155
HEALTH TECHNOLOGY ASSESSMENT OF NEW MODELS IN HEALTHCARE RITIKA KAPOOR (B.Tech, M.Tech) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL OF INTEGRATIVE SCIENCE & ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2018 Supervisor: Professor Teo Yik Ying Examiners: Associate Professor Mikael Hartman Associate Professor Lee Guat Lay Caroline Professor Shuhua Xu, CAS-MPG Partner, Institute of Computational Biology (PICB)

HEALTH TECHNOLOGY ASSESSMENT OF NEW …scholarbank.nus.sg/bitstream/10635/138695/1/Thesis_Ritika(HTA).pdf · Chapter 2 Health Technology Assessment: ... 101 4.5 Discussion ... and

  • Upload
    lynhi

  • View
    215

  • Download
    0

Embed Size (px)

Citation preview

HEALTH TECHNOLOGY ASSESSMENT

OF NEW MODELS IN HEALTHCARE

RITIKA KAPOOR (B.Tech, M.Tech)

A THESIS SUBMITTED

FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

NUS GRADUATE SCHOOL OF INTEGRATIVE

SCIENCE & ENGINEERING

NATIONAL UNIVERSITY OF SINGAPORE

2018

Supervisor:

Professor Teo Yik Ying

Examiners:

Associate Professor Mikael Hartman Associate Professor Lee Guat Lay Caroline Professor Shuhua Xu, CAS-MPG Partner, Institute of Computational Biology (PICB)

1

Declaration

I hereby declare that this thesis is my original work and it has been

written by me in its entirety. I have duly acknowledged all the sources

of information that have been used in this thesis.

This thesis has also not been submitted for any degree in any

university previously.

Ritika Kapoor

8th August 2017

2

Acknowledgements

This Ph.D. has been a wonderful journey of learnings and experiences

for me and today I would like to express my sincere and heartfelt thanks

to the many people who made it more knowledgeable, joyful and

enriching.

First and foremost, I would like to thank NGS, The Graduate School of

Integrative Science and Engineering, NUS, for considering me worthy

for such a wonderful opportunity to embark on this learning adventure.

Thank you for being so considerate, flexible and supporting us to attain

an excellent training and an unmatched international exposure.

I have been very fortunate to find my supervisor in Prof. Teo Yik Ying,

who has always been a strong pillar of supervision, support and

encouragement. His encouragement to contribute as an independent

thinker, advice to skill development, freedom to pursue whatever I am

interested in, trust he had on me and his efforts to foresee and resolve

any problems that might arise for me have been substantial to be able

to achieve this. Thank you YY for always being there irrespective of your

busy schedule, your never ending encouragement and trust which

helped me bring the very best of me in work and in life.

I am eternally thankful to my TAC members – Assoc. Prof. Joanne

Yoong, Assoc. Prof. Cynthia Sung and Prof. Christopher Chen Li Hsian

for their excellent supervision and suggestions throughout these years.

3

Special thanks to Assoc. Prof. Joanne Yoong, A/P Wee Hwee Lin for

working with me and guiding me on projects, career and life.

It was wonderful to work with Prof. Yeoh Khay Guan, Prof. Jimmy So

Bok Yan, Dr. Zhu Feng, Dr. Lihan Zhou, Dr. Calvin J. Koh, Dr Chester

Drum and Dr. Tai E Shyong and I want to express my sincere gratitude

for their suggestions and helping me develop clinical perspectives to

bring more meaning to my work. Also, I want to thank my lab mates and

friends – Xuanyao, Woei-Yuh, Dineli, Tra My, Wang Xu, Anthony, Eryu,

Rick, Wesley, Shiv, Jayantika, Dinesh, Aditi, Mahalakshmi for being

helpful, loving and caring and adding so many colours to these years.

Finally, I want to extend a special thanks to my family – my husband, Mr.

Abhishek Modi, my parents - Mr. R.S Kapoor, Mrs. Asha Kapoor and my

sisters Neha and Nidhi Kapoor for encouraging me to follow my dreams

and for the unconditional love, care and support. A special thanks to my

husband for being the one to encourage me to take this course, for his

unshakable faith in me and for celebrating with me all along.

You all have contributed to make it a joyful journey which I will cherish

for a lifetime. Thank you so much.

