Appendicitis 1889 to 2012: What, No Surgery?

Status

Appendicitis 1889 to 2012: What, No Surgery?

All medical students learn about McBurney’s point—that’s the spot, named for McBurney, in the right lower quadrant of the abdomen where classical appendicitis pain finally localizes.[1] If the patient’s history fits the classic history of appendicitis with vague abdominal pain eventually localizing to McBurney’s point, the norm has been—at least in the U.S. —to take the appendix out. However, as pointed out in a new systematic review done as a meta-analysis, starting in the late 1950s there were reports of success in treating appendicitis with conservative therapy (antibiotics) and good outcomes without resorting to appendectomy.[2]

This systematic review presents a review of our traditions and lack of conclusive evidence about best practices in managing appendicitis and suggests that, for many patients, avoiding appendectomy may be a reasonable option. The current meta-analysis of four selected randomized controlled trials from 59 eligible trials with a total of 900 patients, reported a relative risk reduction for complications (perforation, peritonitis, wound infection) from appendicitis of 31% for antibiotic treatment compared with appendectomy (risk ratio 0.69 (95% confidence interval 0.54 to 0.89); I2=0%; P=0.004). There were no significant differences between antibiotic treatment and appendectomy for length of hospital stay, efficacy of treatment, or risk of complicated appendicitis.

The biggest problem in this meta-analysis is that the results are based on trials with significant threats to validity. Randomization sequence was computer generated in one trial, by “external randomization” in one trial, by date of birth in one trial and unclear in one trial. Concealment of allocation was by sealed envelopes in two trials and not reported in the other two trials. All trials were unblinded. Withdrawal rates are unclear. Therefore, it is uncertain how much the results of this meta-analysis may have been distorted by bias. In addition, as pointed out by an editorialist, in patients who have persistent problems despite antibiotic treatment, delayed appendectomy might be necessary.[3] Delayed appendectomy has been associated with a high complication rate. Also, if a patient develops an inflammatory phlegmon—a palpable mass at clinical examination or an inflammatory mass or abscess at imaging or at surgical exploration—appendectomy sometimes has to be converted to an ileocecal resection—a much more involved operation. Another important issue with antibiotic treatment is the chance of recurrence. The current meta-analysis found a 20% chance of recurrence of appendicitis after conservative treatment within one year. Of the recurrences, 20% of patients presented with a perforated or gangrenous appendicitis. The editorialist questions whether a failure rate of 20% within one year is acceptable.

These four trials and this meta-analysis suggest that antibiotics may be safe for some patients with uncomplicated appendicitis. If this option is considered, we believe detailed information about the uncertainties regarding benefits and risks should be made known to patients. Details are available at http://www.bmj.com/content/344/bmj.e2156

References

1. Thomas CG Jr. Experiences with Early Operative Interference in Cases of Disease of the Vermiform Appendix by Charles McBurney, M.D., Visiting Surgeon to the Roosevelt Hospital, New York City. Rev Surg. 1969 May-Jun;26(3):153-66. PubMed PMID: 4893208.

2. Varadhan KK, Neal KR, Lobo DN. Safety and efficacy of antibiotics compared with appendicectomy for treatment of uncomplicated acute appendicitis: meta-analysis of randomised controlled trials. BMJ. 2012 Apr 5;344:e2156. doi: 10.1136/bmj.e2156. PubMed PMID: 22491789.

3. BMJ 2012;344:e2546 (Published 5 April 2012).

 

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

Empirical Evidence of Attrition Bias in Clinical Trials

Status

Empirical Evidence of Attrition Bias in Clinical Trials

The commentary, “Empirical evidence of attrition bias in clinical trials,” by Juni et al [1] is a nice review of what has transpired since 1970 when attrition bias received attention in a critical appraisal of a non-valid trial of extracranial bypass surgery for transient ischemic attack. [2] At about the same time Bradford Hill coined the phrase “intention-to-treat.”  He wrote that excluding patient data after “admission to the treated or control group” may affect the validity of clinical trials and that “unless the losses are very few and therefore unimportant, we may inevitably have to keep such patients in the comparison and thus measure the ‘intention-to-treat’ in a given way, rather than the actual treatment.”[3] The next major development was meta-epidemiological research which assessed trials for associations between methodological quality and effect size and found conflicting results in terms of the effect of attrition bias on effect size.  However, as the commentary points out, the studies assessing attrition bias were flawed. [4,5,6].

