Understanding breast cancer screening—past, present, and future

Hong Kong Med J 2018;24:Epub 6 Apr 2018
DOI: 10.12809/hkmj177123
REVIEW ARTICLE
Understanding breast cancer screening—past, present, and future
Jacqueline CM Sitt, MB, BS, FHKCR; CY Lui, MB, ChB, FHKCR; Lorraine HY Sinn, MB, BS, FHKCR; Julian CY Fong, MB, BS, FHKCR
Hong Kong Women’s Imaging Limited, Suite 319, 3/F, Central Building, Central, Hong Kong
 
Corresponding author: Dr Jacqueline CM Sitt (jacquelinesitt@gmail.com)
 
 Full paper in PDF
 
Abstract
This article provides an up-to-date overview of breast cancer mammography screening and briefly discusses its history, controversies, current guidelines, practices across Asia, and future directions. An emphasis is made on shared decision-making—instead of giving just a ‘yes’ or ‘no’ answer to patients, the focus should be on providing sufficient information about the pros and cons of screening to help women make a personal, informed choice. Frontline experts, including breast surgeons, oncologists, breast radiologists, and their representative professional associations should all participate in guideline panels, with the goal of improving cancer detection, reducing mortality, and improving patient outcome.
 
 
Introduction
This article provides an up-to-date overview of breast cancer mammography screening and briefly discusses its history, controversies, current guidelines, practices across Asia, and future directions. An emphasis is made on shared decision-making—instead of giving just a ‘yes’ or ‘no’ answer to patients, the focus should be on providing sufficient information about the pros and cons of screening to help women make a personal, informed choice.
 
Goals and advantages of breast cancer screening
The goal of mammographic screening (and other breast-cancer screening tests) is to detect breast cancer earlier than it would otherwise manifest clinically, when it is less likely to have spread. Data clearly show that detection of breast cancers at smaller sizes and lower (earlier) stages is associated with better patient outcomes, lower morbidity, and reduced breast cancer deaths.1 Reduced morbidity is likely to be related to feasibility of breast conservation and hence less extensive surgery, fewer associated complications such as lymphoedema, less chemotherapy, and hence fewer adverse effects.2 Other benefits of diagnosing screen-detected cancers at an earlier stage also include a lower cost of treatment and consequent reduced financial burden on health care resources.3
 
Current guidelines
The Table summarises the mammography guidelines from selected nations.4 5 In common, all organisations emphasise that the benefits of screening outweigh the harm at all ages.3 6 They all endorse informed decision-making and the importance of informing women about both benefits and limitations of screening. However, there remain legitimate concerns about guideline differences, including the complexity of the guidelines; weak adherence to creating opportunities for informed decision-making; unreadiness of referring clinicians to discuss benefits, limitations, and harm associated with screening; and the lack of reminder systems, which results in weaker adherence to recommended screening intervals. Despite these concerns, it is widely accepted that high adherence to even the least aggressive guidelines will save more lives than the current weak adherence to regular screening programmes.4
 

Table. Summary of mammography guidelines from selected nations4 5
 
Current scientific evidence to support screening
Randomised controlled trials (RCTs) have been the gold standard for proving that early detection with mammography decreases mortality from breast cancer. Since the very first screening RCT performed in New York in the 1960s, there have been eight prospective RCTs and numerous subsequent meta-analyses published. Most well-executed RCTs demonstrated a 20% to 30% decrease in mortality from breast cancer when women were invited for screening. These results laid a solid foundation for population-based screening programmes worldwide.1 7 8
 
Subsequent studies that generated data from population-based screening programmes have provided further evidence of the benefits of screening mammography. The true benefit reported (in terms of mortality reduction) ranged from 38% to 49%, even higher than that shown by RCTs. This difference demonstrates that service screening studies measure the direct effect of screening on women who actually underwent mammography, and not just those who were invited to undergo mammography (as opposed to the methodology of RCTs). Service screening studies also tend to measure the effect of more recent screening practices that have benefited from improved mammography technology, better breast positioning techniques, and improved interpretive skills.1 9
 
Understanding screening controversy and ‘mammographic wars’
The Canadian national breast screening study: root of all controversies
One exception to the RCTs that reported unfavourable results of mammographic screening was the Canadian National Breast Screening Study (CNBSS). It was conducted between 1980 and 1985, and was divided into two parts. The first CNBSS included approximately 50 000 volunteer women aged 40 to 49 years, and determined the mortality benefit in the experimental group, who were assigned to annual screening mammography plus clinical breast examination (CBE) versus the control group who received usual care.10 The second CNBSS had almost 40 000 volunteer women aged 50 to 59 years, and compared the benefit of annual mammography plus CBE with that of yearly CBE alone.11
 
From the time the results were first published in 1992 and again after follow-up in 2000, 2002, and 2014, the CNBSS has been controversial, because it is the only RCT to have reported no decrease in mortality associated with an invitation to screening. The study also claimed a 22% overdiagnosis of screen-detected invasive cancer, increasing to up to 35% when cases of ductal carcinoma in situ (DCIS) were included.12 13 14 However, the credibility and scientific value of the CNBSS study have been repeatedly questioned in peer-reviewed publications.15 16 17 18 19 Most criticisms of this study are related to vulnerabilities and shortcomings in its execution, including flaws in the randomisation process, lack of statistical power, non-generalisable results, poor quality imaging, suboptimal mammographic image acquisition and interpretation by untrained personnel, and inconsistent thresholds for interpretation.
 
The flaws in the randomisation process principally arose from three areas. First, unlike all other RCTs, potential participants in the Canadian trials initially underwent a careful physical examination. Second, women with positive findings on physical examination, including palpable lumps, skin or nipple retraction, and even palpable axillary adenopathy, were not excluded from this ‘screening’ trial.18 Finally, randomisation was unblinded and decentralised. Because almost 80% of women with advanced palpable cancers were assigned to the screening arm in the first round of the study, there has been speculation that concerned clinicians did not follow the randomisation process, but rather assigned some symptomatic women to the study group so that they would undergo mammography.19 Whether the imbalance was due to intentional tampering or occurred by chance alone, the net effect was the same—namely, a failure to produce two equal cohorts of patients for comparison.
 
The CNBSS was also criticised at the time of the trial for poor quality mammography, even compared with mammographic imaging of that era.15 20 To reduce radiation dose, mammography for the trial was performed without the benefit of scatter-reducing grids despite their routine use and availability. Standard imaging for much of the trial used a straight lateral view, not a mediolateral-oblique view, which images more tissue. The combination of poor quality imaging and the investigators’ resistance to taking corrective action led two advisors’ resignation in protest. In addition, technologists who participated in the trial received no special training in performing mammography. Radiologists new to mammography also received no training in interpretation.18 There was also a lack of immediate follow-up after recommendations for biopsy had been made. Overall, about 25% of the recommended biopsies were ultimately not performed.18
 
The CNBSS trials are an excellent example of the need to carefully consider all facets of a large-scale screening trial before accepting its results as scientifically valid. The numerous design and execution flaws described above explain in large part why the results of the CNBSS are dramatically different from those of all other RCTs. Ultimately, on the basis of the methodology of the CNBSS, the World Health Organization excluded those results when analysing the breast-screening data in the International Agency for Research on Cancer report.21
 
Controversial meta-analysis results from the Nordic Cochrane Centre
The greatest debate on the value of breast screening arose after the publication of a highly controversial but frequently quoted meta-analysis by Gotzsche (a medical statistician and director of the Nordic Cochrane Centre) and Olsen in The Lancet in 2000. Their study concluded that there was no benefit of mortality reduction by screening, after discarding six of eight RCTs because they deemed the randomisation to be “inadequate”. The only two RCTs included in their analysis showed no benefit, including the Malmo trial and the notorious CNBSS.7 22
 
Gotzsche and Olsen’s critique and methodology have caused much controversy and, in turn, have been criticised heavily by leading expert breast imagers, public health clinicians, and professional bodies such as the Society of Breast Imaging.7 8 23 24 25 26 27 Gotzsche and Olsen’s use of quoted figures from cancer registries rather than actual patient data, their selective approach to studies, and in particular the ignoring of the flaws of the CNBSS, have received the harshest criticism. Many experts have commented that Gotzsche and Olsen overstated the limitations of most of the well-executed RCTs, thereby reflecting a “context-free” application of guidelines in a way that did not address the real issues relevant to the effectiveness of mammographic screening. Moreover, Gotzsche and Olsen’s recommendation to abandon screening altogether has hampered collaborative efforts to improve breast cancer detection and control.27
 
Swiss Medical Board’s decision to stop population-based screening in 2014
In February 2014, the Swiss Medical Board attempted to overturn the widespread practice of mammography screening in Switzerland by stating that new systematic mammography screening programmes should not be introduced, irrespective of women’s age, and recommended that existing programmes should be discontinued. Their main argument was that the absolute risk reduction in breast cancer mortality was low and that the adverse consequences of screening (false-positive test results, overdiagnosis, overtreatment, and high costs and expense of follow-up tests and procedures) were substantial.28 29
 
The Swiss Medical Board’s attempt initiated a new phase of heated arguments and debate about the benefits of screening. Expert breast cancer clinicians in both the United States and Europe (including leading cancer associations in Switzerland) rejected their report. One criticism was that the Swiss Medical Board relied heavily on the controversial work by Gotzsche and Olsen and again quoted data from the flawed CNBSS. Another criticism that attracted great attention was the questionable “expert panels” of the board: they included a medical ethicist, a clinical epidemiologist, a clinical pharmacologist, an oncology surgeon, a nurse scientist, a lawyer, and a health economist. Frontline breast imagers, with expertise in diagnosing breast diseases, were excluded from the review panels because of a “conflict of interest”.28 29
 
The Swiss Medical Board did not adequately consider the fact that assessment of the balance between benefit and harm involves a value judgement that each woman should make only after she is fully informed about the strengths and weaknesses of screening mammography. They also disregarded the extensive literature in support of screening mammography (RCTs and population service screening studies), making their attempt at stopping national mammography screening unjustified.
 
Potential risks of screening overstated
Commonly mentioned potential harms of screening include false-positive mammograms, recall for additional imaging, a false-positive biopsy, missed breast cancer, radiation dose, patient anxiety, and, above all, overdiagnosis.
 
Overdiagnosis is defined as the detection (and subsequent actions taken) of a cancer by screening that would not have progressed to become symptomatic in a woman’s lifetime.1 The estimation of overdiagnosis is complex, highly debated, and very difficult to measure.3 Reported figures range widely, from 0% to 50%, vary greatly in terms of methodological rigour, and testify to the inexact nature of most mathematical models.30 31 32 33 34 When appropriate adjustments for temporal trends, risk factors, and lead time are considered, the level of overdiagnosis should be low, within the range of 0% to 10%.32 Importantly, a recent study of over 5 million women (aged 50-64 years) screened by the United Kingdom’s National Health Service showed that there was a significant negative association between the detection of DCIS at screening and invasive interval cancers. In that study, Duffy and colleagues analysed the data from four consecutive screen years and the 36-month outcome after each relevant screen. For every three screen-detected cases of DCIS, there was one less interval case of invasive cancer over the next 3 years. They agreed that the policy on detection and treatment of DCIS is worthwhile and can prevent subsequent invasive cancers.35
 
The effect of screening on heightening a patient’s anxiety has also been long questioned by critics, but the magnitude of the effect may have been over-exaggerated. In a survey of over 1200 women with a 6-question anxiety scale to understand the short-term and long-term impact of a recall examination, women involved in the digital mammographic imaging screening trial demonstrated only a transient, limited increase in anxiety after a false-positive mammogram compared with those with a negative mammogram, and there was no difference between the two groups’ intention to undergo mammography again in the subsequent 2 years.36 Schwartz et al reported that 96% of American women who received a false-positive mammography report were glad that they underwent the test and remained supportive of screening.37 Most women agreed that the anxiety, inconvenience, and the few image-guided needle biopsies using local anaesthesia associated with a recall from screening, were minor compared with dying of breast cancer.38
 
To summarise, papers citing a high rate of overdiagnosis in screening (in the magnitude of 20% or higher) and claiming that false-positives are a significant cause of patient anxiety are believed by most experts to be overstating the case.
 
Harms of not screening underestimated
Although it is important to discuss all aspects of screening asymptomatic women (including potential harm), the harm of not attending screening is underestimated and not discussed. For instance, women who do not attend screening have significantly larger tumours, a higher stage at diagnosis, poorer overall and disease-specific survival, and higher costs of treatment.39 It has been estimated that the cost of treating advanced metastatic breast cancer exceeds USD 250 000 per patient, and the average cost of treating advanced cancer in the first year after diagnosis is almost double that of early cancers, mainly owing to the difference in costs of chemotherapy.3 40 The cost of treatment and lost productivity each year will far exceed the cost of annual screening and, additionally, do not include the indirect value of the lives saved (as a productive member of workforce).1
 
Situation in Asia
Rising breast cancer incidence: a universal phenomenon among Asian women
The incidence of breast cancer continues to increase worldwide. It remains highest in the United States and Europe, but has been increasing substantially in Asian countries over the past three decades.41 Studies that compare invasive breast cancer data from Asia with those from the United States over a 20-year period have shown that female breast cancer incidence among Asian and Western populations is more similar than expected.42 The incidence of female breast cancer in China will continue to rise, and is expected to exceed 100 per 100 000 women by 2021, giving a total of 2.5 million cases.43
 
According to GLOBOCAN 2012 of the International Agency for Research on Cancer, the specialised cancer research agency of the World Health Organization, almost a quarter (24%) of all breast cancers were diagnosed within the Asia-Pacific region, with the greatest number occurring in China (46%).44 The age-standardised incidence rate was highest among Taiwanese (65.9 per 100 000), followed by Singaporeans, South Koreans, and Japanese.44 In a multiracial country such as Singapore, Chinese women have been noted to have a significantly higher risk of developing breast cancer than Malays and Indians.45
 
The disease burden in Hong Kong is no different. Locally, the age-standardised incidence rate was 58.8 per 100 000 in 2015, with over 3900 new cases per year.46 A study of the local trend in female breast cancer incidence from 1973 to 1999 by the University of Hong Kong showed a significant yearly increase of an average of 3.6%; the increase was most marked and continued to accelerate in the younger age-groups. It was speculated that such trend changes were related to Westernisation of lifestyle.47 All these data indicate that the disease burden in Hong Kong is increasing and comparable to that of all other civilised Asian countries and cities.
 
Breast screening programmes in Asia
Breast screening services in Asian countries and cities are highly variable: some have advanced nationwide screening programmes and others have less developed programmes.48 South Korea and Taiwan are both well recognised for their experience in running such programmes, the former having the highest intake rate and the latter being the most well-structured.
 
South Korea places a very strong emphasis on screening for cancer control in general. Its national health service offers mammography and CBE every 2 years to women aged 40 or older, and at no cost to the 50% of people with the lowest incomes. Their programme is popular and widely accepted by the general public, and achieved an uptake of as high as 66% in 2014. Benefits of downstaging from screening were also observed. However, South Korea encountered a problem of potential overdiagnosis, with a noticeably higher false-positive rate when compared with other places.
 
Taiwan’s health authorities have been recognised for rolling-out well-organised and well-resourced screening programmes, with good support from a local randomised controlled trial showing a reduction in mortality by 40% with mammography screening.49 Since 2004, their health service has provided free breast screening to women aged 50 to 69 years, expanded in 2010 to those aged 40 to 49 years. By 2015, about 40% of the target population participated in screening. It is believed that the cause of the suboptimal participation rate was not due to capacity or outreach, but rather the Taiwanese public’s values and attitude. Nonetheless, with more resources being directed to public education and motivation, Taiwan’s health authorities are pushing their goal to 60% by 2018.
 
The experience of screening programmes in Singapore and Japan is more equivocal. Despite having sufficient scientific evidence to support their role in reducing mortality and reducing invasive cancer incidence, the participation rate has remained lower than expected, mostly owing to cultural barriers and paradigms, or a lack of central governing. Singapore established its national, population-wide screening programme (BreastScreen Singapore) in 2002 and now covers women aged 40 to 69 years. The participation rate has been noted to plateau at 40% since 2010, short of the target of 70%. The health promotion board believes that apart from cultural issues, costs (as screening is paid by an individual’s medical insurance account) constitute the greatest barrier to uptake.
 
The study of population-based screening in Japan has been complex, with scattered data owing to the lack of a single national organisation for monitoring. The participation rate remains lower than in other comparable Asian countries in the past century, again likely because of cultural paradigms. Despite these barriers, in the past decade, Japanese health officials have started designing their own methods and protocols for screening, particularly targeting the higher incidence of cancer among younger women (aged 40-49 years) and the large proportion of patients with dense breasts. After the launch of government-funded screening programmes, a clinical trial that started in 2007 (Japan Strategic Anti-cancer Randomised Trial, J-START) of over 70 000 women undergoing adjunctive ultrasonography to supplement mammography for screening showed an increased sensitivity and detection rate for early preclinical cancers.41
 
In China, there is no nationwide screening programme for breast cancer. A mammographic screening programme was attempted in 2005 but was abandoned because of lack of funding and concerns about false-positive diagnoses. Despite these barriers, national guidelines established in 2007 recommend annual mammography for women aged 40 to 49 years, and every 1 to 2 years for those aged 50 to 69 years. In a Beijing study of 1.46 million women (aged 35 to 59 years) who underwent screening by ultrasonography from 2009 to 2011, the cancer detection rate was 48.0 per 100 000, including 440 cases at early stage that constituted 69.7% of cases detected. The detection rate was lower than anticipated, maybe in part owing to the young age of the screened group and omission of mammography as a screening tool. Subsequently, a second-generation screening programme was initiated in 2012, after modification of the screening methods, cohort size (6 million), and target population that included women aged 35 to 64 years. The new screening procedures include parallel CBE and breast ultrasonography; women with suspicious findings from either examination are recommended to undergo mammographic imaging.50 Although the design of this screening protocol deviates from the standard practice of other countries, we believe that the programme will bring more research data and experience, and eventually lead to more comprehensive guidelines and consensus on a screening approach in China.
 
Breast-screening programmes in Hong Kong: room for development
The awareness of breast cancer and acceptance of screening in Hong Kong is growing, but is still inadequate. According to the latest Breast Cancer Registry Report No 8 (2016), which covers 13 453 breast cancer patients diagnosed from 2006 onwards, the mean and median age of patients at diagnosis was 52.6 and 51.3 years, respectively, and about two-thirds of patients were aged 40 to 59 years. The screening habits among these patients were poor, with over 60% never having undergone mammography screening before their cancer diagnosis.51
 
Although to date there has been no population-based screening for women in Hong Kong, opportunistic screening has long been practised in the private sector. The largest voluntary self-financed and self-referred opportunistic screening programme is run by the Tung Wah Group of Hospitals. In a retrospective review of their performance from 1998 to 2002 involving over 46 600 screening mammograms, a breast cancer detection rate of five cases per 1000 population was noted, which was comparable to the detection rate of Western screening programmes at that time.52
 
Regarding the input of expertise and quality assurance, the Hong Kong College of Radiologists issued their mammographic statement in 2006 (latest revision in 2015).53 Quoting desirable goals recommended by the United Kingdom and United States as a reference the statement sets specific benchmarks for standards of mammographic machines, quality of screening mammograms, radiation dose limits, and accreditation requirements of reporting radiologists.53 Given these guidelines, together with recent advances in mammographic technology, we believe that there should be room for further local development of large-scale quality breast-screening programmes.
 
Designing a screening programme for Hong Kong: can there be a protocol tailor-made for Chinese women?
When planning a breast-screening programme, it is necessary to decide whom to screen (ie, at what age and the target screening population) and how to screen (ie, screening method).
 
For the decision of whom to screen, we should note that the mean age at diagnosis of breast cancer in Chinese women is 45 to 55 years, considerably younger than for western women.43 Starting screening at age 40 or 45 years would likely be a better fit for Chinese women than starting at age 50 years, as recommended by some western guidelines. As for the target screening population, current data favour universal screening over risk-based screening (pre-selecting patients according to risk profile). First, one should note that 80% of women with newly diagnosed breast cancer have no family history (ie, first-degree relative) or other significant previous risk factors, and therefore risk-based screening will miss a majority of screen-detected breast cancers.3 54 Second, a recent 10-year population-based cohort study of over 1.4 million asymptomatic Taiwanese women undergoing various breast-cancer screening regimens showed that universal mammography screening based only on age and sex was more effective than other screening regimens (risk-based biennial mammography screening or annual CBE alone).49 In that study, universal biennial mammography screening was associated with a 41% reduction in mortality and a rate of overdiagnosis of only 13%. In contrast, risk-based biennial mammography (pre-selecting patients according to risk profile or risk score) did not lead to any statistically significant reduction in mortality. Moreover, among all screening regimens, only universal biennial screening was associated with a clear downstaging shift in tumours (30% reduction of stage 2+ cancers), a crucial factor that can improve patient outcome.49
 
Regarding methods of screening, conventional screening uses standard two-view full-field digital (two-dimensional; 2D) mammography. Multiple studies have proven that screening by digital breast tomosynthesis (DBT; also called three-dimensional mammography) can increase cancer detection rates compared with 2D mammography alone, and can reduce the recall rate for benign findings (false-positives). 1 55 A retrospective analysis of over 454 000 screens showed that use of DBT was associated with relative increases of 41% in invasive cancer detection, 49% in positive predictive value (PPV) for recall, and 21% in PPV for biopsy, in addition to a 15% reduction in the overall number of recalls.56 A recent meta-analysis by a Korean group also showed that screening with DBT increased detection of early invasive cancers of <2 cm.57 The American College of Radiology Commission on Breast Imaging now recommends that mammography and DBT are “usually appropriate” for screening of average-risk women, noting that DBT addresses some limitations of standard digital mammography.58 In Hong Kong, DBT has been increasingly adopted to replace or serve as an adjunct to 2D mammography in opportunistic screening. We anticipate that the shift to DBT screening will become a global trend.
 
The use of whole-breast ultrasonography to screen dense breasts is also commonly adopted in Asia, including for opportunistic screening in Hong Kong. In Japan, this practice was reinforced by a government-funded RCT (J-START) that studied the use of adjunctive ultrasonography to supplement mammography in screening over 70 000 women. The J-START study showed favourable results of increased sensitivity and detection rate for early, preclinical cancers.41
 
Screening for high-risk women is often considered a separate entity. According to the American College of Radiology’s Appropriateness Criteria, women at high risk due to prior mantle radiation between the ages of 10 and 30 years should start mammography 8 years after radiation therapy, but not before age 25. For women with a genetic predisposition, annual screening mammography is recommended to begin 10 years earlier than the age that an affected relative had been diagnosed, but not before age 30. Annual screening by magnetic resonance imaging is recommended in high-risk women as an adjunct to mammography.59
 
Future directions for Hong Kong
We believe that health care in Hong Kong should have the capability and expertise to roll out quality, large-scale population-screening programmes that are comparable to those in other developed Asian countries and cities. When we examine the common themes among available guidelines, literature, and expert reviews worldwide, the global trend is to provide women with an informed choice.
 
In the discussion of whether breast-cancer screening is feasible, one should bear in mind that this is an emotive issue. Apart from the critical appraisal of scientific evidence, the interpretation of literature and subsequent formulation of recommendations should always account for the socioeconomic, historical, and contextual realities. The value judgement of women should also be respected.
 
Frontline experts, including breast surgeons, oncologists, breast radiologists, and their representative professional associations should all participate in guideline panels, with a will to end the ‘mammography wars’. Our Holy Grail should always be focused on improving cancer detection, reducing mortality, and improving patient outcome.
 
References
1. Eby PR. Evidence to support screening women annually. Radiol Clin North Am 2017;55:441-56.
2. Society of Breast Imaging Breast Screening Leadership Group. Screening in the 40-49 Age Group. Available from: https://www.sbi-online.org/RESOURCES/BreastScreeningLeadershipGroupResources.aspx. Accessed 19 Nov 2017.
3. Ray KM, Price ER, Joe BN. Evidence to support screening women in their 40s. Radiol Clin North Am 2017;55:429-39.
4. The American College of Radiology. Guidelines for Breast Cancer Screening—An Update. SBI Breast Imaging Symposium 2016. Available from: https://www.sbi-online.org/Portals/0/Breast%20Imaging%20Symposium%202016/Final%20Presentations/4-7%20800am%20Smith%20-%20Guidelines%20for%20Breast%20Cancer%20Screening.pdf. Accessed 19 Nov 2017.
5. Shieh Y, Eklund M, Sawaya GF, et al. Population-based screening for cancer: hope and hype. Nat Rev Clin Oncol 2016;13:550-65. CrossRef
6. Byrne SK. What's the Buzz: Tell me what’s happening in breast cancer screening. Asia Pac J Oncol Nurs 2017;4:122-6. CrossRef
7. Freer P, Moy L, DeMartini W; the Screening Leadership Group. Breast Cancer Screening: Understanding the Randomised Controlled Trials. Available from: https://www.sbi-online.org/Portals/0/Screening%20Leaders/Breast%20Cancer%20Screening_Understanding%20the%20Randomized%20Controlled%20Trials.pdf. Accessed 19 Nov 2017.
8. Duffy SW, Chen TH, Smith RA, Yen AM, Tabar L. Real and artificial controversies in breast cancer screening. Breast Cancer Manage 2013;2:519-28. CrossRef
9. Newell M, Eby PR; the Breast Screening Leadership Group. Benefits of Screening Mammography: Data from Population Service Screening. Available from: https://www.sbi-online.org/Portals/0/Screening Leaders/Benefitsof Screening Mammography_Data from PopulationService Screening.pdf. Accessed 19 Nov 2017.
10. Miller AB, Baines CJ, To T, et al. Canadian National Breast Screening Study: 2. Breast cancer detection and death rates among women aged 50 to 59 years. CMAJ 1992;147:1477-88.
11. Miller AB, Baines CJ, To T, et al. Canadian National Breast Screening Study: 1. Breast cancer detection and death rates among women aged 40 to 49 years. CMAJ 1992;147:1459-76.
12. Miller AB, To T, Baines CJ, et al. Canadian National Breast Screening Study-2: 13-year results of a randomized trial in women aged 50-59 years. J Natl Cancer Inst 2000;92:1490-9. CrossRef
13. Miller AB, To T, Baines CJ, et al. The Canadian National Breast Screening Study-1: breast cancer mortality after 11 to 16 years of follow-up. A randomized screening trial of mammography in women age 40 to 49 years. Ann Intern Med 2002;137:305-12. CrossRef
14. Miller AB, Wall C, Baines CJ, et al. Twenty five year follow-up for breast cancer incidence and mortality of the Canadian National Breast Screening Study: randomised screening trial. BMJ 2014;348:g366. CrossRef
15. Kopans DB, Feig SA. The Canadian National Breast Screening Study: a critical review. AJR Am J Roentgenol 1993;161:755-60. CrossRef
16. Burhenne LJ, Burhenne HJ. The Canadian National Breast Screening Study: a Canadian critique. AJR Am J Roentgenol 1993;161:761-3. CrossRef
17. Kopans D. Breast Imaging. 3rd ed. Lippincott Williams & Wilkins: Philadelphia; 2007.
18. Heywang-Köbrunner SH, Schreer I, Hacker A, et al. Conclusions for mammography screening after 25-year follow-up of the Canadian National Breast Cancer Screening Study (CNBSS). Eur Radiol 2016;26:342-50. CrossRef
19. Boyd NF. The review of randomization in the Canadian National Breast Screening Study. Is the debate over? CMAJ 1997;156:207-9.
20. Baines CJ, Miller AB, Kopans DB, et al. Canadian National Breast Screening Study: assessment of technical quality by external review. AJR Am J Roentgenol 1990;155:743-7; discussion 8-9. CrossRef
21. International Agency for Cancer on Research (IARC), World Health Organization. IARC Handbooks of Cancer Prevention. Volume 7: Breast Cancer Screening. IARC Press; 2002. Available from: http://www.iarc.fr/en/publications/pdfs-online/prev/handbook7/Handbook7_Breast-4.pdf. Accessed 15 Mar 2018.
22. Gotzsche PC, Olsen O. Is screening for breast cancer with mammography justifiable? Lancet 2000;355:129-34. CrossRef
23. Tabár L, Dean PB, Cen TH, et al. The impact of mammography screening on the diagnosis and management of early-phase breast cancer. In: Francescatti D, Silverstein M, editors. Breast Cancer: A New Era in Management. Springer New York; 2014: 31-78. CrossRef
24. Bock K, Borisch B, Cawson J, et al. Effect of population-based screening on breast cancer mortality. Lancet 2011;378:1775-6. CrossRef
25. Patnick J, Perry N, de Wolf C. Effect of population-based screening on breast cancer mortality—Authors’ reply. Lancet 2012;379:1298.
26. Mcneil DG. Confronting cancer: scientist at work—Peter Gotzsche; A career that bristles with against-the-grain conclusions. Available from: http://www.nytimes.com/2002/04/09/science/confronting-cancer-scientist-work-peter-gotzsche-career-that-bristles-with.html. Accessed 19 Nov 2017.
27. Cuzick J. Breast cancer screening—time to move forward. Lancet 2012;379:1289-90. CrossRef
28. Chiolero A, Rodondi N. Lessons from the Swiss Medical Board recommendation against mammography screening programs. JAMA Intern Med 2014;174:1541-2. CrossRef
29. Biller-Andorno N, Juni P. Abolishing mammography screening programs? A view from the Swiss Medical Board. N Engl J Med 2014;370:1965-7. CrossRef
30. Helvie MA, Chang JT, Hendrick RE, et al. Reduction in late-stage breast cancer incidence in the mammography era: Implications for overdiagnosis of invasive cancer. Cancer 2014;120:2649-56. CrossRef
31. Kopans DB, Smith RA, Duffy SW. Mammographic screening and “overdiagnosis”. Radiology 2011;260:616-20. CrossRef
32. Puliti D, Duffy SW, Miccinesi G, et al. Overdiagnosis in mammographic screening for breast cancer in Europe: a literature review. J Med Screen 2012;19 Suppl 1:42-56. CrossRef
33. Duffy SW, Agbaje O, Tabar L, et al. Overdiagnosis and overtreatment of breast cancer: estimates of overdiagnosis from two trials of mammographic screening for breast cancer. Breast Cancer Res 2005;7:258-65. CrossRef
34. Bleyer A, Welch HG. Effect of three decades of screening mammography on breast-cancer incidence. N Engl J Med 2012;367:1998-2005. CrossRef
35. Duffy SW, Dibden A, Michalopoulos D, et al. Screen detection of ductal carcinoma in situ and subsequent incidence of invasive interval breast cancers: a retrospective population-based study. Lancet Oncol 2016;17:109-14. CrossRef
36. Tosteson AN, Fryback DG, Hammond CS, et al. Consequences of false-positive screening mammograms. JAMA Intern Med 2014;174:954-61. CrossRef
37. Schwartz LM, Woloshin S, Fowler FJ, Jr, et al. Enthusiasm for cancer screening in the United States. JAMA 2004;291:71-8. CrossRef
38. Kopans DB. An open letter to panels that are deciding guidelines for breast cancer screening. Breast Cancer Res Treat 2015;151:19-25. CrossRef
39. Duffy SW, Chen TH, Smith RA, Yen AM, Tabar L. Real and artificial controversies in breast cancer screening. Breast Cancer Manage 2013;2:519-28. CrossRef
40. Montero AJ, Eapen S, Gorin B, et al. The economic burden of metastatic breast cancer: a U.S. managed care perspective. Breast Cancer Res Treat 2012;134:815-22. CrossRef
41. Ohuchi N, Ishida T, Kawai M, et al. Randomized controlled trial on effectiveness of ultrasonography screening for breast cancer in women aged 40-49 (J-START): research design. Jpn J Clin Oncol 2011;41:275-7. CrossRef
42. Sung H, Rosenberg PS, Chen WQ, et al. Female breast cancer incidence among Asian and Western populations: more similar than expected. J Natl Cancer Inst 2015;107. CrossRef
43. Fan L, Strasser-Weippl K, Li JJ, et al. Breast cancer in China. Lancet Oncol 2014;15:e279-89. CrossRef
44. Youlden DR, Cramb SM, Yip CH, et al. Incidence and mortality of female breast cancer in the Asia-Pacific region. Cancer Biol Med 2014;11:101-15.
45. Singapore Health Promotion Board. National Registry of Diseases Office. Singapore Cancer Registry Interim Annual Report: Trends in cancer incidence in Singapore 2010-2014. Available from: https://www.nrdo.gov.sg/docs/librariesprovider3/default-document-library/cancer-trends-2010-2014_interim-annual-report_final-%28public%29.pdf?sfvrsn=0. Accessed 19 Nov 2017.
46. Hospital Authority, Hong Kong. Hong Kong Cancer Registry. Available from: http://www3.ha.org.hk/cancereg/. Accessed 19 Mar 2018.
47. Leung GM, Thach TQ, Lam TH, et al. Trends in breast cancer incidence in Hong Kong between 1973 and 1999: an age-period-cohort analysis. Br J Cancer 2002;87:982-8. CrossRef
48. Breast cancer in Asia—The Challenge and Response. A Report from the Economist Intelligence Unit. Available from: https://www.eiuperspectives.economist.com/sites/default/files/EIU Breast Cancer in Asia_Final.pdf. Accessed 19 Nov 2017.
49. Yen AM, Tsau HS, Fann JC, et al. Population-based breast cancer screening with risk-based and universal mammography screening compared with clinical breast examination: a propensity score analysis of 1 429 890 Taiwanese women. JAMA Oncol 2016;2:915-21. CrossRef
50. Song QK, Wang XL, Zhou XN, et al. Breast cancer challenges and screening in China: lessons from current registry data and population screening studies. Oncologist 2015;20:773-9. CrossRef
51. A Hong Kong Breast Cancer Foundation Initiative. Hong Kong Breast Cancer Registry Report No. 8, 2016. Available from: http://www.hkbcf.org/download/bcr_report8/hkbcf_report_2016_full_report.pdf. Accessed 19 Nov 2017.
52. Lui CY, Lam HS, Chan LK, et al. Opportunistic breast cancer screening in Hong Kong; a revisit of the Kwong Wah Hospital experience. Hong Kong Med J 2007;13:106-13.
53. Hong Kong College of Radiologists Mammography Statement. Available from: https://www.hkcr.org/templates/OS03C00336/case/lop/HKCR MammographyStatement_rev20150825.pdf. Accessed 19 Nov 2017.
54. Destounis SV, Arieno AL, Morgan RC, et al. Comparison of breast cancers diagnosed in screening patients in their 40s with and without family history of breast cancer in a community outpatient facility. AJR Am J Roentgenol 2014;202:928-32. CrossRef
55. Rafferty EA, Rose SL, Miller DP, et al. Effect of age on breast cancer screening using tomosynthesis in combination with digital mammography. Breast Cancer Res Treat 2017;164:659-66. CrossRef
56. Friedewald SM, Rafferty EA, Rose SL, et al. Breast cancer screening using tomosynthesis in combination with digital mammography. JAMA 2014;311:2499-507. CrossRef
57. Yun SJ, Ryu CW, Rhee SJ, et al. Benefit of adding digital breast tomosynthesis to digital mammography for breast cancer screening focused on cancer characteristics: a meta-analysis. Breast Cancer Res Treat 2017;164:557-69. CrossRef
58. Monticciolo DL, Newell MS, Hendrick RE, et al. Breast cancer screening for average-risk women: recommendations from the ACR commission on breast imaging. J Am Coll Radiol 2017;14:1137-43. CrossRef
59. Expert Panel on Breast Imaging; Mainiero MB, Moy L, Baron P, et al. ACR Appropriateness Criteria Breast Cancer Screening. J Am Coll Radiol 2017;14:S383-90. CrossRef

Reduction of operative mortality after implementation of Surgical Outcomes Monitoring and Improvement Programme by Hong Kong Hospital Authority

Hong Kong Med J 2018;24:Epub 6 Apr 2018
DOI: 10.12809/hkmj177118
ORIGINAL ARTICLE
Reduction of operative mortality after implementation of Surgical Outcomes Monitoring and Improvement Programme by Hong Kong Hospital Authority
WC Yuen, FHKAM (Surgery)1; K Wong, MSc2; YS Cheung, FHKAM (Surgery)3; Paul BS Lai, FHKAM (Surgery)3
1 Department of Surgery, Ruttonjee and Tang Shiu Kin Hospital, Hospital Authority, Hong Kong
2 Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
3 Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
 
Corresponding author: Prof Paul BS Lai (paullai@surgery.cuhk.edu.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Since 2008, the Hong Kong Hospital Authority has implemented a Surgical Outcomes Monitoring and Improvement Programme (SOMIP) at 17 public hospitals with surgical departments. This study aimed to assess the change in operative mortality rate after implementation of SOMIP.
 
