called synthetic databases, which are essentially prefiltered records for particular topics. However, these sites are usually subscription-based, and the cost is relatively high. You should check with your medical library to get access. Their advantage is that often they provide the best evidence without extensive searches of standard, bibliographic databases. Examples include the Cochrane Database of Systematic Reviews (www.cochrane.org/evidence), the National Guideline Clearinghouse (www.guideline.gov), and UpToDate (www.uptodate.com). UpToDate claims to be the largest clinical community in the world dedicated to synthesized knowledge for clinicians and patients. It features the work of more than 6,000 expert clinician authors/reviewers on more than 10,000 topics in 23 medical specialties. The site offers graded recommendations based on the best medical evidence.

Portals

Portals are web pages that act as a starting point for using the web or web-based services. One popular example is ClinicalKey (www.clinicalkey.com/info), formerly called MD Consult, which offers books, journals, patient education materials, and images. Another popular portal is Ovid (ovid.com), offering books, journals, evidence-based medicine databases, and CINAHL (Cumulative Index to Nursing and Allied Health Literature).

Electronic journals

Many medical journals now have online databases of current and archived issues. Such sites may require membership to access the databases, so again, check with your medical library. Popular examples in pulmonary and critical care medicine include the following:

Electronic books

Amazon.com is a great database search engine for books on specific topics. It even finds out-of-print books. And you don’t have to buy the books, because now you can rent them. Sometimes, I find what I wanted by using the “Look Inside” feature for some books. Note that you can look for books at PubMed. Just change the search box from PubMed to Books on the PubMed home page. Of course, Google also has a book search feature. A great (subscription) resource for medical and technical books is Safari (https://www.safaribooksonline.com). Once again, your library may have a subscription.

General Internet resources

You probably already know about Google Scholar (scholar.google.com) and Wikipedia.com. Because of its open source nature, you should use Wikipedia with caution. However, I have found it to be a very good first step in finding technical information, particularly about mathematics, physics, and statistics.

 

 

Using reference management software

One of the most important things you can do to make your research life easier is to use some sort of reference management software. As described in Wikipedia, “Reference management software, citation management software or personal bibliographic management software is software for scholars and authors to use for recording and using bibliographic citations (references). Once a citation has been recorded, it can be used time and again in generating bibliographies, such as lists of references in scholarly books, articles, and essays.” I was late in adopting this technology, but now I am a firm believer. Most Internet reference sources offer the ability to download citations to your reference management software. Downloading automatically places the citation into a searchable database on your computer with backup to the Internet. In addition, you can get the reference manager software to find a PDF version of the manuscript and store it with the citation on your computer (and/or in the Cloud) automatically.

But the most powerful feature of such software is its ability to add or subtract and rearrange the order of references in your manuscripts as you are writing, using seamless integration with Microsoft Word. The references can be automatically formatted using just about any journal’s style. This is a great time saver for resubmitting manuscripts to different journals. If you are still numbering references by hand (God forbid) or even using the Insert Endnote feature in Word (deficient when using multiple occurrences of the same reference), your life will be much easier if you take the time to start using reference management software.

The most popular commercial software is probably EndNote (endnote.com). A really good free software system with about the same functionality as Zotero (zotero.com). Search for “comparison of reference management software” in Wikipedia. You can find tutorials on software packages in YouTube.

STUDY DESIGN

Schematic of pre-experimental research designs.
Figure 2. Schematic of pre-experimental research designs.

When designing the experiment, note that there are many different approaches, each with their advantages and disadvantages. A full treatment of this topic is beyond the scope of this article. Suffice it to say that pre-experimental designs (Figure 2) are considered to generate weak evidence. But they are quick and easy and might be appropriate for pilot studies.

Schematic of a quasi-experimental research design.
Figure 3. Schematic of a quasi-experimental research design.

Quasi-experimental designs (Figure 3) generate a higher level of evidence. Such a design might be appropriate when you are stuck with collecting a convenience sample, rather than being able to use a full randomized assignment of study subjects.

The randomized controlled study design.
Figure 4. The randomized controlled study design.