Finally a breakthrough in understanding the distorting effect of loss of subjects following randomization was seen by two authors evaluating attrition bias in oncology trials.[7] The investigators compared the results from their analyses which utilized individual patient data, which invariably followed the intention-to-treat principle with those done by the original investigators, which often excluded some or many patients. The results showed that pooled analyses of trials with patient exclusions reported more beneficial effects of the experimental treatment than analyses based on all or most patients who had been randomized. Tierney and Stewart showed that, in most meta-analyses they reviewed based on only “included” patients, the results favored the research treatment (P = 0.03). The commentary gives deserved credit to Tierney and Stewart for their tremendous contribution to critical appraisal and is a very nice, short read.

References

1. Jüni P, Egger M. Commentary: Empirical evidence of attrition bias in clinical  trials. Int J Epidemiol. 2005 Feb;34(1):87-8. Epub 2005 Jan 13. Erratum in: Int J Epidemiol. 2006 Dec;35(6):1595. PubMed PMID: 15649954.

2. Fields WS, Maslenikov V, Meyer JS, Hass WK, Remington RD, Macdonald M. Joint study of extracranial arterial occlusion. V. Progress report of prognosis following surgery or nonsurgical treatment for transient cerebral ischemic attacks. PubMed PMID: 5467158.

3. Bradford Hill A. Principles of Medical Statistics, 9th edn. London: The Lancet Limited, 1971.

4. Schulz KF, Chalmers I, Hayes RJ, Altman D. Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA 1995;273:408–12. PMID: 7823387

5. Kjaergard LL, Villumsen J, Gluud C. Reported methodological quality and discrepancies between large and small randomized trials in metaanalyses. Ann Intern Med 2001;135:982–89. PMID 11730399

6. Balk EM, Bonis PA, Moskowitz H, Schmid CH, Ioannidis JP, Wang C, Lau J. Correlation of quality measures with estimates of treatment effect in meta-analyses of randomized controlled trials. JAMA. 2002 Jun 12;287(22):2973-82. PubMed PMID: 12052127.

7. Tierney JF, Stewart LA. Investigating patient exclusion bias in meta-analysis. Int J Epidemiol. 2005 Feb;34(1):79-87. Epub 2004 Nov 23. PubMed PMID: 15561753.

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

A Caution When Evaluating Systematic Reviews and Meta-analyses

Status

A Caution When Evaluating Systematic Reviews and Meta-analyses

We would like to draw critical appraisers’ attention to an infrequent but important problem encountered in some systematic reviews—the accuracy of standardized mean differences in some reviews. Meta-analysis of trials that have used different scales to record outcomes of a similar nature requires data transformation to a uniform scale, the standardized mean difference (SMD). Gøtzsche and colleagues, in a review of 27 meta-analyses utilizing SMD found that a high proportion of meta-analyses based on SMDs contained meaningful errors in data extraction and calculation of point estimates.[1] Gøtzsche et al. audited two trials from each review and found that, in 17 meta-analyses (63%), there were errors for at least 1 of the 2 trials examined. We recommend that critical appraisers be aware of this issue.

1. Gøtzsche PC, Hróbjartsson A, Maric K, Tendal B. Data extraction errors in meta-analyses that use standardized mean differences. JAMA. 2007 Jul 25;298(4):430-7. Erratum in: JAMA. 2007 Nov 21;298(19):2264. PubMed PMID:17652297.

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

Improving Results Reporting in Clinical Trials: Case Study—Time-to-Event Analysis and Hazard Ratio Reporting Advice

Status

Improving Results Reporting in Clinical Trials: Case Study—Time-to-Event Analysis and Hazard Ratio Reporting Advice

We frequently see clinical trial abstracts—especially those using time-to-event analyses—that are not well-understood by readers. Fictional example for illustrative purposes:

In a 3-year randomized controlled trial (RCT) of drug A versus placebo in women with advanced breast cancer, the investigators presented their abstract results in terms of relative risk reduction for death (19%) along with the hazard ratio (hazard ratio = 0.76, 95% confidence interval [CI] 0.56 to 0.94, P = 0.04). They also stated that, “This reduction represented a 5-month improvement in median survival (24 months in the drug A group vs. 19 months in the placebo group).” Following this information, the authors stated that the three-year survival probability was 29% in the drug A group versus 21.0% in the placebo group.