Methods: The SOMIP included all Hospital Authority patients undergoing major/ultra-major procedures in general surgery, urology, plastic surgery, and paediatric surgery. Patients undergoing liver or renal transplantation or who had multiple trauma or massive bowel ischaemia were excluded. In SOMIP, data retrieval from the Hospital Authority patient database was performed by six full-time nurse reviewers following a set of precise data definitions. A total of 230 variables were collected for each patient, on demographics, preoperative and operative variables, laboratory test results, and postoperative complications up to 30 days after surgery. Mortality at 30 days, 60 days, and 90 days were also retrieved from the HA electronic database. Based on cumulative 5-year data, risk-adjusted 30-day mortality models were generated by hierarchical logistic regression for emergency and elective operations. The models expressed overall performance as an annual observed-to-expected mortality ratio.
 
Results: From 2009/2010 to 2015/2016, the overall crude mortality rate decreased from 10.8% to 5.6% for emergency procedures and from 0.9% to 0.4% for elective procedures. From 2011/2012 to 2015/2016, the risk-adjusted observed-to-expected mortality ratios showed a significant downward trend for both emergency and elective operations: from 1.126 to 0.796 and from 1.150 to 0.859, respectively (Mann-Kendall statistic = –0.8; P<0.05 for both).
 
Conclusion: The Hospital Authority’s overall crude mortality rates and risk-adjusted observed-to-expected mortality ratios for emergency and elective operations significantly declined after SOMIP was implemented.
 
 
New knowledge added by this study
  • A Surgical Outcomes Monitoring and Improvement Programme allows monitoring of performance and fair comparison of individual Hospital Authority hospitals against the overall Hospital Authority average. It enhances the understanding of surgical performance and helps identify areas for improvement.
Implications for clinical practice or policy
  • A properly organised, risk-adjusted clinical audit can accurately measure surgical outcomes and provide information for surgeons to deliver quality improvement.
 
 
Introduction
Audits of surgical mortality are used worldwide to monitor surgical outcome and achieve quality assurance.1 By measuring and comparing properly collected, risk-adjusted surgical outcome data, quality of surgical care could be enhanced in participating institutions.2 It has been demonstrated in several countries that adoption of a national surgical audit programme can reduce mortality.2 3
 
The Hong Kong Hospital Authority (HA) was established in 1991. It is a government statutory body responsible for the management of 42 public hospitals and institutions, 47 specialist out-patient clinics, and 73 general out-patient clinics in Hong Kong. Seventeen HA hospitals have surgical departments; all of them provide an elective surgery service and 14 also provide an emergency surgical service. In 2016, over 80% of all hospital admissions in Hong Kong were under the management of the HA.4 Therefore, it is important for the HA to develop tools with which to measure and improve performance. For this purpose, the HA Coordinating Committee of Surgery set up a Central Surgical Audit Unit in 2001. From 2002 to 2007, the unit conducted clinical audits based on retrospective cumulative data to compare the performance of the 17 surgical departments. One to two major or ultra-major operations, such as major hepatectomy, oesophagectomy, and major lung resection, were selected each year for comparison. The risk-adjustment model used was based on the Physiological and Operative Severity Score for Enumeration of Mortality and Morbidity (POSSUM).5 6 Those audits were procedure-based, mainly focus on mortality, and hence a more comprehensive risk-adjusted outcome audit was needed.
 
In 2008, a new programme—namely, the Surgical Outcomes Monitoring and Improvement Programme (SOMIP)—was launched by the HA with the aim of monitoring and improving surgical quality. The programme was governed by a steering committee comprising surgeons, an anaesthetist, a physician, administrative managers, and statisticians. Risk-adjusted mortality and morbidity rates of elective and emergency major and ultra-major operations at each hospital were measured and reported yearly. Annual forum was held to disseminate the results and allow sharing of best practice.
 
This study aimed to assess the changes in overall surgical outcomes for patients after implementation of SOMIP.
 
Methods
Between July 2008 and June 2016, SOMIP captured data of all elective and emergency major/ultra-major operations (except those in children younger than 1 year) that were performed by general surgery, urology, plastic and reconstructive surgery, and paediatric surgery teams at all 17 HA hospitals. A total of 230 variables were collected from each patient: 10 patient demographic variables, 83 preoperative and operative variables, 31 laboratory test results, and 40 postoperative events and 66 postoperative adverse events in the first 30 days after surgery. Demographic data and laboratory test results were mostly automatically retrieved from various HA clinical information systems. For data that required manual retrieval, six full-time SOMIP nurse reviewers were employed by the HA head office to perform the tasks. Preoperative and operative variables, as well as postoperative complications occurring up to 30 days after the index operation, were retrieved from patient records by the SOMIP nurse reviewers. Mortality at 30 days, 60 days, and 90 days were also retrieved from the HA electronic database. These data were endorsed and submitted by each surgical department’s surgical supervisor within 60 days of surgery.
 
Both the manually captured and automatically captured data were entered into a tailor-made SOMIP electronic database. Data variable definitions were listed in the operation manual of the programme. To ensure data validity and consistency, all nurse reviewers completed comprehensive training on data definition and criteria, and regular nurse reviewer meetings were held to clarify any queries. All data were endorsed by the surgical supervisor of the respective surgical department. When necessary, data definitions were modified.
 
An inter-rater reliability test was performed each May and completed within a month so as to ensure consistency among nurse reviewers as well as data accuracy. Fifty cases were sampled for evaluation using a stratified systematic sampling method. For those selected cases, an independent nurse from the SOMIP team repeated the data collection using a designated data template, without prior knowledge of the information recorded by the original nurse reviewer. Data quality was measured by comparing two sets of data, and inferred by a score defined as the percentage of agreement between nurse reviewer and the SOMIP working team for each data item. The mean score of all data items was used to assess overall performance. The overall result was satisfactory and the mean score of all data items was 99.3% (range of individual item scores, 95.2%-100%).
 
Among the variables collected, preoperative risk factors including demographic data; general health and lifestyle variables; and major respiratory, cardiovascular, hepatobiliary, renal, vascular, central nervous system, and immune co-morbidities were deemed particularly important. These risk factors were modified from those in the American College of Surgeons National Surgical Quality and Improvement Program (NSQIP) to suit the local context.7 8 9 Operative variables included intra-operative blood loss, American Society of Anesthesiologists classification, procedure complexity score, surgical subspecialty, wound classification, operative magnitude, and operative time. Before analysis and reporting, SOMIP data were cleaned and verified by four surgeons and the SOMIP surgical supervisors of each hospital. Questionable cases were reviewed accordingly. Cases of liver transplantation, renal transplantation, multiple trauma, and major bowel ischaemia (Table 1)10 11 12 13 14 15 16 17 were excluded from the risk-adjusted model analysis.
 

Table 1. No. of cases excluded and reasons, from 2008/2009 to 2015/201610 11 12 13 14 15 16 17
 
Comparative risk-adjusted models (different models for emergency operations and elective operations) were generated using hierarchical logistic regression. The 30-day risk-adjusted mortality models expressed hospital performance as expected odds ratios. A risk-adjusted observed-to-expected mortality ratio (O/E ratio) was then calculated for each hospital. The O/E ratio is a quotient between the observed number of deaths and the expected number of deaths; the latter was calculated by a logistic regression method based on significant independent risk factors. Together with the 90% confidence intervals, O/E ratios were depicted by caterpillar plots and boxplots. Hospitals with the lower limit of the 90% confidence interval of O/E ratios greater than 1 were defined as ‘high outliers’; hospitals with the upper limit of the 90% confidence interval of O/E ratios lower than 1 were defined as ‘low outliers’. The risk-adjusted outcome of a ‘high-outlier’ hospital was probably worse than the average outcome, and that of a ‘low-outlier’ hospital was probably better than the average outcome.
 
In addition to risk-adjusted postoperative mortality, various general medical and surgical complications, as well as specific complications (anastomotic leakage, surgical site infection, acute myocardial infarction, pneumonia), were recorded. The list of complications recorded and the method to derive the SOMIP risk-adjustment model have been described in detail in the annual SOMIP Report.10 11 12 13 14 15 16 17 Different levels of confidence were used for different outcome variables—90% confidence interval for mortality rates, 95% confidence interval for major complications, and 99% confidence interval for morbidity rates. Using SOMIP data together with other useful information extracted from the HA Executive Information System (eg, bed occupancy, nursing manpower, intensive care unit support, and surgeon workload), root-cause analyses were performed using multilevel logistic regression, as described in the annual SOMIP Report.10 11 12 13 14 15 16 17
 
The discriminative power of the risk-adjusted models was measured by the C-index, area under the receiver-operating characteristic curve (AUC). The closer the C-index is to 1, the better the discriminative power of the model is: a C-index of ≥0.8 indicates excellent discriminative power.18 The calibration accuracy of the models was assessed by the Hosmer-Lemeshow goodness-of-fit test (HL test). The calibration of the model was rejected if P<0.05. The Mann-Kendall non-parametric trend test was used to identify trends (positive or negative) in the annual data series for both crude mortality rates (2009/2010 to 2015/2016) and risk-adjusted O/E ratios (2011/2012 to 2015/2016). A very high positive value of the Mann-Kendall statistic (S) indicated an increasing trend; a very low negative value indicated a decreasing trend. The test statistic Z-score was used as a measure of trend significance.
 
Results
Descriptive data
Age distribution trends are summarised in Table 2.10 11 12 13 14 15 16 17 From 2008/2009 to 2015/2016, the proportion of people aged 61-70 years increased by 7 percentage points (from 19% to 26%), whereas the proportions of people aged 41-50 years and 71-80 years decreased by 2 and 5 percentage points, respectively.
 

Table 2. Distribution of patients by age-group, from 2008/2009 to 2015/2016.10 11 12 13 14 15 16 17
 
Table 310 11 12 13 14 15 16 17 shows proportions of patients taking regular medication for diabetes mellitus and hypertension before surgery from 2009/2010 to 2015/2016, as well as those currently smoking (within 1 year) and drinking more than 2 units of alcohol per day in the previous 2 weeks. Over 70% of patients had at least one of these four conditions, whereas about 40% had a history of regular use of hypertension medication before surgery.
 

Table 3. Distribution of patients by habit before surgery/admission, from 2009/2010 to 2015/201610 11 12 13 14 15 16 17
 
Annual numbers of elective operations by specialty are summarised in Table 4.10 11 12 13 14 15 16 17 For the 10 listed elective operations, the most frequently performed were in urology, consistently constituting 28% of the caseload from 2008/09 to 2015/16. The least frequently performed procedures were parotid surgery (1%) and paediatric surgery (1%).
 

Table 4. Elective operations by specialty, from 2008/2009 to 2015/201610 11 12 13 14 15 16 17
 
Overall crude mortality rates and risk-adjusted observed-to-expected mortality ratios
From July 2008 to June 2016, eight SOMIP reports were published.10 11 12 13 14 15 16 17 They showed that the HA overall crude mortality rate approximately halved over this time. The crude 30-day mortality rate for emergency operations dropped gradually from 10.8% in the year 2009/2010 to 5.6% in 2015/2016 (Fig 1). Similarly, the crude 30-day mortality rate for elective operations more than halved: from 0.9% in 2009/2010 to 0.4% in 2015/2016 (Fig 2).
 

Figure 1. Crude 30-day mortality rates for emergency operations, 2009/2010 to 2015/2016
 

Figure 2. Crude 30-day mortality rates in elective operations, 2009/2010 to 2015/2016
 
In the 5-year cumulative comparison analysis (2011/2012 to 2015/2016), both models had excellent discriminative power and good calibration accuracy. For emergency operations, the AUC was >0.9 and the HL test statistic was >0.1; for elective operations, the AUC was >0.89 and the HL test statistic was >0.2. The risk-adjusted observed-to-expected 30-day mortality ratio for both types of surgery showed a similar downward trend to the crude mortality rates. For emergency operations, the risk-adjusted O/E ratios were 1.126, 1.022, 1.113, 0.921, and 0.796 across the 5 years (Fig 3). These values show a statistically significant downward (negative) trend (S= –0.8; P<0.05). The results of the Mann-Kendall analyses are summarised in Table 4. For elective operations, risk-adjusted O/E ratios were 1.150, 1.229, 0.881, 0.862, and 0.859 across the 5 years (Fig 4). These values show a statistically significant downward (negative) trend (S= –0.8; P<0.05) [Table 5].
 

Figure 3. Risk-adjusted observed-to-expected 30-day mortality ratio for emergency operations, 2011/2012 to 2015/2016
 

Figure 4. Observed-to-expected 30-day mortality ratio for elective operations, 2011/2012 to 2015/2016
 

Table 5. Mann-Kendall non-parametric trend test results
 
Discussion
Before the turn of the century, most hospital records in HA hospitals were handwritten and retained by individual hospitals. There was no convenient means by which to share patient details among hospitals. Around 2001, the HA implemented a number of clinical management electronic systems, such as the electronic patient record, Operation Theatre Record System, and Clinical Data Analysis and Reporting System, at all HA hospitals. By virtue of this infrastructure, patient records and information about diagnoses and operations could be accessed at a central level. Based on this central clinical database, a Quality Assurance Subcommittee under the Coordinating Committee of Surgery commenced small-scale comparative clinical audits for ultra-major operations in 2002, focusing on one to two ultra-major operations per year. The audits provided basic information about hospital performance for the selected operation, such as number of procedures, age distribution of patients, and mortality rate.
 
Subsequently, the Coordinating Committee of Surgery developed a more robust system to monitor more major operations at the same time. It was decided to follow the framework of NSQIP, which was developed in 1994 by the Veterans Affairs Hospitals in the United States to monitor risk-adjusted surgical operation outcomes. Like HA hospitals, Veterans Affairs Hospitals are managed by a central governing body and equipped with a comprehensive electronic medical records system. Studies19 20 showed a significant improvement in both mortality and morbidity over time and thus, in 2004, NSQIP was extended to private hospitals and endorsed by the American College of Surgeons. Using NSQIP as the blueprint, the HA launched SOMIP in 2008. The SOMIP adopted similar risk-adjustment variables, use of nurse reviewers to collect data, a focus on hospital performance rather than individual surgeon performance, and similar methods of data analysis and determination of outliers. Moreover, SOMIP allowed individual HA hospitals to benchmark their performance against the overall HA average.
 
As all surgical patients have a different health status, their operation outcomes will likewise differ. Appropriate adjustment for different patient risks is essential when interpreting hospital mortality rates. To adjust for different risk factors, over 100 patient risk factors were captured for each enrolled patient. For NSQIP, one risk-adjustment mortality model was constructed for all operations. In contrast to NSQIP, separate models were devised for emergency and elective operations in SOMIP.
 
Hospital outliers can be identified by O/E ratios if the confidence interval of the O/E ratio is greater than 1, meaning that after balancing the different risks of hospital patients, their clinical outcomes are most likely different from the rest. From the results of this study, it was encouraging to find a significant trend of reduction in crude mortality rates and O/E ratios for both elective and emergency operations at HA hospitals over the past 5 years. According to the significant reduction in expected odds ratios over the years, this improvement is genuine and not due to patient selection.
 
There are several possible reasons behind the changes: public identification as a poor performer is a strong incentive for change in HA hospitals; sharing best practices on perioperative patient care in the SOMIP Forum is an important educational activity; investing more resources from the HA Head Office into deficient hospitals; and changing attitudes towards managing surgical complications by other colleagues from the intensive care unit are also helpful. All of these may have contributed to the change.
 
Limitations
The SOMIP has a number of limitations. The coverage of SOMIP is not as complete as that of NSQIP, since many surgical departments such as orthopaedics and neurosurgery are not included. Monitoring is done by retrospective annual case collection (from 1 July to 30 June) because it takes 13 months to complete case enrolment and an additional 3 months for data verification, model building, and statistical analysis. Because of the small number of events, the current programme is not able to determine the risk-adjusted outcomes of individual operations or surgeons. Furthermore, because this programme relies heavily on the HA central electronic database, it is not easy to extend it to hospitals without this information infrastructure. Although the trend of reduction in mortality was statistically significant, we were not able to demonstrate a causal relationship with SOMIP implementation.
 
Potential issues with data quality may have affected the outcomes. In the inter-rater reliability test, the nurse reviewers were not blinded and this may have caused information bias. Also, quality of data collection in the initial 2 years may have been unreliable. As a result, the 5-year cumulative comparison analysis for emergency and elective operations commenced from 2011/2012, rather than 2008/2009. Furthermore, data definitions are updated regularly in the operation manual and could have affected the time trend analysis. Nonetheless, the SOMIP team considered changes in data definitions to be minor and did not expect a significant impact on the risk models.
 
Mortality could be influenced by many factors; ensuring risk adjustments are adequate and appropriate would be a challenge. Disease factors, stage of disease, and treatment options may not be fully taken into account by the risk-adjusted models, and data readiness and availability are further constraints. Surgeon skill and experience was another aspect that could not be accommodated and was difficult to adjust for. In the HA, surgical operations are performed by a team; therefore, it would be difficult to separate individual surgeon experience and credentials from those of the whole team.
 
Conclusion
From 2008 to 2016, the HA’s overall crude mortality rates and risk-adjusted O/E ratios showed a significant trend of reduction for both emergency and elective operations. The SOMIP enhances understanding of surgical performance and helps identify areas for improvement. It allows individual HA hospitals to benchmark their performance against the overall HA average through risk-adjusted O/E ratios.
 
Acknowledgements
We thank the SOMIP Steering Committee of the Hospital Authority, the Coordinating Committee of Surgery, and the Biostatistics team of The Chinese University of Hong Kong for their contributions and helpful comments on this manuscript.
 
Declaration
The authors have no conflicts of interest to disclose.
 
References
1. Lui CW, Boyle FM, Wysocki AP, et al. How participation in surgical mortality audit impacts surgical practice. BMC Surg 2017;17:42. CrossRef
2. Khuri SF, Daley J, Henderson W, et al. Risk adjustment of the postoperative mortality rate for the comparative assessment of the quality of surgical care: results of the National Veterans Affairs surgical risk study. J Am Coll Surg 1997;185:315-27. CrossRef
3. Kiermeier A, Babidge WJ, McCulloch GA, Maddern GJ, Watters DA, Aitken RJ. National surgical mortality audit may be associated with reduced mortality after emergency admission. ANZ J Surg 2017;87:830-6. CrossRef
4. Hospital Authority Statistical Report (2015-16). Available from: http://www.ha.org.hk/haho/ho/stat/HASR15_16.pdf. Accessed 10 Nov 2017.
5. Copeland G, Jones D, Walters M. POSSUM: A scoring system for surgical audit. Br J Surg 1991;78:355-60. CrossRef
6. Copeland G. The POSSUM system of surgical audit. Arch Surg 2002;137:15-9. CrossRef
7. Khuri S, Daley J, Henderson W, et al. The National VA Surgical Risk Study: risk adjustment for the comparative assessment of the quality of surgical care. J Am Coll Surg 1995;180:519-31.
8. Daley J, Khuri SF, Henderson W, et al. Risk adjustment of the postoperative morbidity rate for the comparative assessment of the quality of surgical care: results of the National Veterans Affairs surgical risk study. J Am Coll Surg 1997;185:328-40. CrossRef
9. Daley J. Validating risk-adjusted surgical outcomes: site visit assessment of process and structure. J Am Coll Surg 1997;185:341-51. CrossRef
10. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume One: July 2008-June 2009. Hospital Authority, Hong Kong SAR Government; 2010.
11. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Two: July 2009-June 2010. Hospital Authority, Hong Kong SAR Government; 2011.
12. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Three: July 2010-June 2011. Hospital Authority, Hong Kong SAR Government; 2012.
13. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Four: July 2011-June 2012. Hospital Authority, Hong Kong SAR Government; 2013.
14. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Five: July 2012-June 2013. Hospital Authority, Hong Kong SAR Government; 2014.
15. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Six: July 2013-June 2014. Hospital Authority, Hong Kong SAR Government; 2015.
16. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Seven: July 2014-June 2015. Hospital Authority, Hong Kong SAR Government; 2016.
17. Surgical Outcomes Monitoring & Improvement Program (SOMIP) report. Volume Eight: July 2015-June 2016. Hospital Authority, Hong Kong SAR Government; 2017.
18. Ash A, Schwartz M. Evaluating the performance of risk-adjustment methods: dichotomous variables. In: Iezzoni L, editor. Risk adjustment for measuring health care outcomes. Ann Arbor, MI: Health Administration Press; 1994: 313-46.
19. Hall BL, Hamilton BH, Richards K, Bilimoria KY, Cohen ME, Ko CY. Does surgical quality improve in the American College of Surgeons National Surgical Quality Improvement Program: An evaluation of all participating hospitals. Ann Surg 2009;250:363-76. CrossRef
20. Cohen ME, Liu Y, Ko CY, Hall BL. Improved surgical outcomes for ACS NSQIP hospitals over time: evaluation of hospital cohorts with up to 8 years of participation. Ann Surg 2016;263:267-73. CrossRef

Management of complications of ketamine abuse: 10 years’ experience in Hong Kong

Hong Kong Med J 2018;24:Epub 6 Apr 2018
DOI: 10.12809/hkmj177086
REVIEW ARTICLE
Management of complications of ketamine abuse: 10 years’ experience in Hong Kong
YL Hong, MSc1; CH Yee, FHKAM (Surgery)2, YH Tam, FHKAM (Surgery)1; Joseph HM Wong, FHKAM (Surgery)2; PT Lai, BN2; CF Ng, FHKAM (Surgery)2
1 Division of Paediatric Surgery and Paediatric Urology, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
2 SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
 
Corresponding author: Prof CF Ng (ngcf@surgery.cuhk.edu.hk)
 
 Full paper in PDF
 
Abstract
Ketamine is an N-methyl-d-aspartate receptor antagonist, a dissociative anaesthetic agent and a treatment option for major depression, treatment-resistant depression, and bipolar disorder. Its strong psychostimulant properties and easy absorption make it a favourable candidate for substance abuse. Ketamine entered Hong Kong as a club drug in 2000 and the first local report of ketamine-associated urinary cystitis was published in 2007. Ketamine-associated lower–urinary tract symptoms include frequency, urgency, nocturia, dysuria, urge incontinence, and occasionally painful haematuria. The exact prevalence of ketamine-associated urinary cystitis is difficult to assess because the abuse itself and many of the associated symptoms often go unnoticed until a very late stage. Additionally, upper–urinary tract pathology, such as hydronephrosis, and other complications involving neuropsychiatric, hepatobiliary, and gastrointestinal systems have also been reported. Gradual improvement can be expected after abstinence from ketamine use. Sustained abstinence is the key to recovery, as relapse usually leads to recurrence of symptoms. Both medical and surgical management can be used. The Youth Urological Treatment Centre at the Prince of Wales Hospital, Hong Kong, has developed a four-tier treatment protocol with initial non-invasive investigation and management for these patients. Multidisciplinary care is essential given the complex and diverse psychological factors and sociological background that underlie ketamine abuse and abstinence status.
 
 
Introduction
Ketamine is an N-methyl-d-aspartate (NMDA) receptor antagonist, a dissociative anaesthetic agent that was first synthesised in the United States in 1962. It has been widely used in both human and veterinary medicine since 1971. It has also been used as a treatment for major depression, treatment-resistant depression, and bipolar disorder.1 However, its strong psychostimulant properties and easy absorption make it a favourable candidate for substance abuse. Ketamine abuse has become increasingly common over the past two decades. It entered Hong Kong as a club drug in 2000 and was initially used as a ‘top-up’ drug to ecstasy (3,4-methylenedioxymethamphetamine) by those aiming to elevate and redirect the ‘high’.2 It was viewed as a poor man’s version of cocaine, as it is available in powder form and can be consumed by snorting. By 2002, ketamine had become the drug of choice instead of a ‘top-up’ drug. Users self-administered ketamine to have a ‘time-out’ or ‘sit-down-to-float’ experience.2 3 Within a short period, the number of reported ketamine abusers in Hong Kong increased from 1605 in 2000 to its peak of 5280 in 2009. Ketamine remained the most popular psychotropic substance abused from 2005 to 2014 (Fig 1).4
 

Figure 1. Number of reported drug abusers in Hong Kong, 1996-2016, by common type of psychotropic substances (data source: Narcotics Division, HKSAR Government4)
 
The abuse of ketamine and its popularity nonetheless created a new medical entity. The first report of ketamine-associated urinary cystitis in Hong Kong was published in 2007.5 In the same year, Shahani et al reported a similar condition overseas.6 In the past decade, owing to the joint efforts of urologists, general surgeons, physicians, psychiatrists, pathologists, and basic scientists, we have gained a better understanding of other ketamine-associated conditions. This understanding spans from pathology to clinical management, from urological complications to upper-gastrointestinal (GI) complications, and from a mouse model to humans. We have also explored holistic ways to manage this condition in the long term, such as helping young adults to have a fresh start while living with potentially irreversible complications. This article reviews the evolution of local clinical awareness and management of these complications, with a particular focus on the work and contributions of local researchers.
 
Urological complications
Chu et al5 reported the first local case series of ketamine-associated bladder dysfunction in 2007. Ketamine-associated lower–urinary tract symptoms (LUTS) include frequency, urgency, nocturia, dysuria, urge incontinence, and occasionally painful haematuria. The exact prevalence of ketamine-associated urinary cystitis is difficult to assess because ketamine abuse and many of the associated symptoms often go unnoticed until a very late stage. A survey among 12 000 local secondary school students revealed that 18.5% of non-psychotropic substance users had LUTS, whereas 47.8% and 60.7% of psychotropic substance users and ketamine users had LUTS, respectively.7 Unpublished data from the same study showed that compared with non-psychotropic substance users, sole ketamine users were five times as likely to have LUTS, whereas concomitant users of ketamine and methamphetamine were eight times as likely to have LUTS.
 
In 2015, Yee et al8 reported the largest available cohort of both active and past ketamine users who had ketamine-associated uropathy. Among 463 patients, ketamine users had a significantly higher pelvic pain and urgency/frequency (PUF) score than ex-ketamine users. The PUF score is initially used to assess interstitial cystitis, and a higher PUF score correlates with worse symptoms. Among active ketamine users, a higher PUF score was found to correlate with a poorer quality of life and a smaller functional bladder capacity.8 Achieving abstinence from ketamine use and consuming smaller amounts of ketamine were factors that predicted improvement in PUF score.
 
As well as bladder involvement, upper–urinary tract pathology presenting as hydronephrosis and flank pain has also been reported (Fig 2). A study by Yee et al9 of 572 patients with ketamine-related LUTS found that up to 16.8% (n=96) of patients had hydronephrosis according to ultrasonography. Hydronephrosis was frequently accompanied by ureteral lesions, ureteral wall thickening, or vesicoureteral (vesicoureteric) reflux. Similarly, Chu et al10 reported that 51% (30/59) of their patients had hydronephrosis according to ultrasonography.
 

Figure 2. Intravenous urogram of a 28-year-old female in Prince of Wales Hospital showing bilateral hydronephrosis and small bladder
 
Pathophysiology
Although the exact mechanism of ketamine-associated cystitis remains to be explored, there is evidence that ketamine metabolites in the urine induce chemical irritation of the urothelium, thereby causing an inflammatory response.11 Severe irritation may lead to denudation of the urothelium and consequent transmural inflammation, loss of muscle thickness, fibrosis of the detrusor muscle, and ultimately poor urinary bladder compliance. Vesicoureteral reflux or urinary stasis in the ureter may occur, causing chronic ureteral inflammation and ureteral stricture. There is also evidence that both systemic and local inflammatory markers are elevated in ketamine users.10 12 In addition, the NMDA antagonist properties of ketamine may exert their effect via a central pathway.13
 
Neuropsychiatric complications
As ketamine is a psychostimulant, it is not surprising that it is associated with long-term neurocognitive problems. Chan et al14 found that when ketamine users were compared with healthy controls, they had impaired verbal fluency, cognitive processing speed, and verbal learning. Heavy ketamine use correlated with deficits in verbal memory and visual recognition memory. Liang et al15 also identified predominant verbal and visual memory impairment in ketamine poly-drug users. Unfortunately, these deficits persisted in ex-users. A much higher incidence of psychiatric co-morbidities, including psychosis, depression, and anxiety, was observed among ketamine users.16
 
Pathophysiology
Structural brain damage associated with ketamine abuse was supported by magnetic resonance imaging (MRI) by Wang et al.17 They were the first group to report patches of degeneration in the superficial white matter as early as 1 year after ketamine addiction onset. Cortical atrophy was also found in the frontal, parietal, or occipital cortices of addicts.17 Another MRI study also provided evidence of brain damage in chronic ketamine users. Reduced grey- and white-matter volumes were noted in the bilateral orbitofrontal cortex, right medial prefrontal cortex, and bilateral hippocampi. There was also significantly decreased connectivity inside the brain in chronic ketamine abusers.18
 
A series of studies on the neurotoxicity of ketamine suggested that ketamine could cause apoptosis of neuronal cells in both in-vitro and in-vivo models. Ketamine also potentially causes phosphorylation of tau protein, a marker of Alzheimer’s degeneration in the brain.19 20 21 22
 
Hepatobiliary complications
In 2009, Wong et al23 reported ketamine-associated hepatobiliary complications for the first time. Three ketamine abusers presented with recurrent epigastric pain and dilated bile ducts mimicking choledochal cysts. Subsequently, more similar cases were identified. Fusiform dilatation of the common bile duct was also observed.24 25 26 Liver biopsy confirmed development of active liver and/or bile duct injury. A study of 297 chronic ketamine abusers with urinary tract dysfunction showed that the prevalence of liver injury was 9.8%.27 These studies and reports show the possibility and severity of damage by ketamine to the hepatobiliary and pancreatic system.
 