The fully randomized design (Figure 4) generates the highest level of evidence. This is because if the sample size is large enough, the unknown and uncontrollable sources of bias are evenly distributed between the study groups. 

BASIC MEASUREMENT METHODS

If your research involves physical measurements, you need to be familiar with the devices considered to be the gold standards. In cardiopulmonary research, most measurements involve pressure volume, flow, and gas concentration. You need to know which devices are appropriate for static vs dynamic measurements of these variables. In addition, you need to understand issues related to systematic and random measurement errors and how these errors are managed through calibration and calibration verification. I recommend these two textbooks:

Principles and Practice of Intensive Care Monitoring 1st Edition by Martin J. Tobin MD.

  • This book is out of print, but if you can find a used copy or one in a library, it describes just about every kind of physiologic measurement used in clinical medicine.

Medical Instrumentation: Application and Design 4th Edition by John G. Webster.

  • This book is readily available and reasonably priced. It is a more technical book describing medical instrumentation and measurement principles. It is a standard textbook for biometrical engineers.

STATISTICS FOR THE UNINTERESTED

I know what you are thinking: I hate statistics. Look at the book Essential Biostatistics: A Nonmathematical Approach.15 It is a short, inexpensive paperback book that is easy to read. The author does a great job of explaining why we use statistics rather than getting bogged down explaining how we calculate them. After all, novice researchers usually seek the help of a professional statistician to do the heavy lifting.

My book, Handbook for Health Care Research,16 covers most of the statistical procedures you will encounter in medical research and gives examples of how to use a popular tactical software package called SigmaPlot. By the way, I strongly suggest that you consult a statistician early in your study design phase to avoid the disappointment of finding out later that your results are uninterpretable. For an in-depth treatment of the subject, I recommend How to Report Statistics in Medicine.17

Statistical bare essentials

Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data.
Figure 5. Simple graphs that you should be able to make using a spreadsheet program that contains your experimental data. COPD = chronic obstructive pulmonary disease; PaCOs = partial pressure of carbon dioxide, artery; PS = pressure support; RDS = respiratory distress syndrome; SIMV = synchronized intermittent mandatory ventilation

To do research or even just to understand published research reports, you must have at least a minimal skill set. The necessary skills include understanding some basic terminology, if only to be able to communicate with a statistician consultant. Important terms include levels of measurement (nominal, ordinal, continuous), accuracy, precision, measures of central tendency (mean, median, mode), measures of variability (variance, standard deviation, coefficient of variation), and percentile. The first step in analyzing your results is usually to represent it graphically. That means you should be able to use a spreadsheet to make simple graphs (Figure 5).

Example flowchart for selecting the appropriate statistical test.
Figure 6. Example flowchart for selecting the appropriate statistical test. ANOVA = analysis of variance

You should also know the basics of inferential statistics (ie, hypothesis testing). For example, you need to know the difference between parametric and non-parametric tests. You should be able to explain correlation and regression and know when to use Chi-squared vs a Fisher exact test. You should know that when comparing two mean values, you typically use the Student’s t test (and know when to use paired vs unpaired versions of the test). When comparing more than 2 mean values, you use analysis of variance methods (ANOVA). You can teach yourself these concepts from a book,16 but even an introductory college level course on statistics will be immensely helpful. Most statistics textbooks provide some sort of map to guide your selection of the appropriate statistical test (Figure 6), and there are good articles in medical journals.

You can learn a lot simply by reading the Methods section of research articles. Authors will often describe the statistical tests used and why they were used. But be aware that a certain percentage of papers get published with the wrong statistics.18 

One of the underlying assumptions of most parametric statistical methods is that the data may be adequately described by a normal or Gaussian distribution. This assumption needs to be verified before selecting a statistical test. The common test for data normality is the Kolmogorov-Smirnov test. The following text from a methods section describes 2 very common procedures—the Student’s t test for comparing 2 mean values and the one-way ANOVA for comparing more than 2 mean values.19

“Normal distribution of data was verified using the Kolmogorov-Smirnov test. Body weights between groups were compared using one-way ANOVA for repeated measures to investigate temporal differences. At each time point, all data were analyzed using one-way ANOVA to compare PCV and VCV groups. Tukey’s post hoc analyses were performed when significant time effects were detected within groups, and Student’s t test was used to investigate differences between groups. Data were analyzed using commercial software and values were presented as mean ± SD. A P value < .05 was considered statistically significant.” 