Many readers do not understand hazard ratios and will conclude that a 5 month improvement in median survival is not clinically meaningful. We believe it would have been more useful to present mortality information (which the authors frequently present in  results section, but is not easily found by many readers).

A much more meaningful abstract statement would go something like this: After 3 years, the overall mortality was 59% in the drug A group compared with 68% in the placebo group which represents an absolute risk reduction (ARR) of 9%, P=0.04, number needed to treat (NNT) 11.  This information is much more impressive and much more easily understood than a 5-month increase in median survival and uses statistics familiar to clinicians.

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

Three Questions Patients Should Ask To Improve the Information They Receive

Status

Three Questions Patients Should Ask To Improve the Information They Receive

According to Shepherd  et al [1], the following questions appear to be powerful catalysts for good information exchanges between clinicians and patients:

1. “What are my options?”

2. “What are the benefits and harms of each?”

3. “How likely are the benefits and harms?”

Let’s start with Shepherd’s cross-over trial using the three questions above.[1]  In order to make informed decisions and improve outcomes, patients need reliable information about benefits and risks of the various options.  In this randomized cross-over trial, Shepherd et al. used two standardized patients with identical symptoms— one patient asked the three questions (and also about doing nothing if the physician did not mention this option), the other did not. The patient presented as an otherwise healthy divorced middle-aged female with one prior undiagnosed episode of depression and 3 months of worsening moderate symptoms of depression. Depression was chosen as the condition because evidence is available and patients express differences in preference for treatment. The authors found that the 3 questions were associated with greater provision of information and behavior supporting patient involvement without extending appointment time.

Stiggelbout et al. remind us that shared decision-making (SDM) should be routinely employed to ensure patient autonomy, beneficence (balancing risks and benefits), non-malfeasance (avoiding harm) and justice (patients frequently decline procedures when adequate information has been provided and this may result in improved sharing of limited resources).[2] Pamphlets, videos, tools of various sorts may be employed to facilitate SDM. Tactics and tools that appear to increase SDM include—

  • Creating awareness of equipoise (there is no best choice but a decision must be made—even if it is to do nothing);
  • Presenting or encouraging patients to ask about options and benefits and risks of each option;
  • Use of graphical displays to present risks;
  • Use absolute risk information such as the number of similar patients/100 or number/1000 who will benefit*;
  • Encouraging patients to pay attention to their preferences;
  • Provide appropriate support to help patients make decisions—respect the patient’s preference about his or her role—independent, shared or delegated decision-making role.

*We would add that this information is only useful when also providing information  that provides a more complete picture.  To hear that one’s chance of benefiting from an intervention is 5 out of a hundred has a very different meaning depending upon the specific context:

  • Scenario 1: Out of 100 patients, 10 taking drug A improved as compared to 5 taking placebo, versus—
  • Scenario 2: Out of 100 patients, 90 taking drug A improved as compared to 95 taking placebo.

Examples of decision-aids are available from the following: http://shareddecisions.mayoclinic.org.

Delfini Comment: “Patient demand,”  i.e., activating patients to voice their information needs, has been proposed as a method of improving healthcare consultations for several decades. In our experience, educational programs aimed at increasing the use of  evidence-based information sharing with patients has been hampered by clinicians frequently not possessing accurate answers to the three questions studied here. The two studies discussed above [3,4] indicate  that patient-mediated approaches may be at least part of the answer to improved clinical decision-making.

References

1. Shepherd HL et al. Three questions that patients can ask to improve the quality of information physicians give about treatment options: a cross-over trial. Patient Educ Couns. 2011 Sep;84(3):379-85. Epub 2011 Aug 9.PubMed PMID: 21831558.

2. Stiggelbout AM et al.  Shared decision making: really putting patients at the centre of healthcare. BMJ. 2012 Jan 27;344:e256. doi: 10.1136/bmj.e256. PubMed PMID:22286508.

3. Bell RA et al. Encouraging patients with depressive symptoms to seek care: a mixed methods approach to message development. Patient Educ Couns. 2010 Feb;78(2):198-205. Epub 2009 Aug 11. PubMed PMID: 19674862.