Pathophysiology
The exact mechanism of ketamine-associated bile injury is still unknown. The associated rise in C-reactive protein suggests a possible inflammatory process in the liver parenchyma, including or excluding the bile duct.27 Others have postulated that either central or direct action of ketamine on the biliary smooth muscle in turn leads to the cholestasis and biliary dilatation observed in ketamine abusers.28
 
Gastrointestinal complications
In addition to urological complaints, GI problems are also frequently the symptoms for which ketamine abusers seek medical help. A review of 233 ketamine-related visits to accident and emergency departments found that 49 (21.0%) patients had abdominal pain, 23 (9.9%) had nausea or vomiting, and 41 (17.6%) had abdominal tenderness.29 Gastrointestinal complaints often co-exist with and precede the presentation of urological symptoms. Liu et al30 found that about a quarter (168; 27.5%) of 611 ketamine users who sought treatment for ketamine uropathy reported the presence of upper-GI symptoms, whereas only 42 (5.2%) of 804 non-ketamine users attending a general urology clinic reported similar symptoms (P<0.001). The majority of the symptoms reported were epigastric pain and recurrent vomiting. Nearly three-quarters of patients required hospitalisation for acute or chronic upper-GI symptoms. With the exception of acid reflux and perforated peptic ulcer, the prevalence of all the above-mentioned symptoms and hospitalisation rates were statistically significantly higher in ketamine users than in non-ketamine users. All 168 patients using ketamine had undergone oesophagogastroduodenoscopy during which biopsies were taken. Pathological findings ranged from gastritis to gastroduodenal erosions, peptic ulceration, and intestinal metaplasia.30
 
Liu et al30 also found that more than 80% of patients developed upper-GI symptoms before urological symptoms. Patients developed upper-GI symptoms after a mean (standard deviation) of 5.1 (3.1) years of ketamine use and developed uropathy symptoms after another 4.4 (3.0) years of ketamine use.30 Epigastric symptoms are not common in young people, but common in ketamine abusers. This difference may provide an opportunity to identify hidden ketamine abuse when assessing young patients with epigastric symptoms. The identification of ketamine use is important, as cessation of use can greatly improve GI symptoms.31 Further referral for help and counselling may improve psychological and physical health and promote long-term ketamine abstinence.
 
Pathophysiology
The exact pathophysiological mechanism by which ketamine produces upper-GI toxicity remains unknown but there are several postulations. First, ketamine, as an NMDA antagonist, might act on local smooth muscle or the central nervous system, thereby affecting gastric motility and leading to cramping pain. Second, microvascular damage by ketamine and its metabolites, which was believed to be a possible cause of ketamine uropathy, might also cause similar microvascular damage in the stomach and duodenum, leading to ischaemic pain and inflammation. Likewise, circulating ketamine might also trigger some unknown autoimmune responses, and thus induce interstitial inflammation in the urinary and GI tracts. Finally, as many ketamine abusers like to swallow the nasal drips occurring from ketamine inhalation, the swallowed ketamine might also induce direct cytotoxic injury to the vulnerable GI tract.30
 
Management
As in the management of other substance abuse, abstinence is the key to success in overall management of ketamine use. Whereas other treatment modalities may relieve symptoms and hasten the recovery process, many ketamine abusers have complicated underlying psychosocial problems and psychiatric co-morbidities. Long-term and consistent support and encouragement from doctors, nurses, social workers, family, and friends are vital for success.
 
Abstinence from ketamine use
Recurrence of symptoms after resuming ketamine use highlights the importance of ketamine abstinence. Studies have shown that abstinence leads to symptomatic improvement. Compared with active ketamine abusers, those who had abstained for 1 year had significantly lower PUF scores and a larger voided volume. There was a trend towards higher voided volumes and lower PUF scores as duration of ketamine cessation increased, although neither variable was statistically significant.32 Another follow-up study of 101 participants who had abstained from ketamine and 218 active ketamine users showed that the abstinence group had a statistically significantly lower PUF score, and a higher functional bladder capacity.8 Moreover, abstinence was the only protective factor associated with fewer symptoms, larger voided volume, and bladder capacity.33
 
Nonetheless, abstinence does not lead to immediate and full recovery of symptoms. Gradual improvement can be expected but sustained abstinence is the key to recovery. Patience and continuous support are of paramount importance. A study showed that on admission to a drug rehabilitation centre, 90% of 40 female ex-ketamine users still had active urinary symptoms, with increased 24-hour urinary frequency, lower maximum voided volume, smaller median functional bladder capacity, and higher mean Urogenital Distress Inventory Short Form (UDI-6) and Incontinence Impact Questionnaire Short Form (IIQ-7) scores, when compared with age-matched controls who attended a general gynaecology clinic. After having stopped using ketamine for 3 months or more, mean 24-hour urinary frequency and mean UDI-6 and IIQ-7 scores decreased, and maximum voided volume increased. These scores further improved after another 3 months, although this group continued to perform more poorly in all aspects compared with controls.34
 
Medical and surgical management
As ketamine-associated uropathy is an evolving ‘disease entity’, the exact pathophysiology remains to be elucidated. Some of the clinical features share similarities with interstitial cystitis. Protocols are being developed to cater to the needs of patients in Hong Kong. A one-stop service model has been adopted by the Youth Urological Treatment Centre at the Prince of Wales Hospital since 2011 (Fig 3). The standard treatment protocol involves four tiers of treatment, starting with an initial non-invasive investigative approach, including questionnaire assessment of symptoms and calculation of (1) functional bladder capacity by measuring voided volume using uroflowmetry and (2) residual urine using ultrasound bladder scanning. This non-invasive investigative approach helps gain patients’ trust and improve adherence to later follow-up.
 

Figure 3. One-stop screening clinic model of the Youth Urological Treatment Centre, Prince of Wales Hospital, Hong Kong
 
First-tier treatment includes oral non-steroidal anti-inflammatory drugs (NSAIDs) (eg, diclofenac) and anticholinergics (eg, solifenacin) or COX-II inhibitors (eg, etoricoxib) if patients cannot tolerate NSAIDS. Simple analgesics such as paracetamol and phenazopyridine are used for pain control. The Youth Urological Treatment Centre has reported the largest series of patients with ketamine-associated uropathy and their corresponding outcomes. Of 290 patients with ketamine cystitis who received first-line treatment, 202 (69.7%) reported symptom improvement and a reduction in PUF scores. Functional bladder capacity was also shown to have improved.8
 
The opioid group of analgesics and pregabalin are used in the next tier of pain-control treatment when first-tier treatment is insufficient for symptom relief. Sixty-two patients received second-line treatment and 42 (67.7%) responded to treatment.8
 
Third-tier treatment consists of a course of intravesical instillation of sodium hyaluronate (6-weekly instillations followed by 2-monthly instillations) attempting to repair the glycosaminoglycan layer. The drug is given to patients whose symptoms remain uncontrolled after second-tier treatment. Seventeen patients in the cohort received the third-tier treatment and eight completed the course. Significant improvement in voided volume was noted and five were able to reduce their oral medication usage after treatment. No significant adverse effects were reported.8
 
Unfortunately, for a proportion of patients with extremely refractory symptoms, surgery becomes the fourth-tier treatment of choice. In the Youth Urological Treatment Centre series, one patient in the cohort required hydrodistension and another underwent robotic-assisted laparoscopic augmentation cystoplasty. The patient with hydrodistension experienced a recurrence of symptoms post-treatment.8 Ng et al35 reported on four patients who underwent augmentation cystoplasty. Although they showed initial improvement, all patients relapsed and resumed ketamine use postoperatively. Three of the patients showed a further deterioration in renal function, secondary to new-onset ureteral strictures and/or sepsis. Therefore, patient selection, education, close follow-up, and support are vital to the success of augmentation cystoplasty.35
 
Multidisciplinary care
Given the complex and diverse psychological factors and sociological background contributing to an individual’s decision to abuse ketamine or achieve abstinence, joint multidisciplinary efforts are required to help affected young adults. Doctors, social workers, teachers, psychiatrists, psychologists, nurses, and patients’ families all need to support them on their long road to recovery, to help them rehabilitate physically and achieve sustained abstinence from ketamine.33 36 37
 
Conclusion
Since the initial discovery of ketamine-associated uropathy, the impact of this disease entity has become more prominent in Asian countries. Thanks to the joint efforts of urologists, gynaecologists, surgeons, psychiatrists, pathologists, and social workers, as well as the support of local government, the extent of medical complications has been revealed to also involve the brain, liver, and GI system. Many ketamine abusers are ‘hidden’ and can use ketamine stealthily at home for years without their family noticing. Clinicians must take the opportunity to identify hidden abusers when they consult for non-specific symptoms such as epigastric pain and LUTS. Doing so will not only enable early diagnosis of ketamine-associated uropathy, but it will also help provide appropriate medical treatment in a timely manner. In addition to medical therapy, referral for appropriate psychosocial support is essential to sustain abstinence and manage underlying psychosocial problems.
 
Acknowledgement
The Youth Urological Treatment Centre was developed by joint efforts of The Chinese University of Hong Kong and the Hong Kong Hospital Authority, with generous support from the Beat Drugs Fund of the Narcotics Division, Security Bureau, Government of the Hong Kong Special Administrative Region.
 
Declaration
The authors have no conflicts of interest to disclose.
 
References
1. Hasselmann HW. Ketamine as antidepressant? Current state and future perspectives. Curr Neuropharmacol 2014;12:57-70. CrossRef
2. Joe Laidler KA. The rise of club drugs in a heroin society: the case of Hong Kong. Subst Use Misuse 2005;40:1257-78. CrossRef
3. Joe-Laidler K, Hunt G. Sit down to float: the cultural meaning of ketamine use in Hong Kong. Addict Res Theory 2008;16:259-71. CrossRef
4. Narcotics Division, Security Bureau, HKSAR Government. Statistical tables customization. Available from: http://cs-crda.nd.gov.hk/en/enquiry.php. Accessed 22 Sep 2017.
5. Chu PS, Kwok SC, Lam KM, et al. ‘Street ketamine’–associated bladder dysfunction: a report of ten cases. Hong Kong Med J 2007;13:311-3.
6. Shahani R, Streutker C, Dickson B, et al. Ketamine-associated ulcerative cystitis: a new clinical entity. Urology 2007;69:810-2. CrossRef
7. Tam YH, Ng CF, Wong YS, et al. Population-based survey of the prevalence of lower urinary tract symptoms in adolescents with and without psychotropic substance abuse. Hong Kong Med J 2016;22:454-63. CrossRef
8. Yee CH, Lai PT, Lee WM, et al. Clinical outcome of a prospective case series of patients with ketamine cystitis who underwent standardized treatment protocol. Urology 2015;86:236-43. CrossRef
9. Yee CH, Teoh YC, Lai PT, et al. The risk of upper urinary tract involvement in patients with ketamine-associated uropathy. Int Neurourol J 2017;21:128-32. CrossRef
10. Chu PS, Ma WK, Wong SC, et al. The destruction of the lower urinary tract by ketamine abuse: a new syndrome? BJU Int 2008;102:1616-22. CrossRef
11. Chu PS, Ng CF, Ma WK. Ketamine uropathy: Hong Kong experience. In: Yew DT, editor. Ketamine: Use and Abuse. CRC Press; 2015: 207-26.
12. Division of Urology, Department of Surgery, Princess Margaret Hospital and Tuen Mun Hospital. Research on Urological Sequelae of Ketamine Abuse. Available from: http://www.nd.gov.hk/pdf/20110307_beat_drug_fund_report.pdf. Accessed 22 Sep 2017.
13. Jankovic SM, Jankovic SV, Stojadinovic D, et al. Effect of exogenous glutamate and N-methyl-D-aspartic acid on spontaneous activity of isolated human ureter. Int J Urol 2007;14:833-7. CrossRef
14. Chan KW, Lee TM, Siu AM, et al. Effects of chronic ketamine use on frontal and medial temporal cognition. Addict Behav 2013;38:2128-32. CrossRef
15. Liang HJ, Lau CG, Tang A, et al. Cognitive impairments in poly-drug ketamine users. Addict Behav 2013;38:2661-6. CrossRef
16. Tang WK, Morgan CJ, Lau GC, et al. Psychiatric morbidity in ketamine users attending counselling and youth outreach services. Subst Abus 2015;36:67-74. CrossRef
17. Wang C, Zheng D, Xu J, et al. Brain damages in ketamine addicts as revealed by magnetic resonance imaging. Front Neuroanat 2013;7:23. CrossRef
18. Narcotics Division, Security Bureau, HKSAR Government. Evidence of Brain Damage in Chronic Ketamine Users—a Brain Imaging Study. Available from: http://www.nd.gov.hk/pdf/Evidence-of-Brain-Damage-in-Chronic-Ketamine-Users_Final-report.pdf. Accessed 22 Sep 2017.
19. Yeung LY, Wai MS, Fan M, et al. Hyperphosphorylated tau in the brains of mice and monkeys with long-term administration of ketamine. Toxicol Lett 2010;193:189-93. CrossRef
20. Narcotics Division, Security Bureau, HKSAR Government. Long-term Ketamine Abuse and Apoptosis in Cynomolgus Monkeys and Mice. Available from: http://www.nd.gov.hk/pdf/long_term_ketamine_abuse_apoptosis_in_cynomologus_monkeys_and_mice/final_report_with_all_attachments.pdf. Accessed 22 Sep 2017.
21. Tan S, Rudd JA, Yew DT. Gene expression changes in GABA(A) receptors and cognition following chronic ketamine administration in mice. PLoS ONE 2011;6:e21328. CrossRef
22. Mak YT, Lam WP, Lü L, et al. The toxic effect of ketamine on SH-SY5Y neuroblastoma cell line and human neuron. Microsc Res Tech 2010;73:195-201.
23. Wong SW, Lee KF, Wong J, et al. Dilated common bile ducts mimicking choledochal cysts in ketamine abusers. Hong Kong Med J 2009;15:53-6.
24. Ng SH, Lee HK, Chan YC, et al. Dilated common bile ducts in ketamine abusers. Hong Kong Med J 2009;15:157.
25. Cheung TT, Poon RT, Chan AC, et al. Education and imaging. Hepatobiliary and pancreatic: cholangiopathy in ketamine user—an emerging new condition. J Gastroenterol Hepatol 2014;29:1663. CrossRef
26. Lui KL, Lee WK, Li MK. Ketamine-induced cholangiopathy. Hong Kong Med J 2014;20:78.e1-2. CrossRef
27. Wong GL, Tam YH, Ng CF, et al. Liver injury is common among chronic abusers of ketamine. Clin Gastroenterol Hepatol 2014;12:1759-62.e1. CrossRef
28. Lo RS, Krishnamoorthy R, Freeman JG, et al. Cholestasis and biliary dilatation associated with chronic ketamine abuse: a case series. Singapore Med J 2011;52:e52-5.
29. Ng SH, Tse ML, Ng HW, et al. Emergency department presentation of ketamine abusers in Hong Kong: a review of 233 cases. Hong Kong Med J 2010;16:6-11.
30. Liu SY, Ng SK, Tam YH, et al. Clinical pattern and prevalence of upper gastrointestinal toxicity in patients abusing ketamine. J Dig Dis 2017;18:504-10. CrossRef
31. Poon TL, Wong KF, Chan MY, et al. Upper gastrointestinal problems in inhalational ketamine abusers. J Dig Dis 2010;11:106-10. CrossRef
32. Mak SK, Chan MT, Bower WF, et al. Lower urinary tract changes in young adults using ketamine. J Urol 2011;186:610-4. CrossRef
33. Tam YH, Ng CF, Pang KK, et al. One-stop clinic for ketamine-associated uropathy: report on service delivery model, patients’ characteristics and non-invasive investigations at baseline by a cross-sectional study in a prospective cohort of 318 teenagers and young adults. BJU Int 2014;114:754-60. CrossRef
34. Cheung RY, Chan SS, Lee JH, et al. Urinary symptoms and impaired quality of life in female ketamine users: persistence after cessation of use. Hong Kong Med J 2011;17:267-73.
35. Ng CF, Chiu PK, Li ML, et al. Clinical outcomes of augmentation cystoplasty in patients suffering from ketamine-related bladder contractures. Int Urol Nephrol 2013;45:1245-51. CrossRef
36. Ng CF. Editorial comment to possible pathophysiology of ketamine-related cystitis and associated treatment strategies. Int J Urol 2015;22:826. CrossRef
37. Chu PS, Dasgupta P. Lessons learned from Asian Urology. BJU Int 2014;114:633. CrossRef

Endobronchial valve for treatment of persistent air leak complicating spontaneous pneumothorax

Hong Kong Med J 2018;24:Epub 4 Apr 2018
DOI: 10.12809/hkmj176823
ORIGINAL ARTICLE
Endobronchial valve for treatment of persistent air leak complicating spontaneous pneumothorax
WC Yu, MB, BS, FHKCP1; Ellen LM Yu, MSc2; HC Kwok, MB, BS, FHKCP1; HL She, MB, BS, FRCR3; KK Kwong, MB, BS, FHKCP1; YH Chan, MB, BS, FHKCP1; YL Tsang, BSc4; YC Yeung, MB, BS, FHKCP1
1 Department of Medicine and Geriatrics, Princess Margaret Hospital, Laichikok, Hong Kong
2 Clinical Research Centre, Princess Margaret Hospital, Laichikok, Hong Kong
3 Department of Radiology, Princess Margaret Hospital, Laichikok, Hong Kong
4 Central Endoscopy Unit, Princess Margaret Hospital, Laichikok, Hong Kong
 
Corresponding author: Dr WC Yu (h7537800@connect.hku.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Endobronchial one-way valves have been proposed as treatment for persistent air leak complicating spontaneous pneumothorax in which surgical intervention is not feasible. However, published data on efficacy, safety, and factors associated with success are scanty.
 
Methods: This is a retrospective study of 37 patients at a general hospital from 2008 to 2016. The impact of endobronchial valve implantation on the time to air-leak cessation after bronchoscopy was evaluated.
 
Results: The median patient age was 71 years. The majority of patients were males (92%), were ever-smokers (83%), had at least one co-morbidity (97%), and had secondary spontaneous pneumothorax (89%). Nineteen patients (51%) had a mean of 2.6 endobronchial valves implanted (range, 1-6). The air leak ceased within 72 hours for only eight patients (22% of the complete cohort), with immediate air-leak cessation after completion of endobronchial valve implantation. All six successful cases that had computed tomographic data of the thorax were shown to have bilateral intact interlobar fissures. The median (interquartile range) Charlson co-morbidity index was 1 (0.25-1) and 2 (1-3) for the success group and failure group, respectively (P=0.029). All patients in the no-endobronchial valve group survived, whereas three patients in the endobronchial valve group died within 30 days of endobronchial valve implantation.
 
Conclusion: Only a small proportion of cases of endobronchial valve implantation for air leak complicating pneumothorax had unequivocal success. Intact bilateral interlobar fissures appear to be a necessary, though not sufficient, condition for success. Patients with fewer medical co-morbidities and immediate air-leak cessation after endobronchial valve implantation have a higher likelihood of success.
 
 
New knowledge added by this study
  • Only a small proportion of cases (22%) of persistent air leak complicating spontaneous pneumothorax can be successfully treated by endobronchial valve (EBV).
  • Cases in which the air leak does not cease immediately after completion of EBV implantation are likely to fail.
  • Patients with any non-intact interlobar fissures are likely to experience treatment failure.
Implications for clinical practice or policy
  • EBV treatment may be attempted in patients with persistent air leak complicating spontaneous pneumothorax who are not candidates for surgery, have intact interlobar fissures, and do not have severe medical comorbidities.
  • Plain computed tomography of the thorax should be performed to routinely assess patients who are candidates for EBV treatment.
  • Cases of completed EBV implantation that still have an air leak may have the EBVs removed.
 
 
Introduction
Persistent air leak is a common complication of spontaneous pneumothorax being treated by chest tube drainage. In one report, the median time for spontaneous cessation of air leak was 7 days for primary spontaneous pneumothorax and 11 days for secondary spontaneous pneumothorax.1 In another report, 75% and 100% of cases of primary spontaneous pneumothorax resolved by 7 days and 14 days, respectively, whereas the corresponding proportions for secondary spontaneous pneumothorax were 61% and 79%, respectively.2 It is generally recommended that surgical intervention be considered when an air leak continues for 7 to 14 days after chest tube insertion. Unfortunately, some patients are poor candidates for surgery, and some patients may refuse surgery. Prolonged chest tube in situ, however, is undesirable because of an increased risk of complications, prolonged immobilisation and its consequences, and increased duration of hospital stay. Alternative means to shorten the duration of air leakage are thus needed.
 
Endobronchial one-way valves (EBVs) have been used to treat persistent air leak complicating pneumothorax. The rationale is that air leakage will stop if air is prevented from entering the airway leading to the leak site. Preliminary case reports showed encouraging results.3 4 5 6 7 8 9 10 11 12 13 14 Larger case series showed similarly favourable results.15 16 17 18 19 20 21 22 23 However, most of these reports include a mix of post-surgical pneumothoraces, spontaneous pneumothoraces, and other aetiologies. Moreover, there is scanty information on overall efficacy, short-term safety, and factors related to favourable clinical outcomes.
 
The Princess Margaret Hospital, an acute-care general hospital in Hong Kong, has been using EBV for persistent air leak complicating pneumothorax on compassionate grounds since 2008, and its preliminary experience on three apparently successful cases was reported in 2009.11 However, it was subsequently found that in many cases, the leak site could not be identified, so EBV could not be deployed. In some other cases, despite leak site identification and EBV deployment, the air leak may not resolve completely, or it may recur after a few hours. We therefore hypothesise that EBV treatment has a low success rate in real-life situations. To test this hypothesis, we retrospectively compared the clinical course of patients with and without EBV use for persistent air leak complicating spontaneous pneumothorax during an 8-year period at a single centre.
 
Methods
Patients
This retrospective chart review was based on patients who were managed at the Princess Margaret Hospital from May 2008 to April 2016. Eligible patients were those with spontaneous pneumothorax who were undergoing chest tube drainage and who had had an air leak lasting more than 1 week. All showed bubbling in the drainage bottle during both expiration and inspiration, with water suction at -20 cm. Patients either refused surgery or were considered by the thoracic surgeon to be unsuitable for surgical intervention owing to advanced age, poor lung function, major medical co-morbidities, or a combination of these.
 
Intervention
Bronchoscopy with the intention of EBV implantation to stop the air leak was suggested to eligible patients. Some were reluctant at first but consented after further days of air leakage. Procedures for identifying the leak site and deploying EBV were as described previously.11 Briefly, an endobronchial inflatable balloon was used to occlude lobar or segmental bronchi sequentially. If bubbling from the chest drainage bottle stopped, or was greatly reduced, then the lobe or segment was selected for endobronchial valve implantation. The Zephyr EBV (Emphasys Inc, now Pulmonx Inc, Redwood City [CA], United States) was used throughout. Patients were observed closely for continued air leak after bronchoscopy. Chest tubes were removed as soon as possible after air-leak cessation, typically within 1 or 2 days, as is usual clinical practice.
 
Data collection and outcome measures
Data on demographic and disease characteristics, details of bronchoscopy, and survival after bronchoscopy were recorded. Spirometry data were analysed if available within 1 year of bronchoscopy. Spirometry was performed according to the American Thoracic Society / European Respiratory Society criteria24 and using reference values for Hong Kong adults.25 No spirometry was performed during the period of chest tube drainage or with EBV in situ. Computed tomography images, if available, were viewed in axial, sagittal, and coronal planes by a single radiologist who was blinded to other study data. The integrity of the fissures was defined as more than 90% completeness on at least one axis.26 Outcome measures included whether EBV was implanted, time to air-leak cessation, and 30-day all-cause mortality after bronchoscopy.
 
Definition of therapeutic success
Therapeutic success was defined as cessation of the air leak within 72 hours of EBV implantation. Cases with EBV implanted but with the air leak lasting more than 72 hours afterwards, cessation of air leak within 72 hours of bronchoscopy owing to other interventions (such as surgery), cases without EBV implantation, and cases of chest tube removal while the air leak persisted (such as following unintended displacement) were considered cases of failure.
 
Statistical analysis
The Mann-Whitney U test and Fisher’s exact test were used to examine differences between groups. The Kaplan-Meier log-rank test and Gehan-Breslow-Wilcoxon test were used to analyse time to air-leak cessation after first bronchoscopy among patients with and without EBV implantation. The association between EBV implantation and air-leak cessation was assessed in a multivariable Cox proportional hazards regression model that was adjusted for ‘days on chest tube before first bronchoscopy’ and factors with P<0.2 in univariable regression analyses. The accuracy, sensitivity, specificity, and positive and negative predictive values of using the presence of an intact interlobar fissure as an indicator of a successful outcome were calculated. Statistical analyses were performed using SPSS 22.0 for Windows (IBM Corp., Armonk [NY], United States) and OpenEpi: Open Source Epidemiologic Statistics for Public Health, version 3.01 (http://www.openepi.com). Statistical significance was set at P<0.05.
 
This study was approved by the Research Ethics Committee of the Kowloon West Cluster of the Hong Kong Hospital Authority, with the requirement for patient consent waived. This report conforms to the STROBE 2008 guidelines.27
 
Results
Baseline characteristics
During the study period, 38 patients underwent bronchoscopy for persistent air leak complicating spontaneous pneumothorax at our institution. One patient was excluded because the air leak was subsequently found to result from the chest tube having been inserted into an airway. Of the remaining 37 patients, 35 were assessed by the surgeon and considered unfit for surgery, and two patients with primary spontaneous pneumothorax refused surgery. The median patient age was 71 years and most (34; 92%) were males. The majority (83%) were ever-smokers. All but one (97%) had at least one co-morbidity and the median Charlson co-morbidity index was 1. Fifteen patients (41%) had a left pneumothorax. Thirty-three (89%) had secondary spontaneous pneumothorax, of which 23 had chronic obstructive pulmonary disease with or without other lung diseases. Of the 22 secondary spontaneous pneumothorax cases with spirometric data, the median forced expiratory volume in 1 second (FEV1) was 0.94 L; median percentage of predicted FEV1 was 43.5%; and median FEV1 to forced vital capacity ratio was 0.45. Eleven patients (30%) had at least one attempt at talc pleurodesis before bronchoscopy. The median number of days off chest tube use before bronchoscopy was 25 (Table 1).
 

Table 1. Baseline characteristics of patients with and without endobronchial valve implantation at first bronchosopy
 
Endobronchial one-way valve implantation and time to air-leak cessation
Nineteen patients (51%) had a mean of 2.6 EBVs implanted (range, 1-6). The sites of EBV implantation were as follows: 11 in the right upper lobe, one in the right middle lobe, none in the right lower lobe, five in the left upper lobe, and two in the left lower lobe. Of the 18 patients without an EBV implanted, the target site for EBV implantation could not be identified in 17. In the remaining patient, despite identification of the air leak in the left upper lobe, EBV implantation was followed by severe oxygen desaturation and had to be abandoned.
 
There were eight successful cases among patients with EBVs implanted. Among these, seven patients had immediate and lasting cessation of the air leak, and the chest tube was removed within 2 days. One patient had immediate air-leak cessation but there was recurrence after 2 hours. The air leak subsided completely within 72 hours, and the chest tube was removed on day 4. Among the 11 cases of failure after EBV implantation, the air leak stopped immediately in three cases but recurred soon after and persisted beyond 72 hours. In the other eight cases, the air leak was reduced temporarily but was present beyond 72 hours. In the group without an implanted EBV, the earliest time for air-leak cessation was day 5, and the chest tube was removed on day 8. In a Kaplan-Meier comparison of the EBV and no-EBV groups in the number of days from first bronchoscopy to air-leak cessation, the EBV group did better in the first 30 days, but the no-EBV group caught up by day 45. There was a statistically significant difference between the two groups according to the Gehan-Breslow-Wilcoxon test (P=0.027), but not the log-rank test (P=0.138) [Fig Part a]. When the same comparison was done with the eight successful cases removed from the EBV group, the two curves overlapped throughout and there was no statistically significant difference between the two groups (P=0.881 by Gehan-Breslow-Wilcoxon test and P=0.976 by log-rank test) [Fig Part b].
 

Figure. Kaplan-Meier curves of air-leak cessation after first bronchoscopy in patients with and without an implanted endobronchial valve (EBV): (a) complete cohort; (b) all eight successful cases removed from the EBV implanted group
 
In a comparison of demographic and clinical characteristics between the EBV group and the no-EBV group, the former had a significantly poorer FEV1, but not percentage of predicted FEV1. This group also had the chest tube in place for a significantly longer duration before bronchoscopy (Table 1).
 
Factors associated with a successful outcome
We further examined factors related to air-leak cessation after first bronchoscopy by Cox proportional hazards regression analysis. The implantation of an EBV was significantly associated with air-leak cessation (adjusted hazard ratio=2.39, 95% CI=1.13-5.05; P=0.023), whereas the Charlson co-morbidity index was significantly associated with delayed air-leak cessation (adjusted hazard ratio=0.78, 95% CI=0.63-0.97; P=0.026). The number of days of chest tube use before first bronchoscopy was not associated with air-leak cessation (Table 2).
 

Table 2. Cox proportional hazards regression results for air-leak cessation after first bronchoscopy
 
When comparing the eight successful cases with the 11 failed cases among those with implanted EBVs, we found no difference in any of the demographic or disease variables between the two groups, apart from the Charlson co-morbidity index, which was significantly lower for the success group (Table 3). Additionally, all eight patients (100%) in the success group had air-leak cessation immediately after EBV implantation, versus only three of 11 (27%) in the failure group (P=0.003). When we compared the eight successful cases with all 29 failed cases, again the median (interquartile range) Charlson co-morbidity index was significantly lower for the success group: 1 (0.25-1) vs 2 (1-3) [P=0.029]. This group also had significantly poorer FEV1, but not in percentage of predicted FEV1, and showed a trend towards more severe airflow obstruction, although this did not reach statistical significance (Table 3).
 

Table 3. Comparison between cases of success and failure
 
Twenty-three patients had thoracic computed tomography performed to examine the integrity of interlobar fissures. For the 13 patients with all fissures intact, six had an EBV implanted and were classed as successful cases, four had EBV implanted but were classed as failed cases, and three did not have an EBV implanted. There were five patients each in the group with any non-intact ipsilateral fissure and in the group with any non-intact contralateral fissure. Both groups had two patients with an implanted EBV and three without, and all experienced treatment failure.
 
When using the presence of all intact fissure(s) to screen for successful EBV treatment among the 23 patients who had undergone thoracic computed tomography, the accuracy was 69.6%, the sensitivity and negative predictive value were both 100%, and the specificity and positive predictive value were 58.8% and 46.2%, respectively (Table 4). When the criterion for success was immediate air-leak cessation after EBV implantation, the accuracy among the 19 patients with an implanted EBV was 84.2%, the sensitivity and negative predictive value remained at 100%, and both the specificity and positive predictive value were 72.7% (Table 4).
 

Table 4. Prediction of successful outcome by fissure integrity and air-leak cessation after endobronchial valve implantation
 
Adverse events and mortality
There was no incident of valve displacement, bleeding, or post-obstructive pneumonia. Three patients died within 30 days of EBV implantation and all had advanced age and multiple co-morbidities (Table 5). In two patients, the causes of death were clearly related to ongoing pre-existing disease. One patient had a sudden cardiac arrest on day 29 after being successfully treated with EBV, although the relationship of death to EBV was uncertain. The earliest death in the no-EBV group occurred on day 43 after bronchoscopy and the cause of death was lung cancer.
 

Table 5. Characteristics of patients with endobronchial valve implantation who died within 30 days of bronchoscopy
 
Subsequent treatments
Three patients underwent a second bronchoscopy after the first one failed, with one success and two failures. Sixteen patients received talc pleurodesis (median, 2 times; range, 1-5 times). One young patient with primary spontaneous pneumothorax underwent surgery 2 days after failed identification of the leak site by bronchoscopy. Seventeen patients did not receive any further treatment, mostly owing to early air-leak cessation.
 
Implant removal
Thirteen patients had their implanted EBVs removed after a median of 43 days (range, 21-155 days). For the remaining six patients, three died within 30 days before EBV removal was considered. Another patient had a second bronchoscopy with an additional EBV implanted but died within 30 days of the second bronchoscopy. The fifth patient had advanced lung cancer and removal of EBV was deemed unnecessary; she died 9 months after bronchoscopy. The sixth patient had severe chronic obstructive pulmonary disease and there was subjective improvement of respiratory and health status after EBV implantation, so it was decided that the EBV should remain in situ indefinitely.
 
Discussion
We have reported the first real-life cohort study that consisted entirely of spontaneous pneumothorax cases with persistent air leak treated with EBV. Our results confirm those of previous reports that EBV can be useful in hastening air-leak cessation in patients with this condition. Nonetheless, it also highlights the fact that failures are common. A site for EBV implantation was not identified in nearly half of the patients. For the 19 patients with an EBV implanted, only eight (42%) had a clear success. The overall success rate was thus only 22%.
 