 

 

Estimating sample size and power analysis

One very important consideration in any study is the required number of study subjects for meaningful statistical conclusions. In other words, how big should the sample size be? Sample size is important because it affects the feasibility of the study and the reliability of the conclusions in terms of statistical power. The necessary sample size depends on 2 basic factors. One factor is the variability of the data (often expressed as the standard deviation). The other factor is the effect size, meaning, for example, how big of a difference between mean values you want to detect. In general, the bigger the variability and the smaller the difference, the bigger the sample size required.

As the above equation shows, the effect size is expressed, in general, as a mean difference divided by a standard deviation. In the first case, the numerator represents the difference between the sample mean and the assumed population mean. In the denominator, SD is the standard deviation of the sample (used to estimate the standard deviation of the population). In the second case, the numerator represents the difference between the mean values of 2 samples and the denominator is the pooled standard deviation of the 2 samples.

In order to understand the issues involved with selecting sample size, we need to first understand the types of errors that can be made in any type of decision. Suppose our research goal is to make a decision about whether a new treatment results in a clinical difference (improvement). The results of our statistical test are dichotomous—we decide either yes there is a significant difference or no there isn’t. The truth, which we may never know, is that in reality, the difference exists or it doesn’t.

Types of errors in statistical decision making.
Figure 7. Types of errors in statistical decision making.

As Figure 7 shows, the result of our decision making is that there are 2 ways to be right and 2 ways to be wrong. If we decide there is a difference (eg, our statistical tests yields P ≤ .05) but in realty there is not a difference, then we make what is called a type I error. On the other hand, if we conclude that there is not a difference (ie, our statistical test yields P > .05) but in reality there is a difference that we did not detect, then we have made a type II error.

Probabilities associated with type I and type II errors.
Figure 8. Probabilities associated with type I and type II errors.

The associated math is shown in Figure 8. The probability of making a type I error is called alpha. By convention in medicine, we set our rejection criterion to alpha = 0.05. In other words, we would reject the null hypothesis (that there is no difference) anytime our statistical test yields a P value less than alpha. The probability of making a type II error is called beta. For historical reasons, the probability of not making a type II error is called the statistical power of the test and is equal to 1 minus beta. Power is affected by sample size: the larger the sample the larger the power. Most researchers, by convention, keep the sample size large enough to keep power above 0.80.

Nomogram for calculating power and sample size
Figure 9. Nomogram for calculating power and sample size.

Figure 9 is a nomogram that brings all these ideas together. The red line shows that for your study, given the desired effect size (0.8), if you collected samples from the 30 patients you planned on then the power would be unacceptable at 0.60, indicating a high probability of a false negative decision if the P value comes out greater than .50. The solution is to increase the sample size to about 50 (or more), as indicated by the blue line. From this nomogram we can generalize to say that when you want to detect a small effect with data that have high variability, you need a large sample size to provide acceptable power.

The text below is an example of a power analysis presented in the methods section of a published study.20 Note that the authors give their reasoning for the sample size they selected. This kind of explanation may inform your study design. But what if you don’t know the variability of the data you want to collect? In that case, you need to collect some pilot data and calculate from that an appropriate sample size for a subsequent study.

A prospective power calculation indicated that a sample size of 25 per group was required to achieve 80% power based on an effect size of probability of 0.24 that an observation in the PRVCa group is less than an observation in the ASV group using the Mann-Whitney tests, an alpha of 0.05 (two-tailed) and a 20% dropout.

JUDGING FEASIBILITY

Once you have a draft of your study design, including the estimated sample size, it is time to judge the overall feasibility of the study before committing to it.