4. Kravitz RL et al. Influence of patients’ requests for direct-to-consumer advertised antidepressants: a randomized controlled trial. JAMA. 2005 Apr 27;293(16):1995-2002. Erratum in: JAMA. 2005 Nov 16;294(19):2436. PubMed PMID: 15855433; PubMed Central PMCID: PMC3155410.

 

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

What’s Next in Obesity Treatment?

Status

What’s Next in Obesity Treatment?

QNEXA® (Vivus, Inc.) looks promising. QNEXA is a once-daily weight loss therapy that contains a combination of immediate-release phentermine hydrochloride (PHEN) and extended-release topiramate (TPM). These two agents suppress appetite through complementary mechanisms (decreased hunger and increased satiety), leading to greater weight loss than with either agent alone. Both PHEN and TPM are FDA approved at higher doses than those contained in QNEXA. The Endocrinologic and Metabolic Drugs Advisory Committee voted 20 to 2 to recommend FDA approval on 2/22/12 and the FDA is expected to release a decision in April. The FDA is waiting for Vivus, Inc. to provide assessment of topiramate’s and phentermine/topiramate’s teratogenic potential and a detailed plan and strategy to evaluate and mitigate any potential teratogenic risks in women of childbearing potential (WOCBP) and evidence that QNEXA-associated elevations in heart rate (mean increase of 1.6 bpm on the highest dose) do not increase the risk for major adverse cardiovascular events.

Currently phentermine is restricted to short-term management of obesity. Topiramate is approved for treatment of seizure disorders. The prescription use of these drugs spans more than 52 years for phentermine and more than 15 years for topiramate.

At present, approved pharmacotherapies for obesity are generally associated with <5% weight loss and are often poorly tolerated. The only treatment demonstrated to reliably produce more than 10% sustained weight loss for obesity is bariatric surgery, but is associated with surgical risks, nutritional deficiencies and infections.

From the FDA Advisory Committee briefing document below: The study population evaluated in the QNEXA pivotal clinical development program included a range of adult subjects, from overweight (BMI >27 kg/m2) to severely obese (BMI >60 kg/m2), with a range of obesity-related comorbidities, including type 2 diabetes, hypertension, and hypertriglyceridemia. Within a total of 3807 patients in the one-year cohort, 752 subjects (19.8%) fell into the “low” cardiovascular risk category , 2498 (65.6%) into “moderate” risk, and 557 (14.6%) into the “high” risk group. Thus, the QNEXA clinical sample of patients represents a broad spectrum of cardiovascular risk, and one that represents the range of patients likely to use QNEXA, if approved.

Two trials and an extension of the second trial [1,2,3] and other data [see link at bottom] comprise evidence provided to the FDA by Vivus. [We have not performed any critical appraisals, FYI.]  The two trials reported that a phentermine/topiramate 15 mg/92 mg combination produced a magnitude of weight loss (~10%) that exceeded levels associated with current pharmacotherapies which produce <5% weight loss. Adverse events on the highest dose include paresethesias ~20%, dry mouth ~17%, constipation ~14%, dizziness ~6% to 10%, dysgeusia ~10%, depression ~5% to 8% and irritability ~3% to 5%. Only 1% to 2% discontinued treatment due to adverse events. It appears likely that many will conclude that benefits outweigh harms.

For more information and details of the Phase 3 studies presented to the FDA, go to http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials
/Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM292317.pdf

1: Allison DB, Gadde KM, Garvey WT, Peterson CA, Schwiers ML, Najarian T, Tam PY,  Troupin B, Day WW. Controlled-Release Phentermine/Topiramate in Severely Obese  Adults: A Randomized Controlled Trial (EQUIP). Obesity (Silver Spring). 2012  Feb;20(2):330-42. doi: 10.1038/oby.2011.330. Epub 2011 Nov 3. PubMed PMID:  22051941.

2. Gadde KM, Allison DB, Ryan DH, Peterson CA, Troupin B, Schwiers ML, Day WW.  Effects of low-dose, controlled-release, phentermine plus topiramate combination   on weight and associated comorbidities in overweight and obese adults (CONQUER):   a randomised, placebo-controlled, phase 3 trial. Lancet. 2011 Apr  16;377(9774):1341-52. Epub 2011 Apr 8. Erratum in: Lancet. 2011 Apr  30;377(9776):1494. PubMed PMID: 21481449.