In this retrospective study, we attempted to assess the efficacy of EBV treatment by comparing those who had an EBV implanted with those in whom EBV implantation was denied owing to inability to identify an implantation site or lung function that was too poor. This design is obviously inferior to a prospective randomised design. Nevertheless, the two study groups had a similar number of patients who were similar in many demographic and disease aspects. The Kaplan-Meier comparison suggested that EBV treatment is efficacious in hastening air-leak cessation. The significantly longer duration of air leakage before first bronchoscopy for the EBV group is a potential confounder, because the longer the air leak exists, the higher the probability that spontaneous resolution will occur. Nonetheless, the Cox hazards regression analysis suggested that it was not a significant factor. This finding also lends further support to the efficacy of EBV implantation in enabling early air-leak cessation.
 
A definition of success for using EBV implantation to treat persistent air leak complicating pneumothorax is difficult to formulate and has seldom been discussed in previous studies. Our criteria of cessation of air leak within 72 hours was arbitrary but is supported by our observation that patients who did not meet the criteria behaved almost exactly as if no EBV was implanted. This finding suggests that for cases in which EBV is implanted but fails, other forms of treatment should be sought early.
 
One interesting finding is that higher co-morbidity burden seemed to be a risk factor for delayed air-leak cessation, irrespective of EBV implantation status. Delayed cessation of air leak may be translated as delayed healing of the lesion responsible for the air leak, which is expected in patients with more co-morbidities and who are thus usually sicker. Nonetheless, our study was not designed to examine this association, and a properly designed study would be needed to provide definitive answers.
 
It has been widely accepted that fissure completeness of the target lobe is strongly correlated with significant lobar collapse after implantation of EBVs for volume reduction in severe pulmonary emphysema.28 29 The same is probably true for EBV treatment of persistent air leak complicating pneumothorax, although as far as we are aware there are no published data on this. Not surprisingly, our results showed that intact interlobar fissures were a necessary, but not sufficient, condition for a successful outcome. However, an interesting and unexpected finding is that patients with a non-intact fissure in the contralateral lung, but intact fissure in the ipsilateral lung, behaved similarly to those with a non-intact fissure in the ipsilateral lung. We have no explanation for this, and these findings need to be confirmed with larger studies.
 
Immediate cessation of air leak after completion of EBV implantation was strongly predictive of a successful outcome. Importantly, all eight patients with an implanted EBV and without immediate cessation of air leak failed to respond to treatment. In such cases, the EBV should have been removed immediately to save costs and to avert possible adverse events associated with EBVs. A further implication is that stringent balloon testing of cessation of air leak should be performed and, in the presence of any uncertainty, EBVs should not be implanted.
 
In all our patients, we encountered no adverse events directly attributable to EBVs. Nonetheless, the three early deaths within 30 days of EBV implantation is worrying. The causes of death in two cases were clearly the severe pre-existing illness, but the possibility that recent EBV implantation hastened the terminal event cannot be excluded. The case of sudden cardiac death raises suspicion that the implanted EBV was implicated. More data are needed to determine whether EBV implantation is associated with increased early mortality.
 
There were several limitations to our study. First, it was a retrospective one, so some data were unavailable. Second, it was not a randomised controlled study. Third, it was a single-centre study, making the data less generalisable to a wider setting. Still, single-centre retrospective case series do have the advantage that practices are more uniform and results easier to interpret. Fourth, our case series was small, making it difficult to identify definitive factors associated with clinical outcomes. Fifth, some groups had a very small number of patients, and especially of females, patients with primary spontaneous pneumothorax, and patients with a lesion in the lower lobes. Finally, chart review and data collection (besides radiological data) were performed by an investigator who was not blinded to the study outcomes, and this may have been a source of bias.
 
We conclude that EBV implantation via the flexible bronchoscope can be useful in hastening air-leak cessation in patients with persistent air leak complicating spontaneous pneumothorax. Only about one-fifth of subjects, however, showed unequivocal benefit, and safety of this form of treatment needs further evaluation. Bilateral intact interlobar fissures seem to be a necessary, though not sufficient, condition for treatment success, and patients with fewer medical co-morbidities and immediate air-leak cessation after completion of EBV implantation seem to have a higher likelihood of treatment success. Further evaluation by randomised controlled trials is warranted.
 
Acknowledgements
We thank the doctors and nurses of the Respiratory Team, Princess Margaret Hospital, for taking care of the patients, and nurses of the Central Endoscopy Unit, Princess Margaret Hospital, for providing technical support for the bronchoscopy procedures.
 
Declaration
The authors have no conflicts of interest to disclose.
 
References
1. Mathur R, Cullen J, Kinnear WJ, Johnston ID. Time course of resolution of persistent air leak in spontaneous pneumothorax. Respir Med 1995;89:129-32. CrossRef
2. Chee CB, Abisheganaden J, Yeo JK, et al. Persistent air-leak in spontaneous pneumothorax—clinical course and outcome. Respir Med 1998;92:757-61. CrossRef
3. Snell G, Holsworth L, Fowler S, et al. Occlusion of a broncho-cutaneous fistula with endobronchial one-way valves. Ann Thorac Surg 2005;80:1930-2. CrossRef
4. Ferguson JS, Sprenger K, Van Natta T. Closure of a bronchopleural fistula using bronchoscopic placement of an endobronchial valve designed for the treatment of emphysema. Chest 2006;129:479-81. CrossRef
5. Mitchell KM, Boley TM, Hazelrigg SR. Endobronchial valves for treatment of bronchopleural fistula. Ann Thorac Surg 2006;81:1129-31. CrossRef
6. Feller-Kopman D, Bechara R, Garland R, Ernst A, Ashiku S. Use of a removable endobronchial valve for the treatment of bronchopleural fistula. Chest 2006;130:273-5. CrossRef
7. Fann JI, Berry GJ, Burdon TA. The use of endobronchial valve device to eliminate air leak. Respir Med 2006;100:1402-6. CrossRef
8. Anile M, Venuta F, De Giacomo T, et al. Treatment of persistent air leakage with endobronchial one-way valves. J Thorac Cardiovasc Surg 2006;132:711-2. CrossRef
9. Toma TP, Kon OM, Oldfield W, et al. Reduction of persistent air leak with endoscopic valve implants. Thorax 2007;62:830-3. CrossRef
10. Santini M, Fiorelli A, Vicidomini G, Laperuta P, Di Crescenzo VG. Iatrogenic air leak successfully treated by bronchoscopic placement of unidirectional endobronchial valves. Ann Thorac Surg 2010;89:2007-10. CrossRef
11. Yu WC, Yeung YC, Chang Y, et al. Use of endobronchial one-way valves reveals questions on etiology of spontaneous pneumothorax: report of three cases. J Cardiothorasc Surg 2009;4:63. CrossRef
12. Schiavon M, Marulli G, Zuin A, et al. Endobronchial valve for secondary pneumothorax in a severe emphysema patient. Thorac Cardiovasc Surg 2011;59:509-10. CrossRef
13. El-Sameed Y, Waness A, Al Shamsi I, Mehta AC. Endobronchial valves in the management of broncho-pleural and alveolo-pleural fistulae. Lung 2012;190:347-51. CrossRef
14. Ambrosino N, Ribechini A, Allidi F, Gabbrielli L. Use of endobronchial valves in persistent air leaks: a case report and review of the literature. Expert Rev Respir Med 2013;7:85-90. CrossRef
15. Travaline JM, McKenna RJ Jr, De Giacomo T, et al. Treatment of persistent pulmonary air leaks using endobronchial valves. Chest 2009;136:355-60. CrossRef
16. Gillespie CT, Sterman DH, Cerfolio RJ, et al. Endobronchial valve treatment for prolonged air leaks of the lung: a case series. Ann Thorac Surg 2011;91:270-3. CrossRef
17. Mahajan AK, Verhoef P, Patel SB, Carr G, Kyle Hogarth D. Intrabronchial valves: a case series describing a minimally invasive approach to bronchopleural fistulas in medical intensive care unit patients. J Bronchol Intervent Pulmonol 2012;19:137-41. CrossRef
18. Firlinger I, Stubenberger E, Müller MR, Burghuber OC, Valipour A. Endoscopic one-way valve implantation in patients with prolonged air leak and the use of digital air leak monitoring. Ann Thorac Surg 2013;95:1243-9. CrossRef
19. Dooms CA, Decaluwe H, Yserbyt J, De Leyn P, Van Raemdonck D, Ninane V. Bronchial valve treatment for pulmonary air leak after anatomical lung resection for cancer. Eur Respir J 2014;43:1142-8. CrossRef
20. Cordovilla R, Torracchi AM, Novoa N, et al. Endobronchial valves in the treatment of persistent air leak, an alternative to surgery [in English, Spanish]. Arch Bronconeumol 2015;51:10-5. CrossRef
21. Reed MF, Gilbert CR, Taylor MD, Toth JW. Endobronchial valves for challenging air leaks. Ann Thorac Surg 2015;100:1181-6. CrossRef
22. Hance JM, Martin JT, Mullett TW. Endobronchial valves in the treatment of persistent airleaks. Ann Thorac Surg 2015;100:1780-6. CrossRef
23. Gilbert CR, Casal RF, Lee HJ, et al. Use of one-way intrabronchial valves in air leak management after tube thoracostomy drainage. Ann Thorac Surg 2016;101:1891-6. CrossRef
24. Miller MR, Hankinson J, Bursasco V, et al. Standardisation of spirometry. Eur Respir J 2005;26:319-38. CrossRef
25. Ip MS, Ko FW, Lau AC, et al. Updated spirometric reference values for adult Chinese in Hong Kong and implications on clinical utilization. Chest 2006;129:384-92. CrossRef
26. Sciurba FC, Ernst A, Herth FJ, et al. A randomized study of endobronchial valves for advanced emphysema. N Engl J Med 2010;363:1233-44. CrossRef
27. von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP; STROBE Initiative. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. J Clin Epidemiol 2008;61:344-9. CrossRef
28. Koster TD, van Rikxoort EM, Huebner RH, et al. Predicting lung volume reduction after endobronchial valve therapy is maximized using a combination of diagnostic tools. Respiration 2016;92:150-7. CrossRef
29. Schuhmann M, Raffy P, Yin Y, et al. Computed tomography predictors of response to endobronchial valve lung reduction treatment. Comparison with Chartis. Am J Respir Crit Care Med 2015;191:767-74. CrossRef

Bacteriology and risk factors associated with periprosthetic joint infection after primary total knee arthroplasty: retrospective study of 2543 cases

Hong Kong Med J 2018;24:Epub 29 Mar 2018
DOI: 10.12809/hkmj176885
ORIGINAL ARTICLE
Bacteriology and risk factors associated with periprosthetic joint infection after primary total knee arthroplasty: retrospective study of 2543 cases
KT Siu1; FY Ng2; PK Chan1; Henry CH Fu1; CH Yan3; KY Chiu3
1 Department of Orthopaedics and Traumatology, Queen Mary Hospital, Pokfulam, Hong Kong
2 Private practice, Hong Kong
3 Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong
 
Corresponding author: Prof KY Chiu (pkychiu@hkucc.hku.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Periprosthetic joint infection after total knee arthroplasty is a serious complication. This study aimed to identify risk factors and bacteriological features associated with periprosthetic joint infection after primary total knee arthroplasty performed at a teaching hospital.
 
Methods: We reviewed 2543 elective primary total knee arthroplasties performed at our institution from 1993 to 2013. Data were collected from the Hong Kong Hospital Authority’s Clinical Data Analysis and Reporting System, the Infection Control Team, and the joint replacement division registry. The association between potential risk factors and periprosthetic joint infection was examined by univariable analysis and multivariable logistic regression. Univariable analyses were also performed to examine the association between potential risk factors and bacteriology and between potential risk factors, including bacteriology, and early-onset infection.
 
Results: The incidence of periprosthetic joint infection in our series was 1.34% (n=34). The incidence of early-onset infection was 0.39% (n=24). Of the periprosthetic joint infections, 29.4% were early-onset infections. In both univariable and multivariable analyses, only rheumatoid arthritis was a significant predictor of periprosthetic joint infection. Methicillin-sensitive Staphylococcus aureus was the most common causative organism. We did not identify any significant association between potential risk factors and bacteriology. Periprosthetic joint infection caused by skin flora was positively associated with early-onset infection but the association was not statistically significant.
 
Conclusion: The incidence of periprosthetic joint infection after elective primary total knee arthroplasty performed at our institution from 1993 to 2013 was 1.34%. Rheumatoid arthritis was a significant risk factor for periprosthetic joint infection.
 
 
New knowledge added by this study
  • The incidence of periprosthetic joint infection after elective primary total knee arthroplasty performed at our institution from 1993 to 2013 was 1.34%.
  • Rheumatoid arthritis was the only significant risk factor identified in the series.
Implications for clinical practice or policy
  • Early-onset infection may be associated with infection with skin flora. Therefore, in early-onset periprosthetic joint infection with negative cultures, an empirical antibiotic regimen should preferably provide adequate coverage against skin flora organisms.
 
 
Introduction
Periprosthetic joint infection (PJI) is an uncommon but serious complication after total knee arthroplasty (TKA). Treatment is often challenging and has a major impact on the patient. Multiple operations are often required and patients may suffer from a long period of disability. Moreover, PJI incurs considerable health care costs.1 2 3 Therefore, multiple strategies including antibiotic prophylaxis, body exhaust systems, and laminar airflow systems have been developed to reduce the incidence of PJI. Studies have also identified modifiable risk factors for PJI after elective total joint replacement,4 5 6 7 8 9 10 11 12 13 14 with the aim of further reducing the incidence of PJI. However, local data on the risk factors and bacteriological features associated with PJI are still lacking.
 
This study had several aims. First, it aimed to provide the most up-to-date local data on incidence of and risk factors for PJI, including age, sex, presence of diabetes, presence of rheumatoid arthritis, and one-stage bilateral TKA. Second, this study aimed to provide an update on the bacteriology of PJI after elective primary TKA and to examine the association between potential risk factors and bacteriology. Third, we attempted to determine which risk factors, including bacteriology, were more likely to be associated with early-onset infection after elective primary TKA.
 
It is hoped that risk factors can be optimised or modified to prevent infection after TKA. Furthermore, an improved understanding of local bacteriological patterns and their relationship with various risk factors can help guide antimicrobial therapy.
 
Methods
We reviewed 2543 elective primary TKAs performed at the Queen Mary Hospital, Hong Kong, from 1993 to 2013. Data were collected by an infection control nurse of the Department of Microbiology who was blinded to the study objectives. The cohort data were collected from the Hong Kong Hospital Authority’s Clinical Data Analysis and Reporting System, the Infection Control Team, and the hospital’s joint replacement division registry. The keywords used in the data search were ‘periprosthetic joint infection’, ‘total knee arthroplasty’, and ‘surgical site infection’. Revision arthroplasties and knee arthroplasties for malignant conditions were excluded from the study. In patients with a history of native joint infection, elective primary TKA was performed only after eradication of the infection. Patients with active bacteraemia were also precluded from elective primary TKA until they were infection-free. There were no cases of severe immunosuppression. In relation to infection control, the majority of perioperative protocols for primary TKA were the same throughout the study period. Preoperatively, intravenous antibiotic prophylaxis (1 g of cefazolin) was given within 1 h before skin incision. In patients with penicillin allergy, other antibiotics were prescribed as appropriate. Intra-operatively, laminar airflow and body exhaust systems were used. There was no routine use of antibiotic-loaded cement or postoperative antibiotics. Postoperative wound management was the same throughout the study period.
 
Cohort characteristics, occurrence of PJI, and bacteriological data were retrieved. Bacterial type was defined as infection with skin flora or non-skin flora. Skin flora included methicillin-susceptible Staphylococcus aureus (MSSA), methicillin-resistant S aureus (MRSA), methicillin-susceptible coagulase-negative staphylococci (MSCNS), and methicillin-resistant coagulase-negative staphylococci (MRCNS). Other organisms were considered non-skin flora.
 
The following potential risk factors for PJI were analysed: age, sex, presence of diabetes, presence of rheumatoid arthritis, and one-stage bilateral TKA. They were examined by univariable analyses and then multivariable logistic regression to identify potential predictors of PJI, while controlling for confounders. We also studied the association of those potential risk factors with bacteriology and with the timing of infection onset; culture-negative PJI was excluded from these analyses. According to a working party convened by the Musculoskeletal Infection Society in 2014,15 PJI that occurs within 90 days of the index operation is considered early-onset infection, whereas PJI that occurs later is considered late-onset infection.
 
Both univariable and multivariable logistic regression in this study used the simultaneous entry method, with covariates of age (as a continuous variable) and sex, diabetes, rheumatoid arthritis, and one-stage bilateral TKA (as dichotomous variables). Outcomes are presented as odds ratios (ORs) with 95% confidence intervals (CIs). The regression model and data fitting were assessed using the Hosmer–Lemeshow goodness-of-fit test, and diabetes and one-stage bilateral TKA were excluded from the final model because of poor goodness-of-fit. For associations between potential risk factors and bacteriology and between potential risk factors and early onset of infection, only univariable analyses were used owing to small numbers of events. Categorical variables were compared with the chi-square test, whereas age was compared with the independent t test (two-tailed). Significance was assumed if P<0.05. All statistical analyses were conducted using SPSS version 22.0 (IBM Corporation, Armonk [NY], United States). The study was conducted in accordance with the principles outlined in the Declaration of Helsinki.
 
Results
The incidence of PJI in our series was 1.34% (n=34). The incidence of early-onset infection was 0.39% (n=10) and that of late-onset infection was 0.94% (n=24). Among the cases PJI, 29.4% were early-onset infection. Early-onset infection occurred within a median of 17 days after arthroplasty (interquartile range, 9-32 days). Late-onset infection occurred within a median of 1 year and 8 months after arthroplasty (interquartile range, 7 months to 2 years and 11 months). Fifty-nine percent of infections occurred in the first year of surgery, whereas 74% occurred in the first 2 years.
 
The mean (standard deviation) age was 69 (9) years, with a range from 21 to 91 years; age followed a normal distribution. Overall, PJI developed in 10 males (1.9%) and 24 females (1.2%). In the one-stage bilateral TKA group, PJI occurred in 13 knees (1.2%). For the single-side TKA group, 21 knees (1.4%) developed PJI. Nine patients with diabetes (1.9%) and 25 patients without diabetes (1.2%) developed PJI. The highest rate of PJI, at 3.1%, was found in patients with rheumatoid arthritis, compared with 1.2% in patients without rheumatoid arthritis. The descriptive data are summarised in Table 1.
 

Table 1. Descriptive statistics for potential risk factors according to occurrence of periprosthetic joint infection after primary total knee arthroplasty
 
The most frequent causative organism was MSSA (26.5%, n=9), followed by MRSA (17.6%, n=6), Streptococcus spp (8.8%, n=3), MSCNS (5.9%, n=2), Escherichia coli (5.9%, n=2), Salmonella (5.9%, n=2), MRCNS (2.9%, n=1) and Mycobacterium tuberculosis (2.9%, n=1), The three cases of streptococcal infection comprised two Streptococcus dysgalactiae infections and one Streptococcus agalactiae infection. Culture-negative PJI comprised 23.5% of cases (n=8). Methicillin-resistant strains constituted 39% of all staphylococcal organisms. There was no significant association between the potential risk factors and skin flora infection (Table 2).
 

Table 2. Association between potential risk factors for periprosthetic joint infection after primary total knee arthroplasty and bacteriology
 
Rheumatoid arthritis was a significant risk factor of PJI in the univariable analysis, with an OR of 2.67 (95% CI, 1.15-6.20; P=0.02), as well as in the multivariable analysis, with an OR of 3.12 (CI, 1.29-7.56; P=0.01) [Table 3]. Being male (OR=1.9; P=0.11 in the multivariable analysis) and having diabetes (OR=1.54; P=0.27 in the univariable analysis) were not significantly associated with PJI.
 

Table 3. Results of univariable and multivariable analyses of potential risk factors for periprosthetic joint infection after primary total knee arthroplasty
 
Age (P=0.655), sex (P=0.961), diabetes (P=0.462), and rheumatoid arthritis (P=0.315) were not associated with early-onset infection (Table 4). Infection caused by skin flora was associated with early-onset infection (P=0.099), but the association was not statistically significant.
 

Table 4. Association between potential risk factors for periprosthetic joint infection after primary total knee arthroplasty and onset of infection
 
Discussion
In this study, the incidence of PJI after primary TKA was 1.34% and the incidence of early-onset infection was 0.39%. The majority of PJIs (70%) were late-onset infections. The reported incidence of PJI after primary TKA ranges from 1.1% to 2.18%.16 17 18 Pulido et al16 reported the incidence of PJI after TKA to be 1.1%, of which 27% were diagnosed during the first 30 days after arthroplasty, and a majority of 65% were diagnosed in the first year after surgery. In our study, the average time to diagnosis was 431 days after the index surgery (range, 11-1699 days).
 
Rheumatoid arthritis was a significant risk factor for PJI after primary TKA. This finding is in keeping with the current literature.6 8 11 Although various authors have found male sex to be a risk factor for PJI,4 19 20 the association was not significant in this study. The OR of 1.9 may be of clinical importance but not significant as a result of the small number of PJIs and inadequate statistical power. The correlation between age and PJI has been a matter of controversy, with some reports mentioning young age as a risk factor for PJI4 21 and some otherwise.22 In our study, age was not associated with PJI occurrence. For one-stage bilateral TKA, age has been a controversial risk factor for PJI. Some studies16 23 have suggested that one-stage bilateral TKA is associated with an increased risk of superficial and deep infection. Hussain et al24 nonetheless reported a similar infection rate between one- and two-stage bilateral TKA. Our study did not find an association between one-stage bilateral TKA and PJI occurrence.
 
The local bacteriological pattern for PJI was comparable to that reported in the literature.4 16 In our study, skin flora and gram-positive bacteria were the most commonly isolated organisms, followed by gram-negative bacteria such as Escherichia coli and Salmonella. Coagulase-negative staphylococci were the most common causative organism in one study.4 In contrast, in our series, S aureus was the most common causative organism, particularly methicillin-sensitive strains. Methicillin-resistant strains were less common in our series, constituting 39% of all staphylococcal organisms.
 
Other authors have reported that male sex is a risk factor for PJI, which may be related to a sex difference in immune response to pathogenic bacteria. Studies6 have shown that males (compared with females) have a significantly higher likelihood of being a persistent S aureus carrier. However, our study did not support male sex as a risk factor for infection with skin flora. With regard to onset of infection, PJI caused by skin flora was positively associated with early-onset infection, although the association did not reach statistical significance (P=0.099). Direct inoculation and spread from contiguous foci of infection are more common in early-onset infection caused by wound complications and local soft-tissue conditions. In contrast, distant foci of infection, such as in bacteraemia, play a more important role in late-onset infection. Therefore, in early-onset periprosthetic joint infection with negative cultures, an empirical antibiotic regimen may provide adequate coverage against skin flora organisms.
 
Fan et al20 reported 479 TKAs and rates of 1.9% for superficial wound infection, 0.2% for early deep infection (n=1), and 0.6% for late deep infection (n=2). Methicillin-sensitive S aureus and coagulase-negative staphylococci were causative organisms. Lee et al25 reviewed 1133 primary TKAs and found a 0.71% incidence of PJI. The most common causative organisms in descending order were methicillin-sensitive S aureus, coagulase-negative staphylococci, methicillin-resistant S aureus, and Pseudomonas aeruginosa. This finding is in keeping with our data. Among risk factors identified by Lee et al25 were young age, diabetes, anaemia, thyroid disease, heart disease, lung disease, and long operating time. However, the researchers identified limitations of having only a small number of patients with infection (n=8) and insufficient power for analysis. In addition, multivariable analysis should have been performed to account for the effect of confounders among the multiple risk factors. They also reported the limitation that the mean follow-up duration was only 2 years. A short follow-up period may underestimate the occurrence of late-onset infection.
 
Our study has several limitations. The number of PJI-positive cases was small and thus subgroup analysis was limited. This study included subjects treated at a single centre in Hong Kong; multicentre studies may improve the representativeness of local data. In addition, perioperative management for elective TKA has evolved over the past 20 years, including the introduction of an MRSA-screening programme in 2011. In the screening programme, a nasal swab is taken from all elective joint-replacement patients. Patients with a positive result are prescribed 5 days of decolonisation therapy including a daily chlorhexidine bath. Furthermore, intravenous vancomycin is now administered for prophylaxis instead of cefazolin.26
 
There are many potential risk factors for PJI documented in the literature. Nonetheless, only a limited number were included in this study, most of which are not be modifiable. Thus, it may not provide the necessary guidance for preoperative optimisation. Furthermore, the exclusion of some potential risk factors may have led to inadequate control for potential confounding factors. Inclusion of more risk factors with better characterisation is needed to provide a more comprehensive understanding and to better account for the confounding effect of other variables.
 
Conclusion
The incidence of PJI after elective primary TKA in our institution over two decades from 1993 to 2013 was 1.34%. Rheumatoid arthritis was a significant risk factor for PJI in this series. In the early-onset infection group, PJI was caused by skin flora, but this was not statistically significant. It is hoped that this study has updated the local data for PJI after primary TKA and serves as a model for future related studies.
 
Acknowledgements
We thank colleagues from the Department of Orthopaedics and Traumatology and the Infection Control Team at the Queen Mary Hospital for their assistance in data collection, and those who advised on this project to make its publication possible.
 
Declaration
The authors have no conflicts of interest to disclose.
 
References
1. Kurtz SM, Lau E, Watson H, Schmier JK, Parvizi J. Economic burden of periprosthetic joint infection in the United States. J Arthroplasty 2012;27(8 Suppl):61-5.e1. CrossRef
2. Lamarsalle L, Hunt B, Schauf M, Szwarcensztein K, Valentine WJ. Evaluating the clinical and economic burden of healthcare-associated infections during hospitalization for surgery in France. Epidemiol Infect 2013;141:2473-82. CrossRef
3. Nero DC, Lipp MJ, Callahan MA. The financial impact of hospital-acquired conditions. J Health Care Finance 2012;38:40-9.
4. Crowe B, Payne A, Evangelista PJ, et al. Risk factors for infection following total knee arthroplasty: a series of 3836 cases from one institution. J Arthroplasty 2015;30:2275-8. CrossRef
5. Meller MM, Toossi N, Johanson NA, Gonzalez MH, Son MS, Lau EC. Risk and cost of 90-day complications in morbidly and superobese patients after total knee arthroplasty. J Arthroplasty 2016;31:2091-8. CrossRef
6. Zmistowski B, Alijanipour P. Risk factors for periprosthetic joint infection. In: Springer BD, Parvizi J, editors. Periprosthetic Joint Infection of the Hip and Knee. New York: Springer; 2014: 15-40. CrossRef
7. Jämsen E, Huhtala H, Puolakka T, Moilanen T. Risk factors for infection after knee arthroplasty. A register-based analysis of 43,149 cases. J Bone Joint Surg Am 2009;91:38-47. CrossRef
8. Wilson MG, Kelley K, Thornhill TS. Infection as a complication of total knee-replacement arthroplasty. Risk factors and treatment in sixty-seven cases. J Bone Joint Surg Am 1990;72:878-83. CrossRef
9. Namba RS, Inacio MC, Paxton EW. Risk factors associated with deep surgical site infections after primary total knee arthroplasty: an analysis of 56,216 knees. J Bone Joint Surg Am 2013;95:775-82. CrossRef
10. Pruzansky JS, Bronson MJ, Grelsamer RP, Strauss E, Moucha CS. Prevalence of modifiable surgical site infection risk factors in hip and knee joint arthroplasty patients at an urban academic hospital. J Arthroplasty 2014;29:272-6. CrossRef
11. Chesney D, Sales J, Elton R, Brenkel IJ. Infection after knee arthroplasty: a prospective study of 1509 cases. J Arthroplasty 2008;23:355-9. CrossRef
12. Moucha CS, Clyburn T, Evans RP, Prokuski L. Modifiable risk factors for surgical site infection. J Bone Joint Surg Am 2011;93:398-404.
13. Peersman G, Laskin R, Davis J, Peterson M. Infection in total knee replacement: a retrospective review of 6489 total knee replacements. Clin Orthop Relat Res 2001;392:15-23. CrossRef
14. Rasouli MR, Restrepo C, Maltenfort MG, Purtill JJ, Parvizi J. Risk factors for surgical site infection following total joint arthroplasty. J Bone Joint Surg Am 2014;96:e158. CrossRef
15. Parvizi J, Gehrke T; International Consensus Group on Periprosthetic Joint Infection. Definition of periprosthetic joint infection. J Arthroplasty 2014;29:1331. CrossRef
16. Pulido L, Ghanem E, Joshi A, Purtill JJ, Parvizi J. Periprosthetic joint infection: the incidence, timing, and predisposing factors. Clin Orthop Relat Res 2008;466:1710-5. CrossRef
17. Tsaras G, Osmon DR, Mabry T, et al. Incidence, secular trends, and outcomes of prosthetic joint infection: a population based study, Olmsted County, Minnesota, 1969-2007. Infect Control Hosp Epidemiol 2012;33:1207-12. CrossRef
18. Tande AJ, Patel R. Prosthetic joint infection. Clin Microbiol Rev 2014;27:302-45. CrossRef
19. Herwaldt LA, Cullen JJ, French P, et al. Preoperative risk factors for nasal carriage of Staphylococcus aureus. Infect Control Hosp Epidemiol 2004;25:481-4. CrossRef
20. Fan JC, Hung HH, Fung KY. Infection in primary total knee replacement. Hong Kong Med J 2008;14:40-5.
21. Meehan JP, Danielsen B, Kim SH, Jamali AA, White RH. Younger age is associated with a higher risk of early periprosthetic joint infection and aseptic mechanical failure after total knee arthroplasty. J Bone Joint Surg Am 2014;96:529-35. CrossRef
22. Berbari EF, Osmon DR, Lahr B, et al. The Mayo prosthetic joint infection risk score: implication for surgical site infection reporting and risk stratification. Infect Control Hosp Epidemiol 2012;33:774-81. CrossRef
23. Luscombe JC, Theivendran K, Abudu A, Carter SR. The relative safety of one-stage bilateral total knee arthroplasty. Int Orthop 2009;33:101-4. CrossRef
24. Hussain N, Chien T, Hussain F, et al. Simultaneous versus staged bilateral total knee arthroplasty: a meta-analysis evaluating mortality, peri-operative complications and infection rates. HSS J 2013;9:50-9. CrossRef
25. Lee QJ, Mak WP, Wong YC. Risk factors for periprosthetic joint infection in total knee arthroplasty. J Orthop Surg (Hong Kong) 2015;23:282-6. CrossRef
26. Cheng VC, Tai JW, Wong ZS, et al. Transmission of methicillin-resistant Staphylococcus aureus in the long term care facilities in Hong Kong. BMC Infect Dis 2013;13:205. CrossRef

Intra-operative periarticular multimodal injection in total knee arthroplasty: a local hospital experience in Hong Kong

Hong Kong Med J 2018;24:Epub 14 Mar 2018
DOI: 10.12809/hkmj176804
ORIGINAL ARTICLE
Intra-operative periarticular multimodal injection in total knee arthroplasty: a local hospital experience in Hong Kong
Jason CH Fan, FHKAM (Orthopaedic Surgery)
Department of Orthopaedics and Traumatology, Alice Ho Miu Ling Nethersole Hospital, Tai Po, Hong Kong
 
Corresponding author: Dr Jason CH Fan (fchjason@gmail.com)
 
 Full paper in PDF
 
Abstract
Introduction: Data from a local report revealed the superior outcome of regional anaesthesia and analgesia compared with general anaesthesia and intravenous patient-controlled analgesia in total knee arthroplasty. This retrospective study aimed to assess the efficacy of intra-operative periarticular multimodal injection in improving postoperative pain and reducing morphine consumption with patient-controlled analgesia after total knee arthroplasty in patients with knee osteoarthritis.
 
Methods: From July 2005 to May 2009, 213 total knee arthroplasties without intra-operative periarticular multimodal injection (control group) were performed at a local hospital. From June 2009 to December 2012, 185 total knee arthroplasties were performed with intra-operative periarticular multimodal injection (cocktail group). The inclusion criteria were osteoarthritis of the knee, single method of anaesthesia (general or neuraxial), simple total knee arthroplasty without any metal augmentation or constraint, and postoperative patient-controlled analgesia. Postoperative patient-controlled morphine doses were compared.
 
Results: A total of 152 total knee arthroplasties were recruited to the cocktail group, and 89 to the control group. Duration of tourniquet application and preoperative knee score did not significantly correlate with morphine consumption by patient-controlled analgesia. Multimodal injection significantly decreased such consumption for 36 h. When injection was separately analysed for general and neuraxial anaesthesia, the effect lasted for 42 h and 24 h, respectively.
 
Conclusion: Intra-operative periarticular multimodal injection decreased morphine consumption for up to 42 h postoperatively.
 