Factors to consider when judging the feasibility of a new study
Table 1 shows some of the most important factors in judging feasibility. The first question is whether the outcome will be worth the resources needed to complete the study, implying that you must define costs and benefits. Second, assure yourself that you can both define and measure the outcome variables of interest, which can be a challenge in psychological studies and even in quality improvement projects. Next consider the time constraints, which are affected mainly by the sample size and the time needed to observe all the individuals in that sample. Naturally, if you are studying a rare disorder, the time needed to collect even a modest sample size may make the project impractical.

Every study has associated costs. Those costs and the sources of funding must be identified. Don’t forget costs for consultants, particularly if you need statistical consultation.

Finally, consider your level of experience. If you are contemplating your first study, a human clinical trial might not be the best choice, given the complexity of such a project. Studies such as a meta-analysis or mathematical simulation require special training beyond basic research procedures, and should be avoided.

References
  1. Tobin MJ. Mentoring: seven roles and some specifics. Am J Respir Crit Care Med 2004; 170:114–117.
  2. Chatburn RL. Advancing beyond the average: the importance of mentoring in professional achievement. Respir Care 2004; 49:304–308.
  3. Beveridge WIB. The Art of Scientific Investigation. New York, NY: WW Norton & Company; 1950.
  4. Chatburn RL, Mireles-Cabodevila E, Sasidhar M. Tidal volume measurement error in pressure control modes of mechanical ventilation: a model study. Comput Biol Med 2016; 75:235–242.
  5. Mireles-Cabodevila E, Chatburn RL. Work of breathing in adaptive pressure control continuous mandatory ventilation. Respir Care 2009; 54:1467–1472.
  6. Chatburn RL, Ford RM. Procedure to normalize data for benchmarking. Respir Care 2006; 51:145–157.
  7. Bou-Khalil P, Zeineldine S, Chatburn R, et al. Prediction of inspired oxygen fraction for targeted arterial oxygen tension following open heart surgery in non-smoking and smoking patients. J Clin Monit Comput 2016. https://doi.org/10.1007/s10877-016-9941-6.
  8. Mireles-Cabodevila E, Diaz-Guzman E, Arroliga AC, Chatburn RL. Human versus computer controlled selection of ventilator settings: an evaluation of adaptive support ventilation and mid-frequency ventilation. Crit Care Res Pract 2012; 2012:204314.
  9. Chatburn RL. How to find the best evidence. Respir Care 2009; 54:1360–1365.
  10. Durbin CG Jr. How to read a scientific research paper. Respir Care 2009; 54:1366–1371.
  11. Pierson DJ. How to read a case report (or teaching case of the month). Respir Care 2009; 54:1372–1378.
  12. Callcut RA, Branson RD. How to read a review paper. Respir Care 2009; 54:1379–1385.
  13. Eresuma E, Lake E. How do I find the evidence? Find your librarian—stat! Orthop Nurs 2016; 35:421–423.
  14. Janke R, Rush KL. The academic librarian as co-investigator on an interprofessional primary research team: a case study. Health Info Libr J 2014; 31:116–122.
  15. Motulsky H. Essential Biostatistics: A Nonmathematical Approach. New York, NY: Oxford University Press; 2016.
  16. Chatburn RL. Handbook for Health Care Research. 2nd ed. Sudbury, MA: Jones and Bartlett Publishers; 2011.
  17. Lang TA, Secic M. How to Report Statistics in Medicine. 2nd ed. Philadelphia, PA: American College of Physicians; 2006.
  18. Prescott RJ, Civil I. Lies, damn lies and statistics: errors and omission in papers submitted to INJURY 2010–2012. Injury 2013; 44:6–11.
  19. Fantoni DT, Ida KK, Lopes TF, Otsuki DA, Auler JO Jr, Ambrosio AM. A comparison of the cardiopulmonary effects of pressure controlled ventilation and volume controlled ventilation in healthy anesthetized dogs. J Vet Emerg Crit Care (San Antonio) 2016; 26:524–530.