3. Garvey WT, Ryan DH, Look M, Gadde KM, Allison DB, Peterson CA, Schwiers M, Day  WW, Bowden CH. Two-year sustained weight loss and metabolic benefits with  controlled-release phentermine/topiramate in obese and overweight adults  (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin  Nutr. 2012 Feb;95(2):297-308. Epub 2011 Dec 7. PubMed PMID: 22158731.

 

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

Is “Biologics Versus Biosimilars” A Different Story Than Brand Names Versus Generics?

Status

Is “Biologics Versus Biosimilars” A Different Story Than Brand Names Versus Generics?

In 1984, the Drug Price Competition and Patent Term Restoration Act (Hatch-Waxman Act) created a shortened new drug application pathway for generics by eliminating the need for preclinical and clinical studies if bioequivalence—i.e., the rate and extent of absorption—could be demonstrated. The FDA defines a generic drug as follows: “A generic drug is identical–or bioequivalent—to a brand name drug in dosage form, safety, strength, route of administration, quality, performance characteristics and intended use.”[1] For many drugs, the process was straightforward and payers hoped to reduce expense by purchasing generics.

Although the FDA frequently uses the term “biologic drug,” we have been unable to find an official FDA definition for “biologic drug.” The FDA instead separately defines “biological product” and “therapeutic biological product.” These are the current FDA definitions—Biological Product: “Biological products include a wide range of products such as vaccines, blood and blood components, allergenics, somatic cells, gene therapy, tissues, and recombinant therapeutic proteins. Biologics can be composed of sugars, proteins, or nucleic acids or complex combinations of these substances, or may be living entities such as cells and tissues. Biologics are isolated from a variety of natural sources—human, animal, or microorganism—and may be produced by biotechnology methods and other cutting-edge technologies. Gene-based and cellular biologics, for example, often are at the forefront of biomedical research, and may be used to treat a variety of medical conditions for which no other treatments are available.” The FDA then defines a therapeutic biological product as follows: “A therapeutic biological product is a protein derived from living material (such as cells or tissues) used to treat or cure disease.”[2] Thus, biologics are large-molecule medications produced by living cells. They are produced by using recombinant DNA technology to direct protein synthesis within cells which can then be used to produce medications.

The Patient Protection and Affordable Care Act (Affordable Care Act), signed into law by President Obama on March 23, 2010, amends the Public Health Service Act (PHS Act) to create an abbreviated licensure pathway for biological products that are demonstrated to be “biosimilar” to or “interchangeable” with an FDA-licensed biological product. This pathway is provided in the part of the law known as the Biologics Price Competition and Innovation Act (BPCI Act). Under the BPCI Act, a biological product may be demonstrated to be “biosimilar” if data show that, among other things, the product is “highly similar” to an already-approved biological product. [3] Biosimilars, also called “follow-on biologics,” are in many ways analogous to generics, but are not generic drugs. In Canada they are aptly labeled “subsequent entry biologics (SEBs).” [4] We like the clarity of the Canadian term, but Americans will hear the terms “biologics” and “biosimilars.”

Biologics and biosimilars are large protein molecules. Manufacturing a biosimilar drug poses numerous complexities not seen in the manufacturing of “small molecules” created as generic drugs. A company wishing to manufacture a biosimilar does have access to the commercial biologic product, but does not have access to the biologic cell line used by manufacturerof the reference biologic or details of the manufacturing process such as how fermentation and purification were carried out. This lack of information (and therefore the lack of identical manufactured cellular material), together with the molecular and structural complexity of large biologic proteins makes for immense complexity for the companies who will be creating biosimilars. The bottom line is that even though a biosimilar has the same recombinant DNA sequencing, small differences in structure or chemistry due to the processes and chemicals used in the culture, purification, storage, etc. may result in differences in efficacy, safety and immunological outcomes.