 
New knowledge added by this study
  • Intra-operative periarticular multimodal injection in total knee arthroplasty could decrease parenteral morphine consumption for up to 42 hours.
Implications for clinical practice or policy
  • Intra-operative periarticular multimodal injection should be adopted as a standard local practice for postoperative pain control. This practice may be extended to operations other than total knee arthroplasty.
 
 
Introduction
Postoperative pain following total knee arthroplasty (TKA) is reported to be severe in approximately 60% of patients and moderate in approximately 30%.1 It is associated with arthrofibrosis and diminished range of motion.2 3 Good pain relief is important for rehabilitation following TKA.4 Many modes of perioperative and postoperative analgesia are available, and involve various combinations of systemic and regional analgesia. Intra-operative periarticular multimodal drug injection has been well documented as an excellent method to alleviate postoperative pain following TKA.5 6 7 Nonetheless, a previous retrospective study5 and a randomised trial6 that analysed two different groups of patients with multiple diagnoses and multiple anaesthetic methods revealed that the effect of periarticular injection might have been affected by different causes of end-stage arthritis leading to TKA. Different anaesthetic methods could also have affected patients’ perception of pain and parenteral morphine consumption.
 
In 2006, Chu et al8 reported the superior outcome of regional anaesthesia and regionally delivered analgesia compared with general anaesthesia (GA) and intravenous patient-controlled analgesia (PCA) in TKA at the Alice Ho Miu Ling Nethersole Hospital (AHNH). Since June 2009, intra-operative periarticular multimodal injection (IPMI) consisting of an opioid (morphine), a long-acting local anaesthetic (levobupivacaine) and epinephrine, has been administered by surgeons to control postoperative pain following TKA. This retrospective cohort study analysed the efficacy of IPMI in TKA and also its effect following different types of anaesthesia.
 
Methods
Perioperative pain management
Before June 2009, postoperative pain following primary TKA was managed by a combination of parenteral and oral analgesia. The anaesthetist determined the choice of parenteral analgesia that included regular or as-required subcutaneous morphine injection, PCA with intravenous morphine injection, or epidural analgesia (EpA). Oral paracetamol 1 g every 6 h was prescribed to all patients from day 1 to 3. Since June 2009, IPMI has been routinely added, and comprises 20 mL of 0.5% levobupivacaine, 1 mL of 5 mg/mL morphine, 2 mL of 1:10000 adrenaline, and 17 mL of normal saline. In this study, half of this 40-mL mixture was injected into the posterior capsule, collaterals, and quadriceps incision before implantation of the prosthesis. The other half was injected into the subcutaneous tissue after suturing of the arthrotomy. All patients with PCA were assessed hourly for the first 24 h to monitor vital signs, pain score, and patient-controlled analgesia morphine consumption (PCAMC), and thereafter every 6 h for 2 more days.
 
Patient selection
From July 2005 to May 2009, 213 TKAs without IPMI (control group) were performed at AHNH. They included 196 knees with osteoarthritis (OA) and 17 knees with rheumatoid arthritis. From June 2009 to December 2012, 185 TKAs were performed with IPMI (cocktail group). There were 175 OA knees, nine rheumatoid arthritis knees, and one Charcot knee.
 
All TKAs were performed through an anterior midline incision and medial parapatellar arthrotomy with tourniquet pressure of 300 mm Hg. A cemented posterior stabilised model was used in all cases except for two cases in the control group and nine cases in the cocktail group where a semi-constrained TKA was performed. All operations were performed under GA or neuraxial anaesthesia (NeA) that was either combined spinal epidural or spinal anaesthesia. Four TKAs in the cocktail group and four in the control group were performed with combined GA and NeA. A closed-suction surgical drain was inserted and was routinely removed on postoperative day 2.
 
For postoperative pain control in the control group, PCA was used in 112 TKAs, EpA in 66 TKAs, and subcutaneous morphine injections in 10 TKAs. A 4-point pain scale was completed by a pain nurse to assess pain in 189 PCA patients before October 2008 and a 10-point pain scale used in 23 PCA patients thereafter. In the cocktail group, 152 TKAs were managed with PCA, three with EpA, and three with subcutaneous morphine injections. Pain in all PCA patients was assessed by a 10-point pain scale.
 
The inclusion criteria for this study were OA of the knee, single method of anaesthesia of either GA or NeA, simple TKA without any metal augmentation or constraint, and postoperative PCA. The patients in the control group who were assessed by the 10-point pain scale were excluded to ensure a common pain assessment tool for each group.
 
Method of data retrieval, analysis, and study hypothesis
This was a retrospective cohort observational study carried out in accordance with the principles outlined in the Declaration of Helsinki. Informed patient consent was not required because it was a record-based study that revealed no individual identities or sensitive individual information. The medical records and electronic patient records were traced and the necessary data—including demographic data, TKA model and anaesthetic method, first 72-hour pain score and morphine consumption, and postoperative complications—were entered into an electronic file by a single member of staff blinded to the study hypothesis. The accuracy of the data was selectively double-checked by the author. To enable comparison, pain score was divided by 4 when the 4-point scale was used and by 10 for the 10-point scale. Statistical Package for the Social Sciences (Windows version 13.0; SPSS Inc, Chicago [IL], United States) was used for analysis. The null hypothesis was that IPMI would not alleviate postoperative pain and would not reduce PCAMC. The Chi squared test and two-tailed independent t test were used to analyse categorical and continuous data, respectively. The Pearson correlation test was used to detect any relationship between cumulative PCAMC and tourniquet time, and between PCAMC and preoperative knee score. Statistical significance was set at P<0.05.
 
Results
Perioperative variables
A total of 152 knees (134 patients) in the cocktail group and 89 knees (76 patients) in the control group were recruited (Fig 1). Table 1 shows the demographic data and clinical characteristics of patients, and Table 2 shows the models of primary TKAs and anaesthetic methods. There was no statistically significant difference in age, sex, the side operated on, and mean preoperative knee score or function score between the cocktail and control groups. Tourniquet time was significantly longer in the control group (P<0.05). There was no correlation between tourniquet time and PCAMC for any postoperative period (all correlation coefficients <0.1 and P>0.05). This indicated that tourniquet time was not confounding. Preoperative knee score was not correlated with PCAMC (all correlation coefficients <0.2 and P>0.05). Comparison of the number of Press Fit Condylar Sigma and non–Press Fit Condylar models between the two groups revealed a statistical significance (P<0.001). However, all these models substitute for the posterior cruciate ligament and have a similar design. They were all used in primary simple TKA, and model type would not have caused any difference in early postoperative pain perception.
 

Figure 1. Knee recruitment in the control group and cocktail group
 

Table 1. Patient and clinical characteristics, by study group
 

Table 2. Total knee arthroplasty model and anaesthesia, by study group
 
Cumulative patient-controlled analgesia morphine consumption
The mean cumulative PCAMC in both the cocktail and control groups increased gradually until 72 h postoperatively (Fig 2). The difference between the two groups reached statistical significance in the first 36 h. When effects of GA and NeA were reviewed separately, significantly less PCA morphine was required in the cocktail group than in the control group for the first 42 h (after GA) and 24 h (after NeA).
 

Figure 2. Postoperative morphine consumption by patient-controlled analgesia after total knee arthroplasty
 
Pain scale and complication
Figure 3 shows a decreasing severity of pain for both groups in the initial 72 h after surgery. There was no statistically significant difference between groups when TKA was performed under GA. In patients who underwent TKA under NeA, patients in the control group had a lower pain scale score by 0.1 at 12 h and from 24 to 48 h compared with the cocktail group, although this was gradually reversed up to 72 h. There were no adverse effects or complications as a result of IPMI.
 

Figure 3. Postoperative pain scale score after total knee arthroplasty
 
Discussion
Severe pain following TKA may be related to bone or soft tissue trauma or hyperperfusion following tourniquet release.6 Surgical difficulty in TKA has also been found to be related to postoperative pain9 and related to bone loss, severe deformity, flexion contracture, and poor range, all of which contribute to a low preoperative knee score. Nonetheless, in this study, the duration of tourniquet application was not significantly correlated with morphine consumption; and preoperative knee score was not correlated with PCAMC.
 
Pain management for TKA should start preoperatively and intra-operatively. The preemptive use of analgesia has been shown to prevent central sensitisation and improve postoperative pain control.10 11 12 Busch et al6 conducted a randomised trial of periarticular multimodal drug injection of ropivacaine, ketorolac, epimorphine, and epinephrine in 64 TKA patients. They reported significantly lower pain scores, increased patient satisfaction scores, and decreased requirement for PCA in the first 24 hours after surgery. In another randomised trial of periarticular injection of bupivacaine, fentanyl, and methylprednisolone in either side of bilateral TKAs in 40 patients, pain scores were significantly lower, and active knee flexion ranges were greater until the fourth week after surgery.13 Maheshwari et al7 emphasised the importance of periarticular injection in multimodal pain management following TKA at the Ranawat Orthopaedic Center, United States, and PCA was no longer used because of the high rates of systemic opioid side-effects.
 
The AHNH includes morphine in multimodal injections because opioid receptors are present in peripheral inflamed tissue.14 15 They are expressed within hours of surgical trauma and are thought to be responsible for afferent sensory input to the central nervous system.16 17 The injection also includes levobupivacaine, which is pharmacokinetically similar to bupivacaine. It is a pure left-isomer and has less cardiac and central nervous system toxicity.18 Corticosteroid was not added to the injection, although studies19 20 have shown that methylprednisolone in periarticular injections in total joint surgeries caused no delayed wound healing or wound infection. Mullaji et al13 advocated cautious use of steroid for fear of increasing the risk of surgical site infection in patients who (1) had prior open surgical procedures, (2) were undergoing revision TKA, (3) had poor nutritional status, (4) were immunocompromised, (5) were rheumatoid, or (6) were diabetic. In the current study, periarticular injection of a specific mixture decreased PCAMC for up to 42 hours. In 2013, Andersen et al21 advocated the addition of ketorolac during local infiltration analgesia. They prepared the medication by mixing 150 mL of ropivacaine 2 mg/mL with 1 mL of ketorolac 30 mg/mL; to 100 mL of this mixture was added 0.5 mL of epinephrine 1 mg/mL. The mixture containing epinephrine was injected into the posterior capsule and around the prosthesis, and the 50 mL without epinephrine was injected into the fascia and subcutis. An intra-articular catheter was left in place to enable eight postoperative bolus injections of analgesic without epinephrine. It was found that ketorolac successfully reduced morphine consumption, pain intensity, and length of hospital stay.21 At the AHNH, 1 mL of ketorolac 30 mg/mL has been added to IPMI since July 2014 to provide local anti-inflammatory action and enhance the analgesic effect.
 
Regional anaesthesia is the preferred method.7 8 The previous randomised trials of multimodal drug injection in TKA involved a mixed group of GA and regional anaesthesia,6 or excluded the samples of GA.13 Randomisation of anaesthesia in clinical trials is unethical because of the obvious benefit of regional anaesthesia that avoids central nervous system depression and prevents deep vein thrombosis following TKA.22 A retrospective study stratifying different types of anaesthesia is therefore the preferred method, as in the current study. The present study revealed that IPMI in TKA under NeA could significantly decrease PCAMC for 24 hours.
 
A concordant finding could not be obtained between the effect of IPMI on PCAMC and subjective pain scale. It is possible that the greater use of PCA morphine in the control group in earlier years explained the lower postoperative pain scores. Nonetheless, this could not explain the absence of this phenomenon in the GA subgroup. Rather, it may be explained by the secular change in patient expectations. To many patients early on, TKA was well-known to be associated with a high level of pain. They may have therefore used more PCA morphine. The level of perceived pain was then less than expected with a consequent lower pain score. With increasing popularity of TKA and knowledge of IPMI, patients may have been overly optimistic about the outcome. The 4-point pain scale used in the control group may have exaggerated this discrepancy when one lower grade of pain severity was equal to a 0.25-drop in pain score compared with a 0.1-drop in the 10-point pain scale.
 
Lamplot et al23 reported that the use of periarticular injection and multimodal analgesics could lower pain scores, with fewer adverse effects, lower narcotic usage, higher patient satisfaction, and faster recovery. At the AHNH, TKA protocols for perioperative pain management, blood management, and rehabilitation were altered following the establishment of the Joint Replacement Centre in October 2015. For the pain management protocol, the hospital now uses preemptive oral pregabalin, paracetamol, and etoricoxib if not contra-indicated. The anaesthetist performs a single-injection femoral nerve block or adductor canal block before anaesthesia. Surgeons deliver IPMI. The postoperative cocktail consists of pregabalin, paracetamol, etoricoxib, and tramadol. The new protocols have made a significant contribution to the improvement in postoperative patient recovery.24 25 Further studies will be conducted on the new perioperative analgesic protocol to confirm its efficacy.
 
There were limitations to this retrospective study, which compared two groups of patients with TKA performed during different periods of time. First, possible secular changes to patient expectations and pain assessment tools might have led to discordant outcomes when IPMI was evaluated. Second, the pain scale did not focus separately on rest pain and motion pain. Third, although the data were selectively verified by the author, there might have been errors in data extraction and coding of other data. Last but not least, because TKAs were performed by more than one surgeon, it was difficult to standardise the intra-operative soft-tissue tension and balancing and the injection technique of IPMI. If the knee was made too tight or IPMI missed the quadriceps tendon, the patient would experience greater postoperative pain.
 
In conclusion, IPMI effectively decreases parenteral morphine consumption for up to 42 hours following TKA in patients with OA of the knee.
 
Declaration
The author has disclosed no conflicts of interest. No funding was received for this study.
 
References
1. Bonica JJ. Postoperative pain. In: Bonica JJ, editor. The management of pain. 2nd ed. Philadelphia: Lea and Febiger; 1990: 461-80.
2. Ranawat CS, Ranawat AS, Mehta A. Total knee arthroplasty rehabilitation protocol: what makes the difference? J Arthroplasty 2003;18(3 Suppl 1):27-30. CrossRef
3. Singelyn FJ, Deyaert M, Joris D, Pendeville E, Gouverneur JM. Effects of intravenous patient-controlled analgesia with morphine, continuous epidural analgesia, and continuous three-in-one block on postoperative pain and knee rehabilitation after unilateral total knee arthroplasty. Anesth Analg 1998;87:88-92. CrossRef
4. Shoji H, Solomonow M, Yoshino S, D’Ambrosia R, Dabezies E. Factors affecting postoperative flexion in total knee arthroplasty. Orthopedics 1990;13:643-9.
5. Lavernia C, Cardona D, Rossi MD, Lee D. Multimodal pain management and arthrofibrosis. J Arthroplasty 2008;23(6 Suppl 1):74-9. CrossRef
6. Busch CA, Shore BJ, Bhandari R, et al. Efficacy of periarticular multimodal drug injection in total knee arthroplasty. A randomized trial. J Bone Joint Surg Am 2006;88:959-63. CrossRef
7. Maheshwari AV, Blum YC, Shekhar L, Ranawat AS, Ranawat CS. Multimodal pain management after total hip and knee arthroplasty at the Ranawat Orthopaedic Center. Clin Orthop Relat Res 2009;467:1418-23. CrossRef
8. Chu CP, Yap JC, Chen PP, Hung HH. Postoperative outcome in Chinese patients having primary total knee arthroplasty under general anaesthesia/intravenous patient-controlled analgesia compared to spinal-epidural anaesthesia/analgesia. Hong Kong Med J 2006;12:442-7.
9. Lozano LM, Núñez M, Sastre S, Popescu D. Total knee arthroplasty in the context of severe and morbid obesity in adults. Open Obes J 2012;4:1-10. CrossRef
10. Ringrose NH, Cross MJ. Femoral nerve block in knee joint surgery. Am J Sports Med 1984;12:398-402. CrossRef
11. Heard SO, Edwards WT, Ferrari D, et al. Analgesic effect of intraarticular bupivacaine or morphine after arthroscopic knee surgery: a randomized, prospective, double-blind study. Anesth Analg 1992;74:822-6. CrossRef
12. Woolf CJ, Chong MS. Preemptive analgesia—treating postoperative pain by preventing the establishment of central sensitization. Anesth Analg 1993;77:362-79. CrossRef
13. Mullaji A, Kanna R, Shetty GM, Chavda V, Singh DP. Efficacy of periarticular injection of bupivacaine, fentanyl, and methylprednisolone in total knee arthroplasty: a prospective, randomized trial. J Arthroplasty 2010;25:851-7. CrossRef
14. Mauerhan DR, Campbell M, Miller JS, Mokris JG, Gregory A, Kiebzak GM. Intra-articular morphine and/or bupivacaine in the management of pain after total knee arthroplasty. J Arthroplasty 1997;12:546-52. CrossRef
15. Stein C. The control of pain in peripheral tissue by opioids. N Engl J Med 1995;332:1685-90. CrossRef
16. Stein C. Peripheral analgesic actions of opioids. J Pain Symptom Manage 1991;6:119-24. CrossRef
17. Stein C. Peripheral mechanisms of opioid analgesia. Anesth Analg 1993;76:182-91. CrossRef
18. Leone S, Di Cianni S, Casati A, Fanelli G. Pharmacology, toxicology, and clinical use of new long acting local anesthetics, ropivacaine and levobupivacaine. Acta Biomed 2008;79:92-105.
19. Parvataneni HK, Ranawat AS, Ranawat CS. The use of local periarticular injections in the management of postoperative pain after total hip and knee replacement: a multimodal approach. Instr Course Lect 2007;56:125-31.
20. Parvataneni HK, Shah VP, Howard H, Cole N, Ranawat AS, Ranawat CS. Controlling pain after total hip and knee arthroplasty using a multimodal protocol with local periarticular injections: a prospective randomized study. J Arthoplasty 2007;22(6 Suppl 2):33-8. CrossRef
21. Andersen KV, Nikolajsen L, Haraldsted V, Odgaard A, Soballe K. Local infiltration analgesia for total knee arthroplasty: should ketorolac be added? Br J Anaesth 2013;111:242-8. CrossRef
22. Sharrock NE, Haas SB, Hargett MJ, Urquhart B, Insall JN, Scuderi G. Effects of epidural anesthesia on the incidence of deep-vein thrombosis after total knee arthroplasty. J Bone Joint Surg Am 1991;73:502-6. CrossRef
23. Lamplot JD, Wagner ER, Manning DW. Multimodal pain management in total knee arthroplasty: a prospective randomized controlled trial. J Arthroplasty 2014;29:329-34. CrossRef
24. Ng FY, Ng JK, Chiu KY, Yan CH, Chan CW. Multimodal periarticular injection vs continuous femoral nerve block after total knee arthroplasty: a prospective, crossover, randomized clinical trial. J Arthroplasty 2012;27:1234-8. CrossRef
25. Wu JW, Wong YC. Elective unilateral total knee replacement using continuous femoral nerve blockade versus conventional patient-controlled analgesia: perioperative patient management based on a multidisciplinary pathway. Hong Kong Med J 2014;20:45-51.

Outcomes of a pharmacist-led medication review programme for hospitalised elderly patients

Hong Kong Med J 2018;24:Epub 9 Feb 2018
DOI: 10.12809/hkmj176871
ORIGINAL ARTICLE
Outcomes of a pharmacist-led medication review programme for hospitalised elderly patients
Patrick KC Chiu, FRCP (Glasg), FHKAM (Medicine)1; Angela WK Lee, MPharm, RPharmS (Great Britain)2; Tammy YW See, MClinPharm, RPharmS (Great Britain)2; Felix HW Chan, FRCP (Edin, Glasg, Irel), FHKAM (Medicine)1
1 Geriatric Medical Unit, Grantham Hospital, Wong Chuk Hang, Hong Kong
2 Pharmacy, Grantham Hospital, Wong Chuk Hang, Hong Kong
 
Corresponding author: Dr Patrick KC Chiu (chiukc@ha.org.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Elderly patients are at risk of drug-related problems. This study aimed to determine whether a pharmacist-led medication review programme could reduce inappropriate medications and hospital readmissions among geriatric in-patients in Hong Kong.
 
Methods: This prospective controlled study was conducted in a geriatric unit of a regional hospital in Hong Kong. The study period was from December 2013 to September 2014. Two hundred and twelve patients were allocated to receive either routine care (104) or pharmacist intervention (108) that included medication reconciliation, medication review, and medication counselling. Medication appropriateness was assessed by a pharmacist using the Medication Appropriateness Index. Recommendations made by the pharmacist were communicated to physicians.
 
Results: At hospital admission, 51.9% of intervention and 58.7% of control patients had at least one inappropriate medication (P=0.319). Unintended discrepancy applied in 19.4% of intervention patients of which 90.7% were due to omissions. Following pharmacist recommendations, 60 of 93 medication reviews and 32 of 41 medication reconciliations (68.7%) were accepted by physicians and implemented. After the program and at discharge, the proportion of subjects with inappropriate medications in the intervention group was significantly lower than that in the control group (28.0% vs 56.4%; P<0.001). The unplanned hospital readmission rate 1 month after discharge was significantly lower in the intervention group than that in the control group (13.2% vs 29.1%; P=0.005). Overall, 98.0% of intervention subjects were satisfied with the programme. There were no differences in the length of hospital stay, number of emergency department visits, or mortality rate between the intervention and control groups.
 
Conclusions: A pharmacist-led medication review programme that was supported by geriatricians significantly reduced the number of inappropriate medications and unplanned hospital readmissions among geriatric in-patients.
 
 
New knowledge added by this study
  • This is the first prospective controlled study of the effect of a pharmacist-led medication review programme on medication use and health services utilisation among hospitalised Chinese elderly patients in Hong Kong.
  • The medication review programme led by a clinical pharmacist resulted in a substantial reduction in the use of inappropriate medications among hospitalised elderly patients and all-cause unscheduled readmissions at 1 month after hospital discharge.
Implications for clinical practice or policy
  • A pharmacist-led medication review programme is an important strategy that can enhance the safety and quality of prescription among elderly patients in hospital.
  • It is strongly recommended that these programmes be standardised and implemented in all medical and geriatric wards in Hong Kong.
 
 
Introduction
Elderly patients have multiple co-morbidities and they are consequently prone to multiple medication use. Inappropriate medication use is common among hospitalised older adults. The number of drugs taken is one of the important determinants of risk for receiving an inappropriate medication.1 There is a high prevalence of unnecessary drug use in frail older people. In one hospital study, 44% of patients were prescribed at least one unnecessary drug, with the most common reason being lack of indication.2 The most commonly prescribed unnecessary drug classes were gastrointestinal, central nervous system, and therapeutic nutrients/minerals.2 Appropriate use of drugs is particularly important in the frail older people who are especially at risk of adverse drug reactions.3 It has been shown that implementation of a clinical pharmacist service has a positive effect on medication use and health care service utilisation among hospitalised patients.4 5 A local study in a geriatric hospital demonstrated the effectiveness of a drug rationalisation programme with involvement of a clinical pharmacist in reducing the incidence of polypharmacy and inappropriate medications.6 Interacting with the health care team on patient rounds, interviewing patients, reconciling medications, and providing patient discharge counselling and follow-up all resulted in improved outcomes.7 It is for this reason that patient safety strategies encourage the use of medication reconciliation and clinical pharmacists in health care systems to reduce adverse drug events.8 9 10
 
There is not much information about the effectiveness of a medical review programme among hospitalised elderly patients in Hong Kong. Two recent local reports that examined the effects of a clinical pharmacist–led medication review on hospital readmissions showed conflicting results and did not specifically address elderly patients.11 12 We therefore conducted a prospective controlled study to investigate the effectiveness of a comprehensive pharmacist intervention on medication use and hospital readmission among a group of geriatric in-patients in Hong Kong.
 
Methods
This prospective controlled study was conducted in the geriatric unit of a regional hospital in Hong Kong. The unit has 38 in-patient beds and admits older people aged 65 years or above who are transferred from an acute hospital after initial stabilisation of medical and/or geriatric problems. The unit admits more than 1000 patients per year and provides medical treatment, rehabilitation, and discharge planning services by a multidisciplinary team composed of a geriatrician, residents, nurses, physiotherapists, occupational therapists, and medical social workers. All patients admitted to the unit during December 2013 to September 2014 were included. Patients were excluded if they refused to participate, were terminally ill with a life expectancy of less than 3 months, or if they had already received pharmacist intervention in another hospital prior to this admission. Eligible subjects were assigned to an intervention or control group according to the admission day of the week. Those who were admitted on Monday through Thursday were assigned to the intervention group, and those admitted on Friday through Sunday to the control group. This arrangement was to ensure that pharmacist intervention could be initiated promptly within 48 hours of patient admission. Demographic data, functional status, co-morbidities, and number of drugs on admission were collected at admission.
 
The intervention was conducted by a pharmacist who was present in the unit from Monday to Saturday. The pharmacist provided pharmaceutical care from admission to discharge. Interventions performed by the pharmacist consisted of the following:
 
(1) Medication reconciliation on admission to identify unintended discrepancies between medications prescribed on admission and the usual medications prior to admission—sources to assist medication reconciliation included: electronic patient record; patient’s ward case notes; interview with patient and/or patient carer. The number and type of unidentified discrepancies were recorded.
 
(2) Medication review to check for medication appropriateness on admission and also at discharge—medication appropriateness was assessed by the Medication Appropriateness Index (MAI).13 There are 10 criteria to assess for appropriateness, namely indication, effectiveness, dosage, correct direction, practical direction, drug-drug interaction, drug-disease interaction, duplication, duration, and expense. For a drug item coded as ‘inappropriate’, relative weights for each criterion would apply. A sum of MAI scores could then be calculated to give a score ranging from 0 to 18. The higher the score, the more inappropriate the drug. Recommendations from the pharmacist after the reconciliation and medication review in the intervention group were then communicated to the in-charge doctor via a written note in the medical records. Recommendations were reinforced verbally if deemed appropriate by the pharmacist.
 
(3) Pharmacist counselling on admission and also at discharge was provided to improve patients’ drug knowledge to ensure proper use of drugs and compliance after discharge. A discharge counselling service was provided for all patients who returned home. The counselling included any changes to drug regimen; an explanation of each drug’s indication; any untoward effects that might occur and when to seek medical advice; and drug storage and administration instructions. To ensure patient understanding, written information such as patient information leaflets were given to patients and their carers to remind them of the correct drug regimen. If the patient was illiterate, a simple diagram was drawn on drug labels to demonstrate the time of day and number of tablets to be taken. If necessary, individualised pictorial schedules with drug images and administration instructions could be produced for patients and their carers. The assistance of a family member or external care services such as a community nurse was enlisted if the patient was found to have compliance issues.
 
The control group received routine clinical services. Records of the control group were retrospectively reviewed by the pharmacist after patient discharge to check for medication appropriateness on admission and also at discharge. The primary outcome measure was the appropriateness of prescription as measured by the MAI. Secondary outcomes included the acceptance rate by physicians, number of subjects with unintended discrepancies, patient satisfaction with the programme (for those home-living only), and unplanned hospitalisations 1 and 3 months after discharge.
 
A sample size of 98 patients per group was required to have 85% power to detect an effect size of 0.9 on the MAI. Our sample size was finally set at 210 patients to account for loss of participants due to dropout or death. This sample size was comparable with a study by Spinewine et al14 in which MAI was used as one of the tools to assess appropriateness of prescribing in an acute geriatric care unit and 203 patients were recruited. Our study included 212 patients and was expected to have adequate statistical power to detect differences between groups. Descriptive analyses were performed and included the number and types of unintended discrepancies, MAI score upon admission and at discharge, types of drug-related problems, number of interventions made by pharmacists, and number of recommendations accepted by doctors and implemented. Outcomes for the two groups before and after the programme were compared using the t test and Chi squared test. The Statistical Package for the Social Sciences (Windows version 17.0; SPSS Inc, Chicago [IL], United States) was used and a P value of <0.05 was regarded as statistically significant. The study was approved by the Cluster Research Ethics Committee of the Hospital Authority Hong Kong West Cluster. Written consent was obtained from the patient or their caregiver. The absence of pharmacist intervention in the control group was considered acceptable because a pharmacy service was not a part of routine care at the institution.
 
Results
Figure 1 summarises the patient flow from recruitment to hospital discharge, the components of the medication review programme, and the planned outcome measures. A total of 212 patients were recruited. There were 108 subjects in the intervention group and 104 in the control group (Fig 2). There were no statistical differences in the baseline characteristics of patients (Table 1).
 

Figure 1. Patient flow of the medication review programme from patient recruitment to patient discharge
 

Figure 2. Patient flow diagram
 

Table 1. Baseline demographics and characteristics of the intervention and control groups
 
Appropriateness of prescription
On admission, 51.9% (56/108) of the intervention group and 58.7% (61/104) of the control group had at least one drug classified as inappropriate (P=0.319). Overall, 1996 drug items were reviewed by a pharmacist on admission of which 1020 were from the intervention group and 976 from the control group. Among them, 9.3% and 11.1% of the drugs, respectively, were classified as inappropriate (P=0.282). In the intervention group, 93 recommendations were made by the pharmacist of which 60 (64.5%) were accepted by the physicians and implemented. The mean (± standard deviation) MAI score per patient was 2.19 ± 3.03 in the intervention group and 2.28 ± 3.09 in the control group (P=0.841). The mean MAI score per drug was 0.23 ± 0.30 in the intervention group and 0.25 ± 0.31 in the control group (P=0.628) [Table 2].
 
After the program and at discharge, the proportion of subjects with inappropriate medications in the intervention group was significantly lower than that in the control group (28.0% vs 56.4%; P<0.001). Among the 1999 drug items reviewed by the pharmacist on patient discharge, 3.5% (37 of 1048) of the intervention group and 9.7% (92 of 951) of the control group were classified as inappropriate (P<0.001). The intervention group also had a significantly lower MAI score per patient (0.95 ± 2.02 vs 2.02 ± 2.53; P<0.001) and MAI score per drug (0.09 ± 0.17 vs 0.24 ± 0.30; P<0.001) implying a significant reduction in medication inappropriateness after the pharmacist medication review (Table 2).
 

Table 2. A comparison of the number of subjects with inappropriate medications and the MAI scores between the intervention and control groups on admission and at discharge
 
Types of inappropriateness according to the MAI in the two groups are illustrated in Figure 3. In both the intervention and control groups, the common causes were indication, effectiveness, dosage, practical direction, duration, and expense. After the programme, there was a significant reduction in the number of these drug-related problems in the intervention group.
 

Figure 3. Comparison of the types of inappropriate medication use between the intervention and control groups on admission and at discharge (paired t test)
 
Unintended discrepancy of medications
Among the 108 subjects in the intervention group, 19.4% had at least one unintended discrepancy in medications, involving a total of 43 drug factors. The majority (90.7%) of these factors were omission of drugs, and 4.6% were due to inappropriate dosages. Of all the drug factors involved, 69.8% involved prescribed drugs from hospitals, 25.6% were from a private clinic, and 4.6% were over-the-counter drugs. Overall, 41 recommendations were made, of which 32 (78.0%) were accepted by physicians and implemented.
 
Patient satisfaction
Contact was made with 50 of the 90 non-institutionalised subjects 1 month after discharge to assess satisfaction with the programme. Of those contacted, 98.0% were satisfied with the programme and only one (2.0%) patient expressed a neutral opinion.
 
Impact on health care services utilisation and mortality
There were no statistical differences in the length of hospital stay, in-patient mortality, or mortality at 1 month or 3 months after discharge. There was also no statistical difference in the number of attendances at the accident and emergency department 1 month or 3 months after discharge or in the unplanned hospital readmission rate at 3 months after discharge. The unplanned hospital readmission rate 1 month after discharge, however, was significantly lower in the intervention group than that in the control group (13.2% vs 29.1%; P=0.005) [Table 3].
 

Table 3. Comparison of the impact on health services utilisation and mortality between the intervention and control groups
 
Discussion
To the best of our knowledge, this is the first local prospective controlled study to investigate the effectiveness of a pharmacist-led medication review programme on medication appropriateness and clinical outcomes among geriatric in-patients in Hong Kong. This study has demonstrated superior outcomes that favour a pharmacist-led intervention. There was a substantial reduction in the use of inappropriate medications and all-cause unscheduled readmissions 1 month after hospital discharge. Nonetheless, analysis of length of hospital stay, number of all-cause emergency department visits, and mortality rate favoured neither the intervention nor the usual pharmacist care.
 
This study showed that one in five geriatric in-patients had an unintended medication discrepancy on admission. This figure was slightly higher than that found in a group of 3317 hospitalised medical patients (13%) over 1 year in an acute hospital in Hong Kong by Kwok et al.15 Subjects in our study were all elderly patients, whereas those in Kwok et al’s study were adults of all ages. Elderly subjects tend to have polypharmacy and thus are more vulnerable to unintended medication discrepancy when they move in and out of hospital or are transferred to another health care unit for further care. Unjustifiable medication discrepancies account for more than half of the medication errors that occur during transition of care and up to one third have the potential to cause harm.16 17 This does not bode well for our elderly patients with multiple co-morbidities.
 
Up to 30% of the discrepancies in our study involved medications that had been prescribed by private practitioners or purchased over-the-counter. Unlike medications prescribed from the Hospital Authority, these medications might be overlooked unless the admitting doctor specifically asks for a detailed drug history from the patient. Knowing the medication history and hence resuming these medications are important if new health problems are to be prevented. Pharmacist-led medication reconciliation is therefore a critical process that can enhance patient medication safety by compiling a complete and accurate medication list for patients in hospital.
 