  20. Gruber PC, Gomersall CD, Leung P, et al. Randomized controlled trial comparing adaptive-support ventilation with pressure-regulated volume-controlled ventilation with automode in weaning patients after cardiac surgery. Anesthesiology 2008; 109:81–87.
References
  1. Tobin MJ. Mentoring: seven roles and some specifics. Am J Respir Crit Care Med 2004; 170:114–117.
  2. Chatburn RL. Advancing beyond the average: the importance of mentoring in professional achievement. Respir Care 2004; 49:304–308.
  3. Beveridge WIB. The Art of Scientific Investigation. New York, NY: WW Norton & Company; 1950.
  4. Chatburn RL, Mireles-Cabodevila E, Sasidhar M. Tidal volume measurement error in pressure control modes of mechanical ventilation: a model study. Comput Biol Med 2016; 75:235–242.
  5. Mireles-Cabodevila E, Chatburn RL. Work of breathing in adaptive pressure control continuous mandatory ventilation. Respir Care 2009; 54:1467–1472.
  6. Chatburn RL, Ford RM. Procedure to normalize data for benchmarking. Respir Care 2006; 51:145–157.
  7. Bou-Khalil P, Zeineldine S, Chatburn R, et al. Prediction of inspired oxygen fraction for targeted arterial oxygen tension following open heart surgery in non-smoking and smoking patients. J Clin Monit Comput 2016. https://doi.org/10.1007/s10877-016-9941-6.
  8. Mireles-Cabodevila E, Diaz-Guzman E, Arroliga AC, Chatburn RL. Human versus computer controlled selection of ventilator settings: an evaluation of adaptive support ventilation and mid-frequency ventilation. Crit Care Res Pract 2012; 2012:204314.
  9. Chatburn RL. How to find the best evidence. Respir Care 2009; 54:1360–1365.
  10. Durbin CG Jr. How to read a scientific research paper. Respir Care 2009; 54:1366–1371.
  11. Pierson DJ. How to read a case report (or teaching case of the month). Respir Care 2009; 54:1372–1378.
  12. Callcut RA, Branson RD. How to read a review paper. Respir Care 2009; 54:1379–1385.
  13. Eresuma E, Lake E. How do I find the evidence? Find your librarian—stat! Orthop Nurs 2016; 35:421–423.
  14. Janke R, Rush KL. The academic librarian as co-investigator on an interprofessional primary research team: a case study. Health Info Libr J 2014; 31:116–122.
  15. Motulsky H. Essential Biostatistics: A Nonmathematical Approach. New York, NY: Oxford University Press; 2016.
  16. Chatburn RL. Handbook for Health Care Research. 2nd ed. Sudbury, MA: Jones and Bartlett Publishers; 2011.
  17. Lang TA, Secic M. How to Report Statistics in Medicine. 2nd ed. Philadelphia, PA: American College of Physicians; 2006.
  18. Prescott RJ, Civil I. Lies, damn lies and statistics: errors and omission in papers submitted to INJURY 2010–2012. Injury 2013; 44:6–11.
  19. Fantoni DT, Ida KK, Lopes TF, Otsuki DA, Auler JO Jr, Ambrosio AM. A comparison of the cardiopulmonary effects of pressure controlled ventilation and volume controlled ventilation in healthy anesthetized dogs. J Vet Emerg Crit Care (San Antonio) 2016; 26:524–530.
  20. Gruber PC, Gomersall CD, Leung P, et al. Randomized controlled trial comparing adaptive-support ventilation with pressure-regulated volume-controlled ventilation with automode in weaning patients after cardiac surgery. Anesthesiology 2008; 109:81–87.
Page Number
e10-e19
Page Number
e10-e19
Publications
Publications
Article Type
Display Headline
Basics of study design: Practical considerations
Display Headline
Basics of study design: Practical considerations
Legacy Keywords
study design, statistics, hypothesis testing, sample size, power, Robert Chatburn
Legacy Keywords
study design, statistics, hypothesis testing, sample size, power, Robert Chatburn
Citation Override
Cleveland Clinic Journal of Medicine 2017 September;84(suppl 2):e10-e19
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media