The Health Care Reform Act has outlined a process for abbreviated approval for biosimilars. The FDA has conducted public meetings and published requirements for a drug to be considered biosimilar, emphasizing clinical studies demonstrating that the product is “highly similar” to the reference product, although minor differences are allowed if the differences do not result in clinically meaningful safety, purity and potency differences. In addition, interchangeable biological products may be substituted at the pharmacy level without the intervention of a healthcare provider. [3]

The strength of evidence necessary to result in a decision of biosimilarity and interchangeability remains unclear. It is also currently unclear if separate evidence for each indication will be required. Currently The European Medicines Agency (EMA) has outlined important elements and considerations required to support the designation of biosimilarity in comparison with existing biologics along with fairly detailed scientific guidance documents on biosimilar medicines.[5] Key requirements include similar pharmacokinetics and pharmacodynamics in humans and the demonstration of similar clinical efficacy and safety of the biosimilar compared to that of the reference biologic. Several biosimilar agents have already been licensed in Europe. The American College of Rheumatology has developed a concise position paper which takes a patient-centered approach to biosimilars. The paper is available online. [6] Key points are—

  • Biologics are proteins produced by living cells, including monoclonal antibodies, soluble receptors, receptor antagonists, novel molecules derived by genetic engineering and other types of proteins that can be used to treat human diseases.
  • Biosimilars may represent a cost-saving alternative to reference biologics.
  • Currently the FDA has not provided details of the kind of testing required to demonstrate sufficient similarity in efficacy and safety for approval.
  • A similar production process does not ensure that the biosimilar is functionally equivalent to a reference biologic. Extensive human testing will be required.
  • Even though the FDA has been establishing standards for licensure to ensure the safety and effectiveness of biosimilars and issued a guidance in February 2012 [7], the FDA has not approved a biological product as biosimilar or interchangeable.
  • Several companies are developing biosimilar products and will almost certainly submit applications for licensure under the new law. It is not yet known when the first biosimilar will be on the U.S. market.
For more evidence-based clinical quality improvement help, visit our website at www.delfini.org.  For more commentaries, visit our DelfiniClick™.

References

1.Generics.
http://www.fda.gov/Drugs/ResourcesForYou/Consumers/
BuyingUsingMedicineSafely/UnderstandingGenericDrugs/ucm144456.htm

Accessed 2/1/12.

2. FDA definitions.
http://www.fda.gov/Drugs/informationondrugs/ucm079436.htm#B

3.BPCI Act.
http://www.fda.gov/Drugs/DevelopmentApprovalProcess/
HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/Biosimilars/ucm241719.htm

Accessed 2/1/12.

4. Subsequent Entry Biologics (SEBs).
http://www.hc-sc.gc.ca/dhp-mps/brgtherap/applic-demande/guides/seb-pbu/notice-avis_seb-pbu_2010-eng.php
Accessed 2/1/12

5.Biosimilars EMA.
http://www.ema.europa.eu/ema/index.jsp?curl=pages/special_topics/document_listing/
document_listing_000318.jsp&murl=menus/special_topics/special_topics.jsp&mid=WC0b01ac0580281bf0

Accessed 2/1/12.

6. American College of Rheumatology. http://www.rheumatology.org/practice/clinical/position/biosimilars.pdf
Accessed 2/1/12.

7. Biosimilars FDA Guidance. http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm259797.htm
Accessed 2/13/12.

Other Sources

8. Colbert RA, Cronstein BN. Biosimilars: the debate continues. Arthritis Rheum. 2011 Oct;63(10):2848-50. doi: 10.1002/art.30505. PubMed PMID: 21702015.

9. Reichert JM, Beck A, Iyer H. European Medicines Agency workshop on biosimilar monoclonal antibodies: July 2, 2009, London, UK. MAbs. 2009 Sep-Oct;1(5):394-416. Epub 2009 Sep 25. PubMed PMID: 20065643.

10. Kozlowski S, Woodcock J, Midthun K, Sherman RB. Developing the nation’s biosimilars program. N Engl J Med. 2011 Aug 4;365(5):385-8. PubMed PMID: 21812668.

 

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email

Reversal of Established Practices—Unringing the Bell

Status

Reversal of Established Practices—Unringing the Bell

Many healthcare practices become established before there is sufficient evidence to conclude that the intervention is beneficial and that benefits outweigh harms. Read more at our DelfiniClick:  Reversal of Established Practices—Unringing the Bell

Facebook Twitter Linkedin Digg Delicious Reddit Stumbleupon Tumblr Email