This study revealed that more than half of the subjects (55.2%) received inappropriate medications. The majority of reasons for inappropriateness related to effectiveness, dosage, practical directions, and expense as reflected by the MAI. The inappropriate dosage and the questionable effectiveness might lead to not only failed pharmacological effects, but also potentially an untoward adverse drug reaction, especially in elderly individuals with pre-existing organ dysfunction.18 When a medication is not used according to the practical directions, it may lead to patient non-compliance. Optimising outcomes while reducing costs are the keys for medication management in today’s health care environment.19 Often there are several choices of drugs available to treat a disease or health condition and some are more expensive than others. The involvement of a ward-based pharmacist to review medication can enhance the use of appropriate medications in hospitalised patients and potentially reduce medication costs.
 
Following the medication review (60/93) and medication reconciliation (32/41), there were 92 recommendations that were accepted by the physician. The overall acceptance rate by physicians and the implementation of pharmacist recommendations in our study was 68.7% (92/134). This figure ranged from 39% to 100% in a previous systematic review of 32 studies.4 Our study did not specifically record recommendations accepted by physicians but not implemented. Hence the acceptance rate in our study may be underestimated. Nevertheless, the clinical pharmacist is encouraged to discuss the medication-related problems in person with the physician as well as contacting the patient in order to enhance the implementation rate.4
 
Pharmacy departments within the public hospital system in Hong Kong have strived to implement the aforementioned patient safety strategies in different specialties. Nonetheless, this is not a standard practice simply because of insufficient pharmacist staffing resources. In some hospitals, a pharmacist service is provided in wards, but this does not apply in all cases and is not standardised. Experience of a pharmacist-led medication reconciliation service from an acute teaching hospital in Hong Kong showed promising results over a 1-year trial run with high acceptance and recognition by other health care professionals.16 It is hoped that the clinical role of clinical pharmacists in patient medication management in hospitals can be encouraged. Another local study has demonstrated a positive impact on medication safety in patients with diabetes by pharmacists’ intervention in collaboration with a multidisciplinary team.20 The feasibility of incorporating a pharmacist as part of a multidisciplinary team of health care professionals must be explored in geriatric wards in Hong Kong. With increasing life expectancy, the expanding elderly population will equate to an increase in morbidity and mortality owing to drug-related problems where the need for trained health care professionals to perform medication reviews will be in even greater demand. To enhance safe drug use with limited resources, a systematic approach must be adopted to cover all aspects that affect drug therapy.
 
In terms of the impact on health care services utilisation, a recent systematic review and meta-analysis of the effectiveness of a pharmacist-led medication reconciliation programme revealed a substantial reduction in the rate of all-cause readmissions (19%), all-cause emergency department visits (28%), and adverse drug event–related hospital visits (67%).21 Our study revealed a significant reduction in unplanned hospital admissions (all-cause admission) at 1 month but not at 3 months. This implication might be due to an inadequate sample size to show the difference at 3 months. Alternatively, it might also imply that pharmacist intervention needs to be continued after patient discharge in order to have a sustained effect. This is supported by a study by Schnipper et al22 in which pharmacist intervention after patient discharge was associated with a lower rate of preventable adverse drug events 30 days after hospital discharge.
 
During the pharmacist review, cost-effectiveness of drug use was assessed through MAI. Alternative options such as less-expensive formulations or drugs but of the same quality would be recommended to the doctor in-charge. This was a means of encouraging cost-effective use of drugs in a hospital. Furthermore, the reduction in unscheduled hospital readmissions in the intervention group implies a potential saving in hospital costs. Although a detailed analysis was not performed in this study, a rough estimation is that nearly HK$2 million may be saved annually as a result of lower drug costs and reduced hospital admissions, even after considering the cost of employing a pharmacist. The estimation was based on the following calculations. The estimated drug saving as a result of a switch to a more cost-effective alternative was HK$7500 among the 108 patients in the intervention group. This can be projected to a saving of about HK$69 500 in a unit that admits 1000 patients annually. In the current study, there were 16 fewer readmissions in the intervention group compared with the control group. Assuming a daily cost of an acute hospital bed is HK$4680 and the mean length of hospital stay is 3 days, this equates to a potential saving of about HK$2 080 000 per year in a unit that admits 1000 patients annually. If it is assumed that a pharmacist spends 30 minutes for each patient at an hourly salary of HK$433, the projected cost of an additional pharmacist to run the intervention would be HK$216 500 per year. The net annual saving of this programme to serve 1000 patients in this unit would thus still be close to HK$2 million.
 
This study had several limitations. First, a substantial proportion (35%) of all the admitted patients were not screened by a pharmacist on admission. This was due to a temporary pause in subject recruitment when patients were admitted on public holidays, when the pharmacist was on holiday or when she had to relieve another pharmacist in the hospital. Moreover, a substantial proportion (44%) of eligible subjects were not included owing to no consent or refusal. These factors might have resulted in selection or self-selection bias. Second, subject recruitment was not randomised, but done according to the day of admission. This might be a source of bias. Nevertheless, this would have minimal influence on the outcomes, as the baseline characteristics of the intervention and control groups were comparable. Third, the pharmacist who carried out the review and data extraction was not blinded to the study hypothesis and the group status of the subjects. This could potentially lead to information bias, although this might be partially offset by the fact that the majority of the information or data on the outcome measures were taken with reference to a well-established and validated tool. Fourth, this study was performed in a single unit, so generalisation to other settings is not possible. Fifth, MAI is an implicit tool that is subjective. A single pharmacist as the rater might limit the reliability of the assessment results. Nevertheless, the more explicit tools of STOPP/START criteria23 had also been referred to in addition to the MAI during the review process. Sixth, this study only addressed appropriateness of drug use, whereas underuse of drugs was not investigated. Finally, this study could not conclude a causal relationship between the reduction in inappropriate medications and the reduction in unscheduled hospital readmissions because there were several components in the intervention that included a medication review, medication reconciliation, and discharge counselling. It is difficult to be certain which of these components alone or in combination gave rise to the positive outcome of this study.
 
On the other hand, there were several strengths in this study. This was the first prospective controlled study of the effect of a pharmacist-led medication review programme on medication use and health services utilisation involving over 200 Chinese elderly patients in Hong Kong. Second, a well-validated tool was used to assess medication appropriateness. The use of the MAI tool focused on the patient and the entire medication regimen. Third, there was a comprehensive review of outcomes including quality of prescribing, health services utilisation, mortality, length of hospital stay, and patient satisfaction.
 
Conclusions
This study supported the role of a hospital-based clinical pharmacist to enhance appropriate medication use among elderly Chinese in-patients. A systematic medication review programme in a geriatric unit resulted in a reduced number of drug omissions and fewer inappropriate medications. The service provided by the clinical pharmacist and supported by geriatricians was welcomed by patients and their carers. Together with the potential to reduce hospital readmissions and their associated cost, it is hoped that an in-hospital pharmacist-led medication review programme can be recognised as one of the important strategies to enhance the safety and quality of prescription among elderly patients in hospitals. It is strongly recommended that these programmes be standardised and implemented in all medical and geriatric wards in Hong Kong. Future studies should recruit a larger sample size in a randomised controlled design in other geriatric hospital settings to reiterate our findings. Furthermore, these studies might consider including adverse drug event–related hospital visits as one of the outcome measures.
 
Declaration
All authors have disclosed no conflicts of interest.
 
References
1. Onder G, Landi F, Cesari M, Gambassi G, Carbonin P, Bernabei R; Investigators of the GIFA Study. Inappropriate medication use among hospitalized older adults in Italy: results from the Italian Group of Pharmacoepidemiology in the Elderly. Eur J Clin Pharmacol 2003;59:157-62. CrossRef
2. Hajjar ER, Hanlon JT, Sloane RJ, et al. Unnecessary drug use in frail older people at hospital discharge. J Am Geriatr Soc 2005;53:1518-23. CrossRef
3. Routledge PA, O’Mahony MS, Woodhouse KW. Adverse drug reactions in elderly patients. Br J Clin Pharmacol 2004;57:121-6. CrossRef
4. Graabaek T, Kjeldsen LJ. Medication reviews by clinical pharmacists at hospitals lead to improved patient outcomes: a systematic review. Basic Clin Pharmacol Toxicol 2013;112:359-73. CrossRef
5. Gillespie U, Alassaad A, Henrohn D, et al. A comprehensive pharmacist intervention to reduce morbidity in patients 80 years or older: a randomized controlled trial. Arch Intern Med 2009;169:894-900. CrossRef
6. Chan FH, Pei CK, Chiu KC, Tsang EK. Strategies against polypharmacy and inappropriate medication—are they effective? Australas J Ageing 2001;20:85-9. CrossRef
7. Kaboli PJ, Hoth AB, McClimon BJ, Schnipper JL. Clinical pharmacists and inpatient medical care: a systematic review. Arch Intern Med 2006;166:955-64. CrossRef
8. The ACHS EQuIP5 (5th edition of the ACHS Evaluation and Quality Improvement Program) clinical standard 1.5, criterion 1.5.1.: Medication are managed to ensure safe and effective consumer/patient outcomes. Sydney: The Australian Council on Health Care Standards; 2010.
9. Medication reconciliation. Patient Safety Network. Agency for Healthcare Research and Quality. US Department of Health & Human Services. Available from: http://psnet.ahrq.gov/primer.aspx?primerID=1. Accessed 11 Jun 2017.
10. Gurwitz J, Monane M, Monane S, Avorn J. Polypharmacy. In: Morris JN, Lipsitz LA, Murphy K, Bellville-Taylor P, editors. Quality care in the nursing home. St. Louis, MO: Mosby-Year Book; 1997: 13-25.
11. Wong CY, Lee TO, Lam MP. Pharmacist discharge intervention programme to reduce unplanned hospital use in patients with polypharmacy. Hong Kong Pharm J 2017;24(Suppl 1):S18.
12. Chan HH. To reduce avoidable readmission of patients who are categorized as high risk by 30-day hospital readmission model at medical ward of United Christian Hospital through medication reconciliation and discharge counseling. Hong Kong Pharm J 2017;24(Suppl 1):S21.
13. Hanlon JT, Schmader KE, Samsa GP, et al. A method for assessing drug therapy appropriateness. J Clin Epidemiol 1992;45:1045-51. CrossRef
14. Spinewine A, Swine C, Dhillon S, et al. Effect of a collaborative approach on the quality of prescribing for geriatric inpatients: a randomized, controlled trial. J Am Geriatr Soc 2007;55:658-65. CrossRef
15. Kwok CC, Mak WM, Chui CM. Implementational experience of medical reconciliation in Queen Mary Hospital. Hong Kong Pharm J 2009;16:50-2.
16. Rozich JD, Howard RJ, Justeson JM, Macken PD, Lindsay ME, Resar RK. Standardization as a mechanism to improve safety in health care. Jt Comm J Qual Saf 2004;30:5-14. CrossRef
17. Cornish PL, Knowles SR, Marchesano R, et al. Unintended medication discrepancies at the time of hospital admission. Arch Intern Med 2005;165:424-9. CrossRef
18. Cameron KA. Caregivers’ guide to medications and aging. National Center on Caregiving, Family Caregiver Alliance 2004. Available from: https://www.caregiver.org/caregiver%CA%BCs-guide-medications-and-aging. Accessed 11 Jun 2017.
19. Splawski J, Minger H. Value of the pharmacist in the medication reconciliation process. P T 2016;41:176-8.
20. Chung AY, Anand S, Wong IC, et al. Improving medication safety and diabetes management in Hong Kong: a multidisciplinary approach. Hong Kong Med J 2017;23:158-67. CrossRef
21. Mekonnen AB, McLachlan AJ, Brien JA. Effectiveness of pharmacist-led medication reconciliation programmes on clinical outcomes at hospital transitions: a systematic review and meta-analysis. BMJ Open 2016;6:e010003. CrossRef
22. Schnipper JL, Kirwin JL, Cotugno MC, et al. Role of pharmacist counseling in preventing adverse drug events after hospitalization. Arch Intern Med 2006;166:565-71. CrossRef
23. Lam MP, Cheung BM. The use of STOPP/START criteria as a screening tool for assessing the appropriateness of medications in the elderly population. Expert Rev Clin Pharmacol 2012;5:187-97. CrossRef

Efficacy and tolerability of trastuzumab emtansine in advanced human epidermal growth factor receptor 2–positive breast cancer

Hong Kong Med J 2018 Feb;24(1):56–62 | Epub 12 Jan 2018
DOI: 10.12809/hkmj176808
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
ORIGINAL ARTICLE
Efficacy and tolerability of trastuzumab emtansine in advanced human epidermal growth factor receptor 2–positive breast cancer
Winnie Yeo, FRCP, FHKAM (Medicine)1; MY Luk, FHKCR, FHKAM (Radiology)2; Inda S Soong, FHKCR, FHKAM (Radiology)3; Tony YS Yuen, FHKCR, FHKAM (Radiology)4; TY Ng, FHKCR, FHKAM (Radiology)5; Frankie KF Mo, BSc, PhD6; K Chan, FHKCR, FHKAM (Radiology)3; SY Wong, FHKCR, FHKAM (Radiology)5; Janice Tsang, FHKCP, FHKAM (Medicine)7; Carmen Leung, FHKCR, FHKAM (Radiology)4; Joyce JS Suen, FHKCR, FHKAM (Radiology)8; Roger KC Ngan, FHKCR, FHKAM (Radiology)4
1 Department of Clinical Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong
2 Department of Clinical Oncology, Queen Mary Hospital, Pokfulam, Hong Kong
3 Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong
4 Department of Clinical Oncology, Queen Elizabeth Hospital, Jordan, Hong Kong
5 Department of Clinical Oncology, Tuen Mun Hospital, Tuen Mun, Hong Kong
6 Comprehensive Clinical Trials Unit, Department of Clinical Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong
7 Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
8 Department of Clinical Oncology, Prince of Wales Hospital, Shatin, Hong Kong
 
Corresponding author: Prof Winnie Yeo (wyeo@cuhk.edu.hk)
 
 Full paper in PDF
 
Abstract
Introduction: The management of human epidermal growth factor receptor 2 (HER2)–positive breast cancer has changed dramatically with the introduction and widespread use of HER2-targeted therapies. There is, however, relatively limited real-world information about the effectiveness and safety of trastuzumab emtansine (T-DM1) in Hong Kong Chinese patients. We assessed the efficacy and toxicity profiles among local patients with HER2-positive advanced breast cancer who had received T-DM1 therapy in the second-line setting and beyond.
 
Methods: This retrospective study involved five local centres that provide service for over 80% of the breast cancer population in Hong Kong. The study period was from December 2013 to December 2015. Patients were included if they had recurrent or metastatic histologically confirmed HER2+ breast cancer who had progressed after at least one line of anti-HER2 therapy including trastuzumab. Patients were excluded if they received T-DM1 as first-line treatment for recurrent or metastatic HER2+ breast cancer. Patient charts including biochemical and haematological profiles were reviewed for background information, T-DM1 response, and toxicity data. Adverse events were documented during chemotherapy and 28 days after the last dose of medication.
 
Results: Among 37 patients being included in this study, 28 (75.7%) had two or more lines of anti-HER2 agents and 26 (70.3%) had received two or more lines of palliative chemotherapy. Response assessment revealed that three (8.1%) patients had a complete response, eight (21.6%) a partial response, 11 (29.7%) a stable disease, and 12 (32.4%) a progressive disease; three patients could not be assessed. The median duration of response was 17.3 (95% confidence interval, 8.4-24.8) months. The clinical benefit rate (complete response + partial response + stable disease, ≥12 weeks) was 37.8% (95% confidence interval, 22.2%-53.5%). The median progression-free survival was 6.0 (95% confidence interval, 3.3-9.8) months and the median overall survival had not been reached by the data cut-off date. Grade 3 or 4 toxicities included thrombocytopaenia (13.5%), raised alanine transaminase (8.1%), anaemia (5.4%), and hypokalaemia (2.7%). No patient died as a result of toxicities.
 
Conclusions: In patients with HER2-positive advanced breast cancer who have been heavily pretreated with anti-HER2 agents and cytotoxic chemotherapy, T-DM1 is well tolerated and provided a meaningful progression-free survival of 6 months and an overall survival that has not been reached. Further studies to identify appropriate patient subgroups are warranted.
 
 
New knowledge added by this study
  • This study confirms that the efficacy and toxicity profiles of trastuzumab emtansine (T-DM1) among Chinese patients are similar to the published data that have been based mainly on western populations.
Implications for clinical practice or policy
  • T-DM1 is effective in HER2-positive advanced breast cancer in the second-line setting and beyond. It has tolerable toxicity. Further research is warranted to enable identification of the appropriate patient population to enhance cost-effectiveness.
 
 
Introduction
Breast cancer is the most common female cancer in Hong Kong. The human epidermal growth factor receptor HER2/neu gene is amplified and overexpressed in 15% to 25% of breast cancers.1 The management of human epidermal growth factor receptor 2 (HER2)–positive (HER2+) breast cancer has changed dramatically with the introduction and widespread use of HER2-targeted therapies. The landmark study reported by Slamon et al2 over a decade ago established the combination of trastuzumab with chemotherapy as the standard of care for patients with HER2+ metastatic breast cancer. The later CLEOPATRA trial showed that the combination of pertuzumab with trastuzumab and chemotherapy (specifically, docetaxel) could further improve survival when compared with the standard arm of trastuzumab plus chemotherapy in the first-line setting.3
 
Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that incorporates the HER2-targeted antitumour properties of trastuzumab with the cytotoxic activity of the microtubule inhibitor DM1 (which is a derivative of maytansine). The high potency of the cytotoxic DM1 moiety has been suggested as a key factor in the enhanced activity of this compound.4 5 In the second-line setting, the pivotal EMILIA study compared T-DM1 with lapatinib plus capecitabine among patients with HER2+ breast cancer who had previously been treated with trastuzumab and a taxane; T-DM1 showed remarkable activity with an acceptable toxicity profile.6 There is, however, relatively limited real-world information about the effectiveness and safety of T-DM1 in Hong Kong Chinese patients.
 
In this multicentre retrospective study, we assessed the efficacy and toxicity profiles among local patients with HER2+ advanced breast cancer who had received T-DM1 therapy in the second-line setting and beyond.
 
Methods
This was a retrospective study that involved five local centres that care for over 80% of the local breast cancer population, and included the Pamela Youde Nethersole Eastern Hospital, Prince of Wales Hospital, Queen Mary Hospital, Queen Elizabeth Hospital, and Tuen Mun Hospital between December 2013 and December 2015, the period when the relevant treatment was first started. The institutional ethics committee of each participating centre approved the study.
 
Inclusion criteria included patients who had recurrent or metastatic histologically confirmed HER2+ breast cancer who either had progressed during trastuzumab with chemotherapy in the first-line treatment setting, or had developed progressive disease after at least one line of anti-HER2 agent including trastuzumab. Patients who had received endocrine therapy for recurrent or metastatic disease were included. Exclusion criteria included patients who received T-DM1 as first-line treatment for recurrent or metastatic HER2+ breast cancer.
 
Patient charts were reviewed for background information, T-DM1 response, and toxicity data by medical staff who were not blinded to the study objectives. Biochemical and haematological profiles were extracted from patient charts. Tumour response assessments were recorded according to the Response Evaluation Criteria in Solid Tumors Committee.7 Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 3.0). Adverse events were also documented during chemotherapy and 28 days after the last dose of study medication.
 
Statistical analysis
Outcomes in terms of tumour response, progression-free survival (PFS), and overall survival (OS) were determined. The PFS was assessed from day 1 of treatment cycle 1 to the date when objective disease progression was observed, and OS was calculated from day 1 of treatment cycle 1 to the date of death. Death was regarded as a progression event in those subjects who died before disease progression. Subjects without documented objective progression at the time of the final analysis were censored at the date of their last tumour assessment; data cut-off was on 31 August 2016. Survival curves were constructed using the Kaplan-Meier method.
 
Results
Patient characteristics
Patient characteristics are shown in Table 1. Of a total of 37 patients, 33 (89.2%) had an Eastern Cooperative Oncology Group performance status of 0 or 1.
 

Table 1. Patient characteristics and prior treatments (n=37)
 
Of the 37 patients, tumour biology studies at initial disease presentation showed that 15 (40.5%) patients were oestrogen receptor (ER)–positive, 10 (27.0%) were progesterone receptor (PR)–positive, and 31 (83.8%) had HER2+ breast cancer. Overall, 21 patients had tumour re-biopsy at the time of developing metastatic disease, 10 (47.6%) patients were ER-positive, nine (42.9%) PR-positive, and 21 (100%) had HER2+ (which included six patients who were found to have HER2+ tumours only when anti-HER2 therapy was considered for metastatic disease).
 
At the time of initiating T-DM1 therapy, 21 patients had three or more disease sites involved; the most common sites included lymph nodes (n=27, 73.0%), lungs (n=20, 54.1%), and bones (n=19, 51.4%).
 
Prior treatments
Prior treatments that patients received are listed in Table 1. With regard to adjuvant treatments, 13 (35.1%) patients had prior adjuvant trastuzumab, 21 (56.8%) had adjuvant chemotherapy, eight (21.6%) had adjuvant endocrine therapy, and 19 (51.4%) had adjuvant radiotherapy.
 
With regard to treatment for recurrent/ metastatic disease, nine (24.3%) patients had one line of prior trastuzumab with chemotherapy including three who had trastuzumab in combination with pertuzumab and chemotherapy; 11 (29.7%) had two lines while 17 (45.9%) had three or more lines of anti-HER2 therapy. Overall, 22 (59.5%) patients had received prior lapatinib, and five (13.5%) had received pertuzumab beyond the first-line setting.
 
A total of 26 (70.3%) patients had received two or more lines of palliative chemotherapy, with the majority having received taxanes (n=33, 89.2%), capecitabine (n=23, 62.2%) and vinorelbine (n=17, 45.9%). Nineteen patients had received one or more lines of palliative endocrine therapy, these included eight (21.6%) with tamoxifen, 15 (40.5%) with aromatase inhibitors, and seven (18.9%) with ovarian ablation.
 
Trastuzumab emtansine dose and dose interruptions
The median number of days from last anti-HER2 therapy to the first dose of T-DM1 was 32 days (range, 14-274 days).
 
The median number of cycles was six (range, 1-43). The follow-up data were frozen on 31 August 2016. The median follow-up period was 15.6 months (95% confidence interval [CI], 8.1-20.4 months). Overall, 33 patients were started on the standard dose of 3.6 mg/kg, given once every 3 weeks; 13 patients had dose delay, 10 patients had dose reduction for subsequent cycles, and six patients had both dose delay and dose reductions for subsequent cycles. A total of 326 cycles were administered; 44 (13.5%) cycles were delayed, 11 (3.4%) cycles had further dose reductions in the subsequent cycles, and 51 (15.6%) cycles had both dose delay and dose reductions.
 
At the time of data cut-off, 28 had discontinued T-DM1 treatment: 20 (71.4%) due to progressive disease, four (14.3%) were lost to follow-up, one (3.6%) due to patient withdrawal, and three (10.7%) due to unspecified causes. No patient discontinued treatment due to intolerable toxicities.
 
Response and survival
Among the 37 patients, there were three (8.1%) complete response (CR), eight (21.6%) partial response (PR), 11 (29.7%) stable disease (SD), and 12 (32.4%) progressive disease; three patients could not be assessed (ie they did not have response assessment documented during their treatment). The median duration of response was 17.3 months (interquartile range, 9.4-24.5; 95% confidence interval, 8.4-24.8 months). The clinical benefit rate, defined as CR, PR, or SD of 12 weeks or longer, was 37.8% (95% CI, 22.2%-53.5%).
 
Overall, based on the Kaplan-Meier method, the median PFS was 6.0 (95% CI, 3.3-9.8) months; the 6-month and 12-month PFSs were 51.6% and 23.1%, respectively (Fig a). The median duration of follow-up for PFS was 5.0 (interquartile range, 2.2-10.3) months. The median OS was not reached; the 6-month and 12-month OSs were 82.1% and 74.4%; respectively (Fig b).
 

Figure. Kaplan-Meier estimates of (a) progression-free survival and (b) overall survival
 
Toxicity
Haematological and non-haematological toxicities are listed in Table 2. Grade 3 or 4 toxicities that occurred in one or more patients included thrombocytopenia (n=5, 13.5%), raised alanine transaminase (n=3, 8.1%), anaemia (n=2, 5.4%), and hypokalaemia (n=1, 2.7%). Apart from these, other toxicities that occurred in more than 10% of patients included raised alkaline phosphatase, hyponatraemia, neutropenia, leukopenia, fatigue, raised serum creatinine, and diarrhoea. There was no cardiac toxicity and no patients died as a result of toxicities.
 

Table 2. Haematological and non-haematological toxicities according to the National Cancer Institute Common Toxicity Criteria version 3.0 (n=37)
 
Discussion
During the past decade, the treatment of HER2+ breast cancer has rapidly evolved, and patients with HER2+ metastatic breast cancer have experienced a remarkable improvement in clinical outcomes in terms of OS.8
 
The efficacy of T-DM1 was well demonstrated in the pivotal EMILIA study that compared T-DM1 with lapatinib plus capecitabine among HER2+ breast cancer patients in the second-line setting. The studied patients had previously been treated with trastuzumab and a taxane. For the T-DM1–treated patients, the objective response rate was 44%, the median PFS was 9.6 months, and the median OS was 30.9 months.6
 
In the current multicentre retrospective study among the Chinese patients with breast cancer, over 70% were heavily pretreated with anti-HER2 agents as well as cytotoxic chemotherapy. The efficacy results are consistent with previous findings from the TH3RESA study.9 The latter involved over 600 HER2+ patients with advanced breast cancer who had received two or more anti-HER2–containing regimens, including trastuzumab and lapatinib, and previous taxane therapy. At a median follow-up of 6.5 months, the TH3RESA study reported that among the T-DM1–treated patients, the objective response rate was 31%, the median duration of response was 9.7 months, the median PFS was 6.2 months, and the median OS was not reached.9 Similarly, the safety profile in the current study was consistent with the reported clinical trials, where grade 3 or worse thrombocytopenia was the most commonly reported adverse event (13.5%), followed by raised alanine transaminase (8.1%), anaemia (5.4%), and hypokalaemia (2.7%). Notably there was no grade 3 or worse neutropenia, no febrile neutropenia, and no cardiac toxicity noted in the current study.
 
In heavily pretreated patient populations, two studies, namely the TH3RESA study9 and the EGF104900 study10 (which assessed combination of trastuzumab and lapatinib in the absence of chemotherapy), have shown that even after a median of four prior regimens, the use of anti-HER2 therapy can lead to meaningful clinical benefits. In the TH3RESA study, the PFS benefit with T-DM1 was observed in subgroups including hormone receptor–positive tumours and non-visceral disease, as well as asymptomatic or treated brain metastases. An exploratory analysis conducted in the present study revealed that the median PFSs for patients with hormone receptor–positive disease and hormone receptor–negative disease were 7.5 and 6.0 months, respectively. Owing to small patient numbers, the finding was not significant (P=0.78) but nonetheless lends support to the published data.
 
Among the 37 patients in the current study, five had prior pertuzumab therapy in addition to trastuzumab (including one who also had lapatinib). One of these patients achieved PR and had a total of eight cycles of T-DM1 treatment. The efficacy of T-DM1 among patients previously treated with trastuzumab and pertuzumab has recently been reported in a retrospective study.11 Although the response rate was relatively low at 18%, 30% of the patients had received prolonged T-DM1 therapy, defined as treatment duration of 6 months or longer.
 
It has to be noted that despite the efficacy shown in the second-line and beyond setting among HER2+ patients with advanced breast cancer, the MARIANNE study, which tested three different anti-HER2 regimens in the first-line setting, did not show T-DM1 to be superior to standard treatment.12 In that study, previously untreated patients with HER2+ metastatic breast cancer were randomised to one of the three arms: control (trastuzumab plus taxane), T-DM1 alone, or T-DM1 plus pertuzumab. Although the results revealed that grade 3 or higher adverse events were lower in the T-DM1 arm, efficacy data on PFS were similar in all three arms, at 13.7 months, 14.1 months, and 15.2 months, respectively. In another exploratory analysis in the present study, the PFS of those patients who had undergone only one line of prior anti-HER2 therapy was compared with those who had two or more lines of anti-HER2 therapy revealed corresponding figures of 8.2 and 5.1 months, respectively (P=0.34).
 
In addition, cost-effective analysis has been conducted in a number of countries with regard to the use of T-DM1. For patients with HER2+ metastatic breast cancer, the Canadian analysis demonstrated that utilising T-DM1 could lead to substantial savings for the public health care system when the costs of treatment-related adverse events incurred by other anti-cancer agents were taken into account.13 Nonetheless, analyses based in the United Kingdom and the United States have not supported such findings.14 15 16
 
The identification of an appropriate patient population for the utilisation of T-DM1 may enable better resource allocation. Yet to date, no biomarkers have been identified that can predict better outcome among patients with HER2+ advanced breast cancer treated with T-DM1. Based on the biomarker analyses from EMILIA and TH3RESA studies, T-DM1 was similarly effective in the presence of PI3K wild-type or mutated tumours, and the benefit with T-DM1 was seen irrespective of HER2 mRNA, HER3 mRNA, or PTEN protein level.17 18
 
The current study is limited by its retrospective design, possible information bias during data retrieval/extraction/coding, as well as the small number of patients (especially for subgroup analysis) and inadequate follow-up period for OS. Although the results could not be compared directly with reported prospective trials, patients were representative, and treatment and outcomes reflect routine clinical practice. The T-DM1 therapy provided a meaningful PFS with a favourable toxicity profile among heavily pretreated patients with HER2+ advanced breast cancer. Research is needed to identify biomarkers that will predict sensitivity and resistance to individual anti-HER2 agents, and thereby enable identification of those patients most likely to respond to T-DM1 and appropriate treatment to optimise patient benefit, reduce excessive toxicities, and minimise costs.
 
Conclusions
The T-DM1 therapy has a tolerable toxicity profile among local patients with recurrent or metastatic HER2+ breast cancer. For patients who responded to T-DM1 therapy, there was a durable response. In our study, T-DM1 is associated with a PFS of 6 months and an OS that has not been reached. Further biomarker study is needed to enable appropriate patient selection for this treatment.
 
Acknowledgements
We thank Dr Vicky TC Chan of the Department of Clinical Oncology, Prince of Wales Hospital, and Drs Carol Kwok and Raymond KY Wong of the Department of Oncology, Princess Margaret Hospital, for their support in this study.
 
Declaration
This study has been supported by the Hong Kong Breast Oncology Group. W Yeo has received honoraria for expert opinion from Novartis and Pfizer and has received a research grant from Mundipharma in relation to breast cancer research over the past 12 months. The funder had no role in study selection, quality assessment, data analysis, or writing the manuscript. All other authors have disclosed no conflicts of interest.
 
References
1. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177-82. Crossref
2. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783-92. Crossref
3. Baselga J, Cortés J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012;366:109-19. Crossref
4. Lewis Phillips GD, Li G, Dugger DL, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008;68:9280-90. Crossref
5. Lopus M, Oroudjev E, Wilson L, et al. Maytansine and cellular metabolites of antibody-maytansinoid conjugates strongly suppress microtubule dynamics by binding to microtubules. Mol Cancer Ther 2010;9:2689-99. Crossref
6. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012;367:1783-91. Crossref
7. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 2000;92:205-16. Crossref
8. Lobbezoo DJ, van Kampen RJ, Voogd AC, et al. Prognosis of metastatic breast cancer subtypes: The hormone receptor/HER2-positive subtype is associated with the most favorable outcome. Breast Cancer Res Treat 2013;141:507-14. Crossref
9. Krop IE, Kim SB, González-Martín A, et al. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol 2014;15:689-99. Crossref
10. Blackwell KL, Burstein HJ, Storniolo AM, et al. Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol 2010;28:1124-30. Crossref
11. Dzimitrowicz H, Berger M, Vargo C, et al. T-DM1 activity in metastatic human epidermal growth factor receptor 2–positive breast cancers that received prior therapy with trastuzumab and pertuzumab. J Clin Oncol 2016;34:3511-7. Crossref
12. Perez EA, Barrios C, Eiermann W, et al. Trastuzumab emtansine with or without pertuzumab versus trastuzumab plus taxane for human epidermal growth factor receptor 2-positive, advanced breast cancer: primary results from the phase III MARIANNE study. J Clin Oncol 2017;35:141-8. Crossref
13. Piwko C, Prady C, Yunger S, Pollex E, Moser A. Safety profile and costs of related adverse events of trastuzumab emtansine for the treatment of HER2-positive locally advanced or metastatic breast cancer compared to capecitabine plus lapatinib from the perspective of the Canadian health-care system. Clin Drug Investig 2015;35:487-93. Crossref
14. Diaby V, Adunlin G, Ali AA, et al. Cost-effectiveness analysis of 1st through 3rd line sequential targeted therapy in HER2-positive metastatic breast cancer in the United States. Breast Cancer Res Treat 2016;160:187-96. Crossref
15. Le QA, Bae YH, Kang JH. Cost-effectiveness analysis of trastuzumab emtansine (T-DM1) in human epidermal growth factor receptor 2 (HER2): positive advanced breast cancer. Breast Cancer Res Treat 2016;159:565-73. Crossref
16. Squires H, Stevenson M, Simpson E, Harvey R, Stevens J. Trastuzumab emtansine for treating HER2-positive, unresectable, locally advanced or metastatic breast cancer after treatment with trastuzumab and a taxane: an evidence review group perspective of a NICE single technology appraisal. Pharmacoeconomics 2016;34:673-80. Crossref
17. Baselga J, Lewis Phillips GD, Verma S, et al. Relationship between tumor biomarkers and efficacy in EMILIA, a phase III study of trastuzumab emtansine in HER2-positive metastatic breast cancer. Clin Cancer Res 2016;22:3755-63. Crossref
18. Kim SB, Wildiers H, Krop IE, et al. Relationship between tumor biomarkers and efficacy in TH3RESA, a phase III study of trastuzumab emtansine (T-DM1) vs. treatment of physician’s choice in previously treated HER2-positive advanced breast cancer. Int J Cancer 2016;139:2336-42. Crossref

Injuries and envenomation by exotic pets in Hong Kong

Hong Kong Med J 2018 Feb;24(1):48–55 | Epub 5 Jan 2018
DOI: 10.12809/hkmj176984
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
ORIGINAL ARTICLE
Injuries and envenomation by exotic pets in Hong Kong
Vember CH Ng, FHKCEM, FHKAM (Emergency Medicine)1; Albert CH Lit, FRCSEd, FHKAM (Emergency Medicine)2; OF Wong, FHKAM (Anaesthesiology), FHKAM (Emergency Medicine)2; ML Tse, FHKCEM, FHKAM (Emergency Medicine)1; HT Fung, FRCSEd, FHKAM (Emergency Medicine)3
1 Hong Kong Poison Information Centre, United Christian Hospital, Kwun Tong, Hong Kong
2 Accident and Emergency Department, North Lantau Hospital, Tung Chung, Lantau, Hong Kong
3 Accident and Emergency Department, Tuen Mun Hospital, Tuen Mun, Hong Kong
 
Corresponding author: Dr OF Wong (oifungwong@yahoo.com.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Exotic pets are increasingly popular in Hong Kong and include fish, amphibians, reptiles, and arthropods. Some of these exotic animals are venomous and may cause injuries to and envenomation of their owners. The clinical experience of emergency physicians in the management of injuries and envenomation by these exotic animals is limited. We reviewed the clinical features and outcomes of injuries and envenomation by exotic pets recorded by the Hong Kong Poison Information Centre.
 
Methods: We retrospectively retrieved and reviewed cases of injuries and envenomation by exotic pets recorded by the Hong Kong Poison Information Centre from 1 July 2008 to 31 March 2017.
 
Results: There were 15 reported cases of injuries and envenomation by exotic pets during the study period, including snakebite (n=6), fish sting (n=4), scorpion sting (n=2), lizard bite (n=2), and turtle bite (n=1). There were two cases of major effects from the envenomation, seven cases with moderate effects, and six cases with mild effects. All major effects were related to venomous snakebites. There were no mortalities.
 
Conclusion: All human injuries from exotic pets arose from reptiles, scorpions, and fish. All cases of major envenomation were inflicted by snakes.
 
 
New knowledge added by this study
  • This is the first case series of injuries and envenomation by exotic pets in Hong Kong.
  • Reptiles, scorpions, and fish that are kept as exotic pets can potentially cause injuries to and envenomation of their owners.
  • All cases of major envenomation were inflicted by snakes. Envenomation by a highly venomous exotic snake was also encountered.
Implications for clinical practice or policy
  • A variety of exotic animals, including venomous species, are kept as pets in Hong Kong. Emergency physicians in Hong Kong, however, have limited knowledge about the management of injuries caused by these exotic animals.
  • The Hong Kong Poison Information Centre provides an expert consultation service for the management of injuries and envenomation by such exotic animals.
 
 
Introduction
A variety of exotic animals are kept as ‘pets’ including fish, amphibians, reptiles, and arthropods. The keeping of exotic, and sometimes venomous pets, is becoming increasingly common worldwide. Some of these exotic pets are capable of causing injury to or even life-threatening envenomation of their owners.1
 
Reptiles are the most popular exotic pets worldwide. It has been estimated that 1.5 to 2.0 million households in the United States (US) own one or more pet reptiles. Snakes account for approximately 11% of the imported reptiles in the US, and up to 9% of these are venomous.2 Envenomation by exotic pets, particularly snakes, is an increasing cause for concern in both the US and Europe.3 In a study of exotic snake envenomation in the US, data from the National Poison Data System database revealed 258 cases of exotic snakebites involving at least 61 unique exotic venomous species between 2005 and 2011. Among these, 40% of bites occurred in a private residence.4 Another study of bites and stings by exotic pets in Europe reported 404 cases in four poison centres in Germany and France from 1996 to 2006. Exotic snakebites from rattlesnakes, cobras, mambas, and other venomous snakes were the cause of approximately 40% of envenomations.5 Another survey conducted in the United Kingdom reviewed the data from the National Health Service Health Episode Statistics from 2004 to 2010. A total of 709 hospital admissions associated with injuries from exotic pets were reported and approximately 300 hospital admissions were related to contact with scorpions, venomous snakes, and lizards.6 Nonetheless, no such epidemiological study has been conducted in Hong Kong. According to the thematic household survey report in 2006, 286 300 households in Hong Kong kept pets at home, of which 5% were pets other than dogs, cats, turtles, tortoises, birds, hamsters, and rabbits.7 The number of imported pet reptiles into Hong Kong has increased rapidly in recent years. In 2016, the Agriculture, Fisheries and Conservation Department (AFCD) recorded that almost 1 000 000 pet reptiles were imported into Hong Kong (Table 1).
 

Table 1. Number of imported pet reptiles in Hong Kong from 2012 to 2016 (data from the Agriculture, Fisheries and Conservation Department)
 
The knowledge of local emergency physicians about the management of injuries by these exotic animals is limited. Since 2005, the Hong Kong Poison Information Centre (HKPIC) has provided a 24-hour telephone consultation service (tel: 2635 1111) for health care professionals in Hong Kong, offering poison information and clinical management advice. The objectives of this study were to use HKPIC records to describe the variety of reported exotic species and the clinical features and outcomes of injuries and envenomation caused by exotic pets.
 
Methods
This was a case series based on the database of the HKPIC. It included cases encountered by clinical frontline staff and surveillance data from routine reporting of poisoning cases by all accident and emergency departments (AEDs) under the Hospital Authority (HA). Cases of injuries and envenomation by exotic pets recorded by the HKPIC from 1 July 2008 to 31 March 2017 were retrospectively retrieved. Demographic data of the patients—including the involved species, clinical presentations, and outcomes—were reviewed from the patient electronic Health Record. This study was done in accordance with the principles outlined in the Declaration of Helsinki.
 
The severity of injuries and the effects of envenomation were defined as major (life-threatening or resulting in significant residual disability or disfigurement), moderate (pronounced, prolonged, or systemic signs and symptoms), or mild (minimal and rapidly resolving signs and symptoms).
 
Results
During the study period, 15 cases of injuries and envenomation by exotic pets were reported to the HKPIC. Among the 15 patients, nine consulted the HKPIC for management advice and one was managed by the toxicology team of the AED. Local zoologists were consulted in five cases for species identification and opinion about the venomous nature of the species. All bites and stings were unintentional and occurred in a private household. The mean age of the exotic pet owners was 28.2 (range, 14-59) years and the majority (73%) were male. There were six cases of snakebite, four cases of fish sting, two cases of scorpion sting, two cases of lizard bite, and one case of turtle bite. The severity of injury and envenomation effect are summarised in Table 2.
 

Table 2. Severity of injury and envenomation effect caused by exotic pets
 
All major effects occurred in patients with snakebite. A 16-year-old boy was bitten by a short-tailed mamushi (Gloydius blomhoffii brevicaudus; Fig 1a) on his left middle finger. The short-tailed mamushi is not native to Hong Kong but was being kept as a pet. The patient had a history of snakebite by a bamboo snake (Trimeresurus albolabris) that required antivenom treatment, sustained while attempting to catch the snake in the suburbs. Following the bite by the short-tailed mamushi, the patient developed severe local envenomation over his left hand and required admission to the intensive care unit for close observation of the rapidly progressing local envenomation. No systemic envenomation was observed. A local zoologist was consulted for snake identification. A total of three vials of antivenom for Agkistrodon halys were administered as treatment but ischaemia due to compartment syndrome developed in the left hand. Debridement and fasciotomy were eventually performed. The patient had a residual flexion contraction deformity of his left middle finger 2 months later. He recovered with full movement of the left middle finger 6 months after the injury. Another boy, aged 15 years, was bitten by a bamboo snake on his left thumb. The snake had been caught by the patient in the suburbs and kept as a pet. He developed severe local envenomation and was given three vials of antivenom for Agkistrodon halys and three vials of antivenom for green pit viper. The patient developed tenosynovitis of his left thumb and required emergency surgery for debridement. Another four patients were bitten by ‘nonvenomous’ snakes including a rainbow boa (Epicrates cenchria; Fig 1b), corn snake (Pantherophis guttatus), and eastern hognose snake (Heterodon platirhinos; Fig 1c). All snakes were kept as pets. An 18-year-old girl was accidentally bitten by a rainbow boa on her left hand but had no signs of local or systemic envenomation after the injury. Another patient developed a wound infection after being bitten by a corn snake 2 weeks previously (Fig 1d). She recovered after a course of antibiotic therapy. Two young men were bitten by hognose snakes. One developed local envenomation with progressive swelling over the injured hand (Fig 1e). The local envenomation resolved with conservative management. The HKPIC was consulted in all cases, of which three required consultation with a zoologist.
 

Figure 1. (a) Short-tailed mamushi (Gloydius blomhoffii brevicaudus), (b) rainbow boa (Epicrates cenchria), and (c) eastern hognose snake (Heterodon platirhinos). (d) Local wound infection after being bitten by a corn snake (Pantherophis guttatus), and (e) local envenomation after a bite by an eastern hognose snake
 
Injuries from reptiles other than snakes were also recorded. There were two cases of lizard bite. In one case, a 22-year-old man was bitten by a common iguana (Iguana iguana) on his left wrist. In the other case, a 41-year-old man presented to the AED approximately 2 hours after being bitten on his right hand by a Gila monster (Fig 2a). He developed intense pain and local swelling over the site of injury. The pain lasted for about 12 hours and then gradually improved. His haemodynamic state remained stable and no airway oedema or neurological symptoms were observed during his stay in the emergency medicine ward. He was eventually discharged. A young woman attended the AED because of a turtle bite over her left face with consequent minor physical injury.
 

Figure 2. (a) Gila monster (Heloderma suspectum) and (b) freshwater stingray (Potamotrygon species). (c) Thick-tailed scorpion (Parabuthus transvaalicus) and (d) cave-claw scorpion (Pandinus cavimanus). (e) Wound infection after cave-claw scorpion sting
 
Stings by aquarium fish were the second most common injuries by exotic pets. Four cases were recorded, including one sting by a blue tang fish and three by freshwater stingrays (Fig 2b). All patients developed severe pain over the site of injury that responded to immersion in hot water. One of the patients with a freshwater stingray sting developed a wound infection that required emergency surgery for wound exploration and irrigation.
 
Two male patients were stung by their pet scorpions: a thick-tailed scorpion (Parabuthus transvaalicus; Fig 2c) and a cave-claw scorpion (Pandinus cavimanus; Fig 2d). No systemic envenomation was observed. The patient with the cave-claw scorpion sting developed a local wound infection (Fig 2e) that recovered after a course of antibiotics.
 
The characteristics and management of the 15 cases are summarised in Table 3.
 

Table 3. Detailed description of 15 reported cases of injuries and envenomation by exotic pets
 
Discussion
Injuries by a variety of exotic pets were encountered in this study. More than half of the injuries (9/15) were inflicted by reptiles. Reptiles are becoming increasingly popular to keep as pets in Hong Kong. According to the records of the AFCD over the past 5 years, the top 10 most common reptile species imported to Hong Kong are the European pond turtle (Emys orbicularis), razor-backed musk turtle (Sternotherus carinatus), common snapping turtle (Chelydra serpentina), red-bellied cooter (Pseudemys nelsoni), yellow-spotted Amazon River turtle (Podocnemis unifilis), Hermann’s tortoise (Testudo hermanni), African spurred tortoise (Geochelone sulcata), leopard tortoise (Stigmochelys pardalis), common iguana (Iguana iguana), and ball python (Python regius). Commonly imported pet snakes include the ball python (Python regius), king snake (Lampropeltis getula), corn snake (Pantherophis guttatus), rat snake (Elaphe obsoleta), milk snake (Lampropeltis triangulum), and western hognose snake (Heterodon nasicus). With the exception of the hognose snake, which is a mildly venomous species, they are all nonvenomous. Nonetheless, a much wider variety of species, including venomous reptiles, may be sold on the black market. Bites may occur during the care and handling of these exotic animals.3 Envenomation by exotic venomous species is an uncommon but often serious medical emergency.
 
The keeping of venomous snakes is common in the US.4 Amateur collectors are at risk of bites and envenomation and fatalities have been reported.8 Although envenomation from exotic snakes is rarely encountered in Hong Kong, it poses a great challenge to emergency physicians owing to their lack of experience and limited supplies of antivenom, as illustrated by our case of bite by a short-tailed mamushi. Currently, the HA stocks principally snake antivenom for local venomous species (Table 4). Bites by nonvenomous pet snakes may also result in local envenomation and complications; for instance, although the hognose snake is known as a nonvenomous species, one patient developed local envenomation after being bitten. Another patient developed a wound infection after being bitten by a corn snake.
 

Table 4. Antivenoms currently available in the Hong Kong Poison Information Centre and public hospitals under the Hospital Authority
 
As well as snakes, lizards are popular as pets. Bites by large species such as the common green iguana (Iguana iguana) can result in serious injury.9 Envenomation from lizard bites is rare in Hong Kong. Two lizards are well known to be venomous: the Gila monster (Heloderma suspectum)10 and the Mexican beaded lizard (Heloderma horridum).11 12 Both have venom-secreting glands and bites. The Gila monster is native to the southwestern US extending into Mexico, whereas the beaded lizard is native only to Mexico. The Gila monster is listed in the Convention on International Trade in Endangered Species of Wild Fauna and Flora (CITES) as a protected species.13 Captive-bred Gila monsters are traded in international pet markets. Venom of the Gila monster consists of a variety of proteins including gilatoxin, a kallikrein-like protease that can hydrolyse kininogen and produce bradykinin.11 14 The common envenomation effects are intense pain at the injured site, oedema, paraesthesia, weakness, dizziness, and nausea. Hypotension occurs in severe envenomation.15 The intense pain, oedema, and hypotension are likely due to the bradykinin-mediated effects. Airway oedema has been reported regardless of the site of bite and may occur up to 12 hours after the bite.14 Nevertheless, severe envenomation from the Gila monster occurs in only a minority of patients. In a retrospective study of all cases of Gila monster bite reported to the two Arizona poison control centres from 2000 to 2011, 105 cases of human exposure to Gila monsters were recorded and 70 cases were referred to health care facilities for medical treatment. Eleven cases required admission to hospital and five required care in an intensive care unit. Six patients developed airway oedema and three required emergent airway management including one cricothyrotomy.14 Treatment of Gila monster bites is mainly supportive. Intravenous crystalloid infusion and vasopressors may be required for treatment of hypotension in severe envenomation. Radiographic assessment is needed to look for retained teeth and subcutaneous air due to the chewing-like action during the bites.16 No antivenom to Gila monster is commercially available.17 Observation for at least 12 hours after the bite for delayed-onset airway oedema is recommended.14
 
Among all the reptiles, tortoises and turtles are the most popular in pet markets. All species of tortoises and turtles are nonvenomous although some, such as the alligator snapping turtle (Macrochelys temminckii) and the common snapping turtle (Chelydra serpentina), are aggressive and can grow to a very large size. Bites by these large species can result in severe limb injuries.18
 
Stings by aquarium fish contributed to the second largest group of injuries in our case series. The most commonly encountered aquarium fish was freshwater stingray. Freshwater stingrays (Potamotrygon species) are native to South America. They are regarded as dangerous by the native people of the Amazon and frequent sting during fishing season.19 Freshwater stingrays are not aggressive by nature; stings frequently occur when people step on them or handle them improperly. Different species of freshwater stingrays have different colour patterns on their body. They are popular aquarium fish as they are easy to keep although stings may result in severe envenomation.20 The most common feature of envenomation from freshwater stingrays is intense local pain. Systemic manifestations are rare. Skin necrosis is frequently observed in victims wounded by large freshwater stingrays in the wild.21 In addition, skin necrosis is more commonly observed in victims injured by freshwater stingrays than marine stingrays. A study of tissue extracts from the stingers of freshwater and marine stingrays showed that both tissue extracts had gelatinolytic, caseinolytic, and fibrinogenolytic activity but hyaluronidase activity was detected only in the extracts from freshwater stingrays.22 In our case series, no patient injured by a freshwater stingray developed skin necrosis. The risk of developing skin necrosis is likely related to the venom load. Larger stingrays possess a much larger venom load in their stingers. Small freshwater stingrays are commonly kept in an aquarium and skin necrosis as a result of their sting is uncommon. Hot water immersion is effective in controlling acute pain but does not prevent skin necrosis.21 Wounds caused by freshwater stingray stings such as the Aeromonas species can be complicated by severe secondary infection with virulent bacteria.23 Prophylactic antibiotic is often required.
 
Apart from freshwater stingrays, the stinging catfish (Heteropneustes fossilis) is another commonly reported freshwater aquarium fish that can cause injuries and envenomation. It possesses venom in the sting that is located in front of the soft-rayed portion of the pectoral and dorsal fins. Apart from intense local pain, systemic envenomation including weakness and hypotension can result from a sting.24 25 There was no case reported to the HKPIC of injury by this venomous catfish during the study period. Coral reef fish are also popular pets in Hong Kong. Some coral reef fish, such as the lionfish (Pterois volitans), are venomous.26 Nonetheless, injuries by aquarium coral reef fish were rarely encountered in the AED of Hong Kong.
 
Exotic pet owners also enjoy keeping arthropods such as scorpions and spiders. There are approximately 2000 species of scorpion in the world but only a few (30 to 40) are highly venomous and able to cause severe envenomation in humans.27 Scorpion envenomation is reported throughout the world, mainly in subtropical and tropical regions.28 The majority of scorpion stings cause mild or no envenomation. Species that cause serious medical problems mainly belong to the Buthidae family. The genera of the Buthidae family include Centruroides, Tityus, Leiurus, Androctonus, Buthus and Parabuthus.29 Scorpions have a special venom apparatus, the telson, that produces venom. Scorpion venom comprises numerous toxins including several neurotoxins. Unlike snake venom, scorpion venom generally lacks enzyme activity. The main molecular targets of scorpion neurotoxins are the voltage-gated sodium channels and the voltage-gated potassium channels. Scorpion α-toxin, one of the most medically important neurotoxins in the scorpion venom, acts on the voltage-gated sodium channels. Once the toxin binds to voltage-gated sodium channels, it inhibits inactivation of the channel with consequent prolonged depolarisation and, hence, neuronal excitation. The autonomic centres, both sympathetic and parasympathetic, are stimulated. In most situations of scorpion envenomation, the sympathetic nerves are predominantly affected. Scorpion envenomation is characterised by relatively similar neurotoxic excitation syndromes, irrespective of the species. Parasympathetic effects tend to occur early and then sympathetic effects persist due to the release of catecholamines that are responsible for the severe envenomation. Parasympathetic (cholinergic) effects include hypersalivation, diaphoresis, lacrimation, miosis, diarrhoea, vomiting, bradycardia, hypotension, increased respiratory secretion, and priapism. Sympathetic (adrenergic) effects are manifested as tachycardia, hypertension, mydriasis, hyperthermia, hyperglycaemia, and agitation. Fatal effects of scorpion envenomation are largely due to cardiovascular effects. Various cardiac conduction abnormalities have been reported in patients with scorpion envenomation as well as catecholamine-induced cardiomyopathy, pulmonary oedema, and cardiogenic shock. Other manifestations of systemic envenomation include vomiting, abdominal pain, abnormal oculomotor movements, muscle fasciculation, and spasms of the face and limbs.29 Pancreatitis is also a well-reported complication of envenomation by certain species, such as Leiurus quinquestriatus.30 Nonetheless, severe local envenomation is generally uncommon. Differences in the clinical manifestations of systemic envenomation exist in some species. Delayed localised necrosis has been reported in patients stung by an Iranian scorpion (Hemiscorpius lepturus).31 Patients with envenomation from the thick-tailed scorpion (Parabuthus transvaalicus) in Zimbabwe have been reported to develop predominant symptoms from parasympathetic nerve system stimulation, including profuse sialorrhoea, sweating, and urinary retention, in the absence of sympathetic stimulation.32
 
Scorpion stings and envenomation are uncommon in Hong Kong. Most of the locally reported cases of scorpion sting occurred while patients were handling langsat, a type of tropical fruit from South-East Asia. The Chinese stropped bark scorpion (Lychas mucronatus) hides in the fruit and is subsequently imported into Hong Kong.33 Scorpions are also sold as fish food in aquarium shops in Hong Kong. People use scorpions to feed arowana, which are popular aquarium fish. Importation of endangered scorpion species (CITES-listed species) for commercial purposes is regulated by the Protection of Endangered Species of Animals and Plants Ordinance Cap. 586 in Hong Kong.34 According to the data from the AFCD for importation of CITES-listed scorpions, more than 1000 heads of emperor scorpion (Pandinus imperator) have been imported as pets to Hong Kong each year for the last 4 years. The emperor scorpion is a nonvenomous species and is native to the rainforests and savannas of West Africa. Most scorpions in the pet trade, such as the forest scorpion (Heterometrus species), have no potential for dangerous envenomation. Nonetheless venomous species may also be kept by hobbyists and severe envenomation may occur after stings.
 
Management of scorpion stings includes local wound care and supportive care for systemic envenomation. Expert opinion should be sought from a zoologist for species identification and to determine the venomous nature of the species. Patients with severe systemic envenomation may require antivenom therapy. Specific antivenom (Scorpifav; Sanofi Pasteur, France) for Androctonus australis, Buthus occitanus, and Leiurus quinquestriatus is currently available in the HKPIC.
 
Spiders, such as tarantulas, are popular exotic pets and are common in the pet trade in Hong Kong. Nonetheless, inexperienced owners may be unaware of the potential risk of ocular injury from the barbed urticating hairs on the abdomen of the tarantulas. Eye injuries occur when the barbed hairs come into contact with the eyes, either directly from the tarantula’s ejection or when the owners rub their eyes after handling the spider.35 Embedment of the hairs in the cornea can result in severe complications, including ophthalmia nodosa, iritis, and even permanent visual impairment.36 37
 
Conclusion
The diversity of pets is changing and keeping exotic animals is increasingly popular. Injuries from these exotic pets are expected to increase and envenomation may result from stings or bites from some species. In our case series, reptiles, scorpions, and fish were responsible for human injuries, and all cases of major envenomation were inflicted by snakes. Emergency physicians need to be aware of the appropriate management of injuries and envenomation by these exotic animals. The HKPIC plays an important role in the provision of expert advice about management of these special toxicological cases.
 
Acknowledgement
The authors would like to thank AFCD for providing the data of imported reptiles, scorpions, and spiders.
 
Declaration
All authors have disclosed no conflicts of interest.
 
References
1. Bey TA, Boyer LV, Walter FG, McNally J, Desai H. Exotic snakebite: envenomation by an African puff adder (Bitis arietans). J Emerg Med 1997;15:827-31. Crossref
2. McNally J, Boesen K, Boyer L. Toxicologic information resources for reptile envenomations. Vet Clin North Am Exot Anim Pract 2008;11:389-401. Crossref
3. de Haro L, Pommier P. Envenomation: a real risk of keeping exotic house pets. Vet Hum Toxicol 2003;45:214-6.
4. Warrick BJ, Boyer LV, Seifert SA. Non-native (exotic) snake envenomations in the U.S., 2005-2011. Toxins (Basel) 2014;6:2899-911. Crossref
5. Schaper A, Desel H, Ebbecke M, et al. Bites and stings by exotic pets in Europe: an 11 year analysis of 404 cases from Northeastern Germany and Southeastern France. Clin Toxicol (Phila) 2009;47:39-43. Crossref
6. Warwick C, Steedman C. Injuries, envenomations and stings from exotic pets. J R Soc Med 2012;105:296-9. Crossref
7. Thematic household survey report 2006. Census and Statistics Department, HKSAR Government. Available from: http://www.statistics.gov.hk/pub/ B11302262006XXXXB0100.pdf. Accessed 1 Jun 2017.
8. Marsh N, DeRoos F, Touger M. Gaboon viper (Bitis gabonica) envenomation resulting from captive specimens—a review of five cases. Clin Toxicol (Phila) 2007;45:60-4. Crossref
9. Heffelfinger RN, Loftus P, Cabrera C, Pribitkin EA. Lizard bites of the head and neck. J Emerg Med 2012;43:627-9. Crossref
10. Hooker KR, Caravati EM, Hartsell SC. Gila monster envenomation. Ann Emerg Med 1994;24:731-5. Crossref
11. Cantrell FL. Envenomation by the Mexican beaded lizard: a case report. J Toxicol Clin Toxicol 2003;41:241-4.Crossref
12. Ariano-Sánchez D. Envenomation by a wild Guatemalan beaded lizard Heloderma horridum charlesbogerti. Clin Toxicol (Phila) 2008;46:897-9. Crossref
13. Wijnstekers W. The Convention on International Trade in Endangered Species of Wild Fauna and Flora (CITES)—35 years of global efforts to ensure that international trade in wild animals and plants is legal and sustainable. Forensic Sci Rev 2011;23:1-8.
14. French R, Brooks D, Ruha AM, et al. Gila monster (Heloderma suspectum) envenomation: descriptive analysis of calls to United States Poison Centers with focus on Arizona cases. Clin Toxicol (Phila) 2015;53:60-70. Crossref
15. Piacentine J, Curry SC, Ryan PJ. Life-threatening anaphylaxis following gila monster bite. Ann Emerg Med 1986;15:959-61. Crossref
16. French RN, Ash J, Brooks DE. Gila monster bite. Clin Toxicol (Phila) 2012;50:151-2. Crossref
17. Miller MF. Gila monster envenomation. Ann Emerg Med 1995;25:720. Crossref
18. Johnson RD, Nielsen CL. Traumatic amputation of finger from an alligator snapping turtle bite. Wilderness Environ Med 2016;27:277-81. Crossref
19. Junior VH, Cardoso JL, Neto DG. Injuries by marine and freshwater stingrays: history, clinical aspects of the envenomations and current status of a neglected problem in Brazil. J Venom Anim Toxins Incl Trop Dis 2013;19:16. Crossref
20. Brisset IB, Schaper A, Pommier P, de Haro L. Envenomation by Amazonian freshwater stingray Potamotrygon motoro: 2 cases reported in Europe. Toxicon 2006;47:32-4. Crossref
21. Haddad V Jr, Neto DG, de Paula Neto JB, de Luna Marques FP, Barbaro KC. Freshwater stingrays: study of epidemiologic, clinic and therapeutic aspects based on 84 envenomings in humans and some enzymatic activities of the venom. Toxicon 2004;43:287-94. Crossref
22. Barbaro KC, Lira MS, Malta MB, et al. Comparative study on extracts from the tissue covering the stingers of freshwater (Potamotrygon falkneri) and marine (Dasyatis guttata) stingrays. Toxicon 2007;50:676-87. Crossref
23. Polack FP, Coluccio M, Ruttimann R, Gaivironsky RA, Polack NR. Infected stingray injury. Pediatr Infect Dis J 1998;17:349,360.
24. Dorooshi G. Catfish stings: a report of two cases. J Res Med Sci 2012;17:578-81.
25. Satora L, Kuciel M, Gawlikowski T. Catfish stings and the venom apparatus of the African catfish Clarias gariepinus (Burchell, 1822), and stinging catfish Heteropneustes fossilis (Bloch, 1794). Ann Agric Environ Med 2008;15:163-6.
26. Chan HY, Chan YC, Tse ML, Lau FL. Venomous fish sting cases reported to Hong Kong Poison Information Centre: a three-year retrospective study on epidemiology and management. Hong Kong J Emerg Med 2010;17:40-4. Crossref
27. Santos MS, Silva CG, Neto BS, et al. Clinical and epidemiological aspects of scorpionism in the world: a systematic review. Wilderness Environ Med 2016;27:504-18. Crossref
28. Chippaux JP, Goyffon M. Epidemiology of scorpionism: a global appraisal. Acta Trop 2008;107:71-9. Crossref
29. Kounis NG, Soufras GD. Scorpion envenomation. N Engl J Med 2014;371:1558-9.
30. Sofer S, Shalev H, Weizman Z, Shahak E, Gueron M. Acute pancreatitis in children following envenomation by the yellow scorpion Leiurus quinquestriatus. Toxicon 1991;29:125-8.Crossref
31. Pipelzadeh MH, Jalali A, Taraz M, Pourabbas R, Zaremirakabadi A. An epidemiological and a clinical study on scorpionism by the Iranian scorpion Hemiscorpius lepturus. Toxicon 2007;50:984-92.Crossref
32. Bergman NJ. Scorpion sting in Zimbabwe. S Afr Med J 1997;87:163-7.
33. Centre for Food Safety, Hong Kong SAR Government. Scorpion stings and langsat. Food Safety Focus (16th Issue, Nov 2007). Available from: http://www.cfs.gov.hk/english/ multimedia/multimedia_pub/multimedia_pub_fsf_16_04. html. Accessed 1 Jun 2017.
34. Agriculture, Fisheries and Conservation Department, Hong Kong SAR Government. Protection of Endangered Species of Animals and Plants Ordinance. Available from: http://www.afcd.gov.hk/english/conservation/con_end/ con_end_reg/con_end_reg_ord/con_end_reg_ord.html. Accessed 1 Jun 2017.
35. Waggoner TL, Nishimoto JH, Eng J. Eye injury from tarantula. J Am Optom Assoc 1997;68:188-90.
36. Belyea DA, Tuman DC, Ward TP, Babonis TR. The red eye revisited: ophthalmia nodosa due to tarantula hairs. South Med J 1998;91:565-7. Crossref
37. Blaikie AJ, Ellis J, Sanders R, MacEwen CJ. Eye disease associated with handling pet tarantulas: three case reports. BMJ 1997;314:1524-5. Crossref

Optimising the utility of pleural fluid adenosine deaminase for the diagnosis of adult tuberculous pleural effusion in Hong Kong

Hong Kong Med J 2018 Feb;24(1):38–47 | Epub 22 Dec 2017
DOI: 10.12809/hkmj176238
© Hong Kong Academy of Medicine. CC BY-NC-ND 4.0
 
ORIGINAL ARTICLE
Optimising the utility of pleural fluid adenosine deaminase for the diagnosis of adult tuberculous pleural effusion in Hong Kong
KC Chang, MSc, FHKAM (Medicine)1; MC Chan, MB, BS, MRCP (UK)2; WM Leung, MB, ChB, FHKAM (Medicine)1; FY Kong, MB, BS, FHKAM (Medicine)3; Chloe M Mak, MD, FHKAM (Pathology)4; Sammy PL Chen, MRes(Med), FHKAM (Pathology)4; WC Yu, FRCP, FHKAM (Medicine)2
1 Tuberculosis and Chest Service, Department of Health, Hong Kong
2 Department of Medicine and Geriatrics, Princess Margaret Hospital, Laichikok, Hong Kong
3 Department of Medicine and Geriatrics, Yan Chai Hospital, Tsuen Wan, Hong Kong
4 Chemical Pathology Laboratory, Department of Pathology, Princess Margaret Hospital, Laichikok, Hong Kong
 
Corresponding author: Dr KC Chang (kc_chang@dh.gov.hk)
 
 Full paper in PDF
 
Abstract
Introduction: Pleural fluid adenosine deaminase level can be applied to rapidly detect tuberculous pleural effusion. We aimed to establish a local diagnostic cut-off value for pleural fluid adenosine deaminase to identify patients with tuberculous pleural effusion, and optimise its utility.
 
Methods: We retrospectively reviewed the medical records of consecutive adults with pleural fluid adenosine deaminase level measured by the Diazyme commercial kit (Diazyme Laboratories, San Diego [CA], United States) during 1 January to 31 December 2011 in a cluster of public hospitals in Hong Kong. We considered its level alongside early (within 2 weeks) findings in pleural fluid and pleural biopsy, with and without applying Light’s criteria in multiple scenarios. For each scenario, we used the receiver operating characteristic curve to identify a diagnostic cut-off value for pleural fluid adenosine deaminase, and estimated its positive and negative predictive values.
 
Results: A total of 860 medical records were reviewed. Pleural effusion was caused by congestive heart failure, chronic renal failure, or hypoalbuminaemia caused by liver or kidney diseases in 246 (28.6%) patients, malignancy in 198 (23.0%), non-tuberculous infection in 168 (19.5%), tuberculous pleural effusion in 157 (18.3%), and miscellaneous causes in 91 (10.6%). All those with tuberculous pleural effusion had a pleural fluid adenosine deaminase level of ≤100 U/L. When analysis was restricted to 689 patients with pleural fluid adenosine deaminase level of ≤100 U/L and early negative findings for malignancy and non-tuberculous infection in pleural fluid, the positive predictive value was significantly increased and the negative predictive value non-significantly reduced. Using this approach, neither additionally restricting analysis to exudates by Light’s criteria nor adding closed pleural biopsy would further enhance predictive values. As such, the diagnostic cut-off value for pleural fluid adenosine deaminase is 26.5 U/L, with a sensitivity of 87.3%, specificity of 93.2%, positive predictive value of 79.2%, negative predictive value of 96.1%, and accuracy of 91.9%. Sex, age, and co-morbidity did not significantly affect prediction of tuberculous pleural effusion using the cut-off value.
 
Conclusion: We have established a diagnostic cut-off level for pleural fluid adenosine deaminase in the diagnosis of tuberculous pleural effusion by restricting analysis to a level of ≤100 U/L, and considering early pleural fluid findings for malignancy and non-tuberculous infection, but not Light’s criteria.
 
 
New knowledge added by this study
  • There are limitations to the use of pleural fluid adenosine deaminase (pfADA) level as a surrogate marker for tuberculous pleural effusion (TBPE); thus, it must be interpreted alongside other findings that help exclude non-tuberculous diseases, thereby increasing the pre-test probability of TBPE and the positive predictive value (PPV).
  • We demonstrated that TBPE was unlikely when pfADA level was >100 U/L.
  • Restricting analysis to patients with pfADA level of ≤100 U/L and early (within 2 weeks) negative findings for malignancy and non-tuberculous infection in pleural fluid significantly increased PPV and non-significantly reduced the negative predictive value (NPV). Using this approach, neither additionally restricting analysis to exudates by Light’s criteria nor adding closed pleural biopsy would further enhance predictive values of pfADA for TBPE. As such, the local pfADA diagnostic cut-off value is set at 26.5 U/L, with a sensitivity of 87.3%, specificity of 93.2%, PPV of 79.2%, NPV of 96.1%, and accuracy of 91.9%.
Implications for clinical practice or policy
  • Among patients with pfADA level of ≤100 U/L, when pfADA level is ≥26.5 U/L with early negative findings in pleural fluid for malignancy and non-tuberculous infection, it is probably appropriate to manage the patient as a case of TBPE, without additionally performing pleural biopsy (also a surrogate marker for TBPE), but remain vigilant for a 20.8% (1 minus PPV) chance of mistaking non-tuberculous diseases as TBPE.
  • When pfADA level is <26.5 U/L with early negative findings in pleural fluid for malignancy and non-tuberculous infection, tuberculosis is highly unlikely, but caution should be exercised because of a 3.9% (1 minus NPV) chance of mistaking TBPE for another disease.
  • Other investigations are always indicated when the clinical progress is incompatible with the working diagnosis.
 
 
Introduction
Adenosine deaminase (ADA) is an enzyme involved in purine metabolism, with its primary function in the development and maintenance of the immune system. There are at least two ADA isoforms: ADA1 and ADA2. Whereas ADA1 is found in most body cells (especially lymphocytes and macrophages), ADA2 is predominantly found in the human plasma and serum, and co-exists with ADA1 in macrophages. Absence of ADA1 causes severe combined immunodeficiency. Serum ADA2 level is increased in collagen vascular disease,1 2 and most cancers.
 
Many studies have suggested that pleural fluid adenosine deaminase (pfADA) is useful in the diagnosis of tuberculous pleural effusion (TBPE).3 4 5 6 7 8 9 10 11 12 13 The merits of using pfADA include its low cost, short turnaround time, and high sensitivity and specificity.3 12 Notwithstanding possibly better sensitivity and specificity for detecting TBPE by combining ADA1 or ADA2 in pleural fluid (PF) with other PF biomarkers such as tumour necrosis factor–alpha, interleukin 27, interferon-gamma and dipeptidyl peptidase IV,14 15 16 17 it may not be cost-effective to combine pfADA with other PF biomarkers.18 Although ADA2 is predominantly increased in TBPE, and ADA1 is more commonly associated with pleural effusion due to pyogenic bacteria,19 determination of ADA1 and ADA2 may not provide a diagnostic advantage over the use of total pfADA.20
 
A standardised and automated method (Diazyme commercial kit; Diazyme laboratories, San Diego [CA], United States) has been developed to determine pfADA activity. The test performance of pfADA has largely been evaluated by including all cases with pleural effusion, and estimating its sensitivity and specificity with reference to an optimal cut-off value. Some studies fine-tuned the test performance by restricting the analysis to subjects with lymphocytic exudates9 13 or to young adults.21 In Hong Kong, pfADA has been measured centrally by the Chemical Pathology Laboratory at the Princess Margaret Hospital using the Diazyme commercial kit. In the absence of a diagnostic cut-off value established from local data, pfADA level of ≥30 U/L has been used territory-wide in Hong Kong for detecting TBPE. This is with reference to a retrospective Thai study of 59 (33.1%) patients with TBPE among 178 patients with predominantly exudative lymphocytic pleural effusion.22 It suggested a sensitivity of 82% and specificity of 91% for pfADA level of ≥30 U/L, as measured by the Diazyme commercial kit.22 Corresponding estimates of positive predictive value (PPV) and negative predictive value (NPV) were 81.4% and 90.8%, respectively.22
 
Although pfADA rapidly detects TBPE, it is often assessed alongside other tests that include sputum bacteriology for acid-fast bacilli (AFB) and other pathogens, sputum cytology, PF bacteriology for AFB and other pathogens, PF biochemistry, PF cytology, and pleural biopsy. Restricting analysis to patients with exudative pleural effusion may help optimise the utility of pfADA for detecting TBPE. Excluding patients with an early diagnosis of non-tuberculous disease, notably malignancy and non-tuberculous infection, may also help improve the utility of pfADA for TBPE.5 9 23 24 25 26
 
Diagnostic test accuracy depends on sensitivity and specificity, which are relatively stable, and pre-test probability that can be enhanced by selecting appropriate patients. In this study, we aimed to optimise its utility by increasing the pre-test probability, and establish a local diagnostic pfADA cut-off value for adult TBPE. Additionally, we evaluated whether the prediction of TBPE using the pfADA cut-off value was affected by sex, age, or co-morbidity.
 
Methods
We retrospectively searched a centralised computerised database for consecutive PF specimens tested for ADA from 1 January 2011 to 31 December 2011 and assembled a cohort of patients with exudative pleural effusion. These patients were all managed at a cluster of public hospitals that served a large population in western Kowloon of Hong Kong. At least 90% of patients with pleural effusion had PF tested for ADA. We considered pfADA alongside early (within 2 weeks) findings in PF and pleural biopsy, with and without applying Light’s criteria27 in multiple scenarios. For each scenario, we used the receiver operating characteristic (ROC) curve and the Youden Index (the point of maximal summation of sensitivity and specificity estimates) to identify an optimal pfADA diagnostic cut-off value for TBPE, and estimated the corresponding PPV and NPV. The Youden Index maximises the difference between the true-positive rate (sensitivity) and the false-positive rate (1 minus specificity), thereby maximising the correct classification rate. When the Youden Index comprised more than 1 point, we also considered the point at minimal distance between the ROC curve and the coordinate with 100% specificity and 100% sensitivity.
 
The following data were collected by review of medical records that had been created and maintained by clinicians who were unaware of the study hypothesis: sex, age (at the time of initial diagnosis), smoking history, drinking history, co-morbidity (chronic obstructive pulmonary disease, diabetes mellitus, chronic renal failure), use of immunosuppressive treatment for at least 1 month in the past year, nature of PF (exudate vs transudate by Light’s criteria), sputum AFB smear and culture, PF AFB smear and culture, PF bacterial and fungal stain, PF culture of other bacteria or fungus, pleural biopsy findings, other significant findings related to initial or definitive diagnosis, the early diagnosis (within 2 weeks after checking pfADA), and the definitive diagnosis (by 1 year after checking pfADA).
 
This study was conducted in accordance with the amended Declaration of Helsinki, and approved by the Kowloon West Cluster Research Ethics Committee (IRB approval number: KW/EX-13-139(69-17)) and the Department of Health Ethics Committee (IRB approval number: L/M 400/2013).
 
Definitions
The exudative versus transudative nature of PF was established by reference to Light’s criteria that classify PF as exudative in the presence of any one of the following: ratio of protein in PF to serum >0.5, ratio of lactate dehydrogenase (LDH) in PF to serum >0.6, and PF LDH level of >200 IU/L.27
 
A definitive diagnosis of TBPE was made when Mycobacterium tuberculosis complex was isolated in culture of PF or parietal pleura, or any one of the following in the absence of an alternative diagnosis by 1 year after pfADA checking: (i) granulomatous inflammation of parietal pleura, (ii) culture-proven pulmonary tuberculosis (TB) with pleural effusion and compatible response to TB treatment, (iii) a clinical diagnosis of TBPE with compatible response to TB treatment, or (iv) AFB and/or positive findings from nucleic acid amplification tests in PF or parietal pleura. An early diagnosis of TBPE was made when pleural biopsy showed granulomatous inflammation in the absence of an alternative cause, or rarely, the presence of AFB or positive findings from nucleic acid amplification tests in PF or parietal pleura.
 
Parapneumonic effusion refers to any pleural effusion secondary to pneumonia or lung abscess.28 The PF is often exudative with a predominance of neutrophils.28 It can be ‘simple’ (with sterile exudate) or ‘complicated’ (with progression to a fibrinopurulent state), characterised by pH <7.2, glucose level of <2.2 mmol/L, and LDH level of >1000 IU/L.29 Empyema thoracis is a complicated parapneumonic effusion with frank pus.28 A definitive diagnosis of simple non-tuberculous parapneumonic effusion was made if the PF was exudative and sterile with LDH level of ≤1000 IU/L and if there was a compatible clinical response to empirical antibiotic treatment, in the absence of an alternative diagnosis by 1 year after the first attempt of diagnostic thoracentesis. Without an identifiable non-tuberculous pathogen, we considered it impossible to confidently make an early diagnosis of simple non-tuberculous parapneumonic effusion. A definitive diagnosis of complicated non-tuberculous parapneumonic effusion, or empyema thoracis in the presence of frank pus or compatible radiological signs on chest computed tomographic scan was made if the PF was exudative with a non-tuberculous pathogen (demonstrated by positive stain/culture) in PF or parietal pleura, or LDH level of >1000 IU/L, and compatible clinical response to empirical antibiotic treatment and/or drainage, in the absence of an alternative diagnosis by 1 year after the first attempt of diagnostic thoracentesis. An early diagnosis of complicated non-tuberculous parapneumonic effusion, or empyema thoracis in the presence of frank pus or compatible radiological signs, was made when a non-tuberculous pathogen could be identified in PF or parietal pleura.
 
Malignant pleural effusion refers to the presence of malignant cells in PF and/or parietal pleura.30 A definitive diagnosis of malignant pleural effusion was made if malignant cells were found in PF and/or parietal pleura, or clinical/radiological findings were compatible with malignant pleural effusion in the absence of an alternative diagnosis by 1 year after the first attempt of diagnostic thoracentesis. An early diagnosis of malignant pleural effusion was made when malignant cells could be demonstrated in PF or parietal pleura.
 
Statistical analysis
Chi squared test (for categorical data), Fisher’s exact test (for categorical data), McNemar’s test (for paired data), Student’s t test (for continuous variables normally distributed), and Mann-Whitney U test (for continuous variables not normally distributed) were used as appropriate to evaluate the association between TBPE and the pfADA cut-off as well as demographic factors and co-morbidity. Factors with a P value of <0.25 by univariate analysis were forced into a logistic regression model after considering multicollinearity.
 
Laboratory methods
Throughout the study period, ADA activity was measured by the same automated method, the Diazyme commercial kit in the Beckman Coulter UniCel DxC 800 Synchron Clinical System. The automated Diazyme method has been validated.31
 
Results
Search from the computerised database of ADA assay from 1 January to 31 December 2011 identified a total of 903 independent PF specimens from 903 patients. We evaluated 860 patients with pleural effusion and pfADA after excluding 42 cases that were peritoneal rather than PF and one case with no medical record. Table 1 shows their definitive diagnoses and the corresponding pfADA value. Pleural effusion was caused by congestive heart failure, chronic renal failure, hypoalbuminaemia, or nephrotic syndrome/nephropathy with membranous glomerulonephritis in 246 (28.6%) cases, malignancy in 198 (23.0%), non-tuberculous infection (simple non-tuberculous parapneumonic effusion, complicated non-tuberculous parapneumonic effusion other than empyema, and non-tuberculous empyema thoracis) in 168 (19.5%), TBPE in 157 (18.3%), and miscellaneous or unknown causes in 91 (10.6%). By Light’s criteria, 626 (72.8%) cases were classified as exudative, 222 (25.8%) as transudative, and 12 (1.4%) as indeterminate (lack of data).
 

Table 1. Definitive diagnosis of pleural effusion among a cohort of 860 patients with pfADA level measured
 
Among the 198 patients with malignant pleural effusion, an early diagnosis could be established by detecting malignant cells in PF in 136 (68.7%), including 21 also detected by pleural biopsy (20 closed and 1 open), and seven (3.5%) by pleural biopsy alone (5 closed and 2 open). Malignant pleural effusion was caused by lung cancer in 152 (76.8%) patients, lymphoid or haematological malignancy in 12 (6.1%), unknown primary in nine (4.5%), gastric cancer in five (2.5%), ovarian cancer in four (2.0%), breast cancer in four (2.0%), liver cancer in two (1.0%), pancreatic cancer in two (1.0%), and one (0.5%) each by cancer in the nasopharynx, tongue, oesophagus, unspecified gastrointestinal tract, kidney, urinary bladder, prostate, and nerve.
 
Among the 168 patients with non-tuberculous infection, infection was bacteriologically confirmed by PF culture in 21 (12.5%) cases with non-tuberculous empyema thoracis (including 19 with early diagnosis), and 10 (6.0%) with complicated non-tuberculous parapneumonic effusion (including 9 with early diagnosis).
 
Among the 157 patients with TBPE, the diagnosis was (1) bacteriologically confirmed by PF culture in 62 (39.5%) including four also confirmed by pleural tissue culture and 26 also suggested by pleural biopsy; (2) bacteriologically confirmed by pleural tissue culture in 12 (7.6%) including four also confirmed by PF culture and nine also suggested by pleural biopsy; (3) histologically suggested by pleural biopsy in 74 (69 closed and 5 open; 47.1%) including 26 also confirmed by PF culture and nine also confirmed by pleural tissue culture; (4) clinically suggested by pulmonary TB in 44 (28.0%) including 26 solely by clinical correlation with radiological progress; and (5) clinically suggested by culture-proven TB ascites in one (0.6%). Sputum AFB smear was positive in nine (5.7%) patients, with M tuberculosis complex isolated in the sputum culture of 51 (32.5%). An early diagnosis could be established in 65 (41.4%), using pleural biopsy in 64 and polymerase chain reaction in PF in one. The majority (n=152) of patients with TBPE were Chinese.
 
Among 90 patients with TBPE and closed pleural biopsy performed, TBPE was detected by pleural biopsy in 69 (76.7%) and pfADA cut-off level in 85 (94.4%). The difference was statistically significant (P<0.005 by McNemar’s test).
 
Table 2 shows the distribution of pfADA levels stratified by the nature of PF and tuberculous versus non-tuberculous pleural effusion. The prevalence (pre-test probability) of TBPE was significantly higher among exudative (24.3%) than transudative (2.3%) cases. All cases with TBPE had pfADA level of ≤86 U/L. With pfADA level of >86 U/L, all cases (n=18) with exudative non-tuberculous pleural effusion had pfADA level of >100 U/L of whom 13 patients had non-tuberculous empyema thoracis, two had lymphoma, one had plasmacytoma, one had liver abscess, and one had an uncertain diagnosis.
 

Table 2. Distribution of pfADA levels stratified by the nature of pleural fluid and tuberculous versus non-tuberculous pleural effusion
 
Figure 1 shows how we proceeded to increase the pre-test probability of TBPE by first excluding transudates, and then stepwise excluding non-tuberculous patients to further increase the pre-test probability. For each scenario, the pfADA cut-off value was tabulated alongside estimates of sensitivity, specificity, PPV, and NPV. Restricting analysis to 461 patients with exudative pleural effusion, pfADA level of ≤100 U/L, and early negative findings for non-tuberculous infection and malignancy in PF significantly increased PPV from 66.3% to 79.5% and non-significantly reduced NPV from 97.1% to 94.5%. Further excluding seven patients with an early diagnosis of malignancy by pleural biopsy resulted in no change to PPV and a non-significant decrease in NPV. Figure 2 shows an alternative approach that disregards Light’s criteria. Restricting analysis to 689 patients with pfADA level of ≤100 U/L and early negative findings for non-tuberculous infection and malignancy in PF also significantly increased PPV from 66.3% to 79.2% and non-significantly reduced NPV from 97.1% to 96.1%. Further excluding seven patients with an early diagnosis of malignancy by pleural biopsy resulted in no change to PPV and a non-significant decrease in NPV from 96.12% to 96.07%. With no significant difference in PPV (P=0.938) or NPV (P=0.279) between the two approaches, the utility of pfADA may be optimised by applying a diagnostic cut-off among patients with pfADA level of ≤100 U/L and early negative findings for malignancy and non-tuberculous infection in PF, without considering Light’s criteria or pleural biopsy. As such, pfADA level of ≥26.5 U/L, ascertained from the ROC curve using the Youden Index, detected TBPE with a sensitivity of 87.3%, specificity of 93.2%, PPV of 79.2%, NPV of 96.1%, and accuracy of 91.9% (Fig 3).
 

Figure 1. Optimising predictive values of pfADA by first considering Light’s criteria
 

Figure 2. Optimising predictive values of pfADA without considering Light’s criteria
 

Figure 3. Receiver operating characteristic curve based on data of 689 patients with pleural fluid adenosine deaminase level of ≤100 U/L and early (within 2 weeks) negative findings for malignancy and non-tuberculous infection in pleural fluid
 
Table 3 shows different causes of pleural effusion above and below the diagnostic pfADA cut-off value of 26.5 U/L among the 689 patients with pfADA level of ≤100 U/L, and early negative findings for malignancy and non-tuberculous infection in PF. It is noteworthy that in the seven (4.0%) patients with pfADA level of ≥26.5 U/L and 64 (12.4%) patients with pfADA level of <26.5 U/L, the diagnosis was uncertain. Among 157 patients with TBPE, 137 (87.3%, sensitivity) tested positive (pfADA ≥26.5 U/L), with false-negative results in 20 (12.7%, the false-negative rate or 1 minus sensitivity). Among 532 patients with non-tuberculous pleural effusion, 496 (93.2%, specificity) tested negative (pfADA <26.5 U/L), with false-positive results in 36 (6.8%, false-positive rate or 1 minus specificity). Among 173 patients who tested positive, 137 (79.2%, PPV) were true-positive, and 36 (20.8%, 1 minus PPV) were false-positive with non-tuberculous diseases mistaken for TBPE: 10 with complicated non-tuberculous parapneumonic effusion, 10 with non-tuberculous empyema thoracis, seven with uncertain diagnosis, four with malignant pleural effusion, four with simple non-TB parapneumonic effusion, and one with chronic renal failure. Among 516 patients tested negative, 496 (96.1%, NPV) were true-negative, and 20 (3.9%, 1 minus NPV) were false-negative with TBPE mistaken for non-tuberculous pleural effusion. Among 689 test results, 633 (91.9%) were accurate and comprised 137 true-positive and 496 true-negative results.
 

Table 3. Different causes of pleural effusion above and below the diagnostic pfADA cut-off value among 689 patients with pfADA level of ≤100 U/L, and early (within 2 weeks) negative findings for malignancy and non-tuberculous infection in pleural fluid
 
A logistic regression model that considered sex, age, and co-morbidity alongside the pfADA diagnostic cut-off value identified pfADA level of ≥26.5 U/L as the only significant predictive variable of TBPE (Table 4).
 

Table 4. Univariate and multiple logistic regression analyses of tuberculous versus non-tuberculous exudative pleural effusion among 461 patients with exudative pleural effusion, pfADA level of ≤100 U/L, and early (within 2 weeks) negative findings for malignancy and non-tuberculous infection in pleural fluid
 
Discussion
As a limited surrogate marker for TBPE, pfADA level must be interpreted alongside other clinical, radiological, and laboratory findings that help exclude non-tuberculous diseases, thereby increasing the pre-test probability of TBPE and the PPV. Using local data as measured by the Diazyme commercial kit, we demonstrated that TBPE was unlikely when pfADA level was >100 U/L. Restricting analysis to patients with pfADA level of ≤100 U/L and early (within 2 weeks) negative findings for malignancy and non-tuberculous infection in PF significantly increased the PPV and non-significantly reduced the NPV. Using this approach, neither additionally restricting analysis to exudates by Light’s criteria, nor adding closed pleural biopsy, would further enhance the predictive value of pfADA for TBPE. This might be explained by the fact that pfADA level of >13 U/L excluded all non-tuberculous transudative cases (Table 2), and that pfADA was significantly more sensitive than closed pleural biopsy for TBPE. A recent study, which demonstrated a need to suspect empyema or lymphoma when the pfADA level was extremely high,32 corroborated our findings regarding the low likelihood of TBPE when pfADA level was >100 U/L. Furthermore, we demonstrated that the prediction of TBPE using the pfADA diagnostic cut-off value was not affected by sex, age, or co-morbidity. Another study that developed a predictive model for TBPE also failed to show any significant association between TBPE and either age or sex.33
 
Among patients with pfADA level of ≤100 U/L, when pfADA level is ≥26.5 U/L with early negative findings in PF for malignancy and non-tuberculous infection, it is probably appropriate to manage the patient as a case of TBPE, without additionally performing pleural biopsy (also a surrogate marker for TBPE). Nonetheless, it is important to remain vigilant due to a 20.8% (1 minus PPV) chance of mistaking non-tuberculous diseases for TBPE and prescribing unnecessary TB treatment. When pfADA level is <26.5 U/L with early negative findings in PF for malignancy and non-tuberculous infection, TB is highly unlikely. Again caution should be exercised in the presence of a 3.9% (1 minus NPV) chance of mistaking TBPE for other diseases. Tuberculosis is potentially fatal although effective treatment can reduce morbidity and mortality. Yet standard TB treatment is not without harmful side-effects that include hepatotoxicity. This occurs in 1% to 3% of patients on average and becomes more prevalent among the elderly people and those with underlying liver disease.34 35 Additionally, treating non-tuberculous disease as TB may also delay the diagnosis of other diseases including malignancy. It is important to balance the benefits of TB treatment against the risks when using a pfADA cut-off value to diagnose TBPE. In general, if the test suggests TBPE, and the risk of morbidity or mortality from untreated TB is substantial, it is prudent to promptly start TB treatment, and closely monitor treatment progress, with further investigations for other diseases conducted concurrently or as soon as treatment response is considered suboptimal. Other investigations are always indicated when the clinical progress is incompatible with the working diagnosis.
 
A major drawback of this study was its retrospective nature and related selection and misclassification bias. Selection bias may be modest as public hospitals provide approximately 90% of hospital care in Hong Kong, and we included every consecutive and non-duplicated PF sample from all patients managed during the study period in a large public hospital cluster in which at least 90% patients with pleural effusion had PF tested for ADA. Misclassification bias may occur. Efforts made by clinicians to confirm TB disease may be selectively affected by knowledge about the association between pfADA and TB. Non-tuberculous infection could have been misclassified as TB, thereby overestimating PPV or underestimating NPV. On the other hand, TBPE could also have been misclassified as non-TB, thereby underestimating PPV or overestimating NPV. Another possible source of misclassification bias was uncertain diagnosis (Table 3), which was considered as non-tuberculous during analysis. Of note, TBPE labelled as uncertain diagnosis could have caused an underestimation of PPV or overestimation of NPV. Nonetheless, the lack of a definitive diagnosis by 1 year might suggest a low likelihood of TBPE, thereby reducing the impact of this misclassification bias.
 
We have established a pfADA diagnostic cut-off value for TBPE by restricting analysis to patients with pfADA level of ≤100 U/L, and considering early PF findings for malignancy and non-tuberculous infection, but not Light’s criteria.
 
Declaration
All authors have disclosed no conflicts of interest.
 
References
1. Sari RA, Taysi S, Yilmaz O, Bakan N. Correlation of serum levels of adenosine deaminase activity and its isoenzymes with disease activity in rheumatoid arthritis. Clin Exp Rheumatol 2003;21:87-90.
2. Stancíková M, Lukác J, Istok R, Cristalli G, Rovensky J. Serum adenosine deaminase activity and its isoenzyme pattern in patients with systemic lupus erythematosus. Clin Exp Rheumatol 1998;16:583-6.
3. Gupta DK, Suri JC, Goel A. Efficacy of adenosine deaminase in the diagnosis of pleural effusions. Indian J Chest Dis Allied Sci 1990;32:205-8.
4. Bañales JL, Pineda PR, Fitzgerald JM, Rubio H, Selman M, Salazar-Lezama M. Adenosine deaminase in the diagnosis of tuberculous pleural effusions. A report of 218 patients and review of the literature. Chest 1991;99:355-7. Crossref
5. Valdés L, Alvarez D, San José E, et al. Value of adenosine deaminase in the diagnosis of tuberculous pleural effusions in young patients in a region of high prevalence of tuberculosis. Thorax 1995;50:600-3. Crossref
6. Burgess LJ, Maritz FJ, Le Roux I, Taljaard JJ. Combined use of pleural adenosine deaminase with lymphocyte/neutrophil ratio. Increased specificity for the diagnosis of tuberculous pleuritis. Chest 1996;109:414-9. Crossref
7. Riantawan P, Chaowalit P, Wongsangiem M, Rojanaraweewong P. Diagnostic value of pleural fluid adenosine deaminase in tuberculous pleuritis with reference to HIV coinfection and a Bayesian analysis. Chest 1999;116:97-103. Crossref
8. Reechaipichitkul W, Kawamatawong T, Teerajetgul Y, Patjanasoontorn B. Diagnostic role of pleural fluid adenosine deaminase in tuberculous pleural effusion. Southeast Asian J Trop Med Public Health 2001;32:383-9.
9. Lee YC, Rogers JT, Rodriguez RM, Miller KD, Light RW. Adenosine deaminase levels in nontuberculous lymphocytic pleural effusions. Chest 2001;120:356-61. Crossref
10. Jiménez Castro D, Díaz Nuevo G, Pérez-Rodríguez E, Light RW. Diagnostic value of adenosine deaminase in nontuberculous lymphocytic pleural effusions. Eur Respir J 2003;21:220-4. Crossref
11. Chen ML, Yu WC, Lam CW, Au KM, Kong FY, Chan AY. Diagnostic value of pleural fluid adenosine deaminase activity in tuberculous pleurisy. Clin Chim Acta 2004;341:101-7. Crossref
12. Baba K, Hoosen AA, Langeland N, Dyrhol-Riise AM. Adenosine deaminase activity is a sensitive marker for the diagnosis of tuberculous pleuritis in patients with very low CD4 counts. PLoS ONE 2008;3:e2788. Crossef
13. Garcia-Zamalloa A, Taboada-Gomez J. Diagnostic accuracy of adenosine deaminase and lymphocyte proportion in pleural fluid for tuberculous pleurisy in different prevalence scenarios. PLoS ONE 2012;7:e38729. Crossref
14. Küpeli E, Karnak D, Elgün S, Argüder E, Kayacan O. Concurrent measurement of adenosine deaminase and dipeptidyl peptidase IV activity in the diagnosis of tuberculous pleural effusion. Diagn Microbiol Infect Dis 2009;65:365-71. Crossref
15. Wu YB, Ye ZJ, Qin SM, Wu C, Chen YQ, Shi HZ. Combined detections of interleukin 27, interferon-γ, and adenosine deaminase in pleural effusion for diagnosis of tuberculous pleurisy. Chin Med J (Engl) 2013;126:3215-21.
16. Keng LT, Shu CC, Chen JY, et al. Evaluating pleural ADA, ADA2, IFN-γ and IGRA for diagnosing tuberculous pleurisy. J Infect 2013;67:294-302. Crossref
17. Li M, Wang H, Wang X, Huang J, Wang J, Xi X. Diagnostic accuracy of tumor necrosis factor-alpha, interferon-gamma, interleukin-10 and adenosine deaminase 2 in differential diagnosis between tuberculous pleural effusion and malignant pleural effusion. J Cardiothorac Surg 2014;9:118. Crossref
18. Sharma SK, Banga A. Pleural fluid interferon-gamma and adenosine deaminase levels in tuberculosis pleural effusion: a cost-effectiveness analysis. J Clin Lab Anal 2005;19:40-6. Crossref
19. Ungerer JP, Oosthuizen HM, Retief JH, Bissbort SH. Significance of adenosine deaminase activity and its isoenzymes in tuberculous effusions. Chest 1994;106:33-7. Crossref
20. Andreasyan NA, Hairapetian HL, Sargisova YG, Mardanyan SS, Badalyan LT, Khanoyan AS. Activity of adenosine deaminase and its isoforms in pleural fluid in tuberculous pleuritis. Med Sci Monit 2002;8:CR708-12.
21. Yildiz PB, Yazar EE, Gorgun D, Secik F, Cakir G. Predictive role of adenosine deaminase for differential diagnosis of tuberculosis and malignant pleural effusion in Turkey. Asian Pac J Cancer Prev 2011;12:419-23.
22. Kawamatawong T, Panompong K, Kiatboonsri S, Khupulsup K. The appropriate cutoff level of pleural fluid adenosine deaminase activity by diazyme commercial kit for diagnosis pleural tuberculosis in Ramathibodi hospital. Chest 2008;134(Suppl 2):p55001. Crossref
23. Valdés L, San José E, Alvarez D, Valle JM. Adenosine deaminase (ADA) isoenzyme analysis in pleural effusions: diagnostic role, and relevance to the origin of increased ADA in tuberculous pleurisy. Eur Respir J 1996;9:747-51. Crossref
24. Strankinga WF, Nauta JJ, Straub JP, Stam J. Adenosine deaminase activity in tuberculous pleural effusions: a diagnostic test. Tubercle 1987;68:137-40. Crossref
25. Porcel JM, Esquerda A, Bielsa S. Diagnostic performance of adenosine deaminase activity in pleural fluid: a singlecenter experience with over 2100 consecutive patients. Eur J Intern Med 2010;21:419-23. Crossref
26. Ogata Y, Aoe K, Hiraki A, et al. Is adenosine deaminase in pleural fluid a useful marker for differentiating tuberculosis from lung cancer or mesothelioma in Japan, a country with intermediate incidence of tuberculosis? Acta Med Okayama 2011;65:259-63.
27. Light RW, Macgregor MI, Luchsinger PC, Ball WC Jr. Pleural effusions: the diagnostic separation of transudates and exudates. Ann Intern Med 1972;77:507-13. Crossref
28. Light RW. Parapneumonic effusions and empyema. Proc Am Thorac Soc 2006;3:75-80. Crossref
29. Davies HE, Davies RJ, Davies CW, BTS Pleural Disease Guideline Group. Management of pleural infection in adults: British Thoracic Society Pleural Disease Guideline 2010. Thorax 2010;65 Suppl 2:ii41-53. Crossref
30. Antunes G, Neville E, Duffy J, Ali N, Pleural Diseases Group, Standards of Care Committee, British Thoracic Society. BTS guidelines for the management of malignant pleural effusions. Thorax 2003;58 Suppl 2:ii29-38. Crossref
31. Feres MC, Martino MC, Maldijian S, Batista F, Gabriel Júnior A, Tufik S. Laboratorial validation of an automated assay for the determination of adenosine deaminase activity in pleural fluid and cerebrospinal fluid [in English, Portuguese]. J Bras Pneumol 2008;34:1033-9. Crossref
32. Wu YH, Zhao GW, Wang XF, Wang MS. Pleural effusion adenosine deaminase is not accurate in diagnosis of pediatric tuberculous pleural effusion: a retrospective study. Eur Rev Med Pharmacol Sci 2015;19:1706-10.
33. Neves DD, Dias RM, Cunha AJ. Predictive model for the diagnosis of tuberculous pleural effusion. Braz J Infect Dis 2007;11:83-8. Crossref
34. Chang KC, Leung CC, Yew WW, Tam CM. Standard anti-tuberculosis treatment and hepatotoxicity: do dosing schedules matter? Eur Respir J 2007;29:347-51. Crossref
35. Chang KC, Leung CC, Yew WW, Lau TY, Tam CM. Hepatotoxicity of pyrazinamide: cohort and case-control analyses. Am J Respir Crit Care Med 2008;177:1391-6. Crossref

Pages