4

Table of Contents

Summary .............................................................................................. 8

Publications ........................................................................................ 11

List of Tables ...................................................................................... 12

List of Figures ..................................................................................... 14

List of Abbreviations ........................................................................... 16

Chapter 1 Role of Pharmacogenomics in public health and clinical healthcare: A SWOT Analysis ............................................................ 18

1.1 Strengths of Pharmacogenomics in Clinical Medicine .............. 20

1.1.1 Faster achievement of optimal drug dosages ................. 20

1.1.2 Minimising toxicity and adverse side effects ................... 21

1.1.3 Identifying efficacious drugs ............................................ 22

1.1.4 Reduce overall costs to the healthcare system ............... 23

1.2 Weaknesses of Pharmacogenomics in Clinical Medicine ......... 24

1.2.1 Costs of Pharmacogenomic tests to the individuals ........ 24

1.2.2 Speed of testing to the physicians .................................. 25

1.2.3 Imperfect understanding of genetically driven drug response variability ...................................................................... 25

1.3 Opportunities of pharmacogenomics in clinical medicine ......... 26

1.3.1 Technological innovations and falling prices of genetic tests ........................................................................................ 26

1.3.2 Next generation sequencing to encourage individualized pharmacogenomics ..................................................................... 27

1.3.3 Innovation in healthcare that impacts drug development and utility 28

1.3.4 Public pharmacogenomics network on clinical implementation ............................................................................ 28

1.3.5 Uptake of pharmacogenomics in outpatient settings ....... 29

1.4 Threats of pharmacogenomics in clinical medicine ................... 30

5

1.4.1 Availability of non-genetics alternatives to physicians ..... 30

1.4.2 Misaligned incentives for clinical discovery versus clinical implementation activities ............................................................. 31

1.4.3 Lack of healthcare infrastructure ..................................... 32

1.4.4 Mistrust over management and use of genetic information 32

1.4.5 Lack of buy-in by healthcare financiers ........................... 33

1.4.6 Higher institutional risk and threats of lawsuits ............... 33

1.4.7 Widening inequality in healthcare accessibility ............... 34

1.5 Discussion ................................................................................ 35

Chapter 2 Health Technology Assessment: A logic based approach to regulatory decision making ............................................................. 37

2.1 Perspective of Analysis ............................................................. 41

2.2 Allele frequencies, Disease Severity and Prevalence ............... 43

2.3 Patients Adherence .................................................................. 45

2.4 Willingness-To-Pay (Cost-effectiveness threshold) .................. 46

2.5 Healthcare system .................................................................... 47

2.6 Discussion ................................................................................ 48

Chapter 3 Cost Effectiveness Analysis of HLA-B*5701 Genotyping in Newly Diagnosed Persons Living with HIV/AIDS in Singapore, by Ethnicity 49

3.1 Introduction ............................................................................... 49

3.2 Methods .................................................................................... 51

3.2.1 Model Structure............................................................... 51

3.2.2 Model Inputs ................................................................... 54

3.3 Results ...................................................................................... 61

3.3.1 Sensitivity Analysis ......................................................... 62

3.4 Discussion ................................................................................ 68

6

Chapter 4 Cost-Effectiveness Analysis for including a Serum-microRNA Biomarker Panel as a Screen before Endoscopy, for Detection of Gastric Cancer among High Risk Patients in Singapore 72

4.1 Introduction ............................................................................... 72

4.1.1 Gastric Cancer statistics in Singapore ............................ 75

4.2 Methods .................................................................................... 80

4.2.1 Target Population: ........................................................... 80

4.2.2 Strategies Compared ...................................................... 83

4.2.3 Methodology ................................................................... 83

4.2.4 Model Inputs ................................................................... 86

4.2.5 Sensitivity Analysis ......................................................... 91

4.3 Analysis for the Korean population screening program ............ 93

4.4 Results ...................................................................................... 94

4.4.1 Hospital-clinic setting ...................................................... 94

4.4.2 Population screening for Chinese males (50-69 years) .. 97

4.4.3 Population screening in Korea ...................................... 101

4.5 Discussion .............................................................................. 105

Chapter 5 Cost-Effectiveness and Value of Information Analysis of SLCO1B1 Genotyping (rs4149056) before Simvastatin Prescription for Secondary Prevention of AMI in Singapore ...................................... 111

5.1 Introduction ............................................................................. 111

5.2 Methods .................................................................................. 114

5.2.1 Model Structure............................................................. 114

Side effects on Cholesterol Lowering Drugs .............................. 115

Clinical Management of Side effects ......................................... 116

5.2.2 Sources of Information .................................................. 117

5.2.3 Sensitivity analysis ........................................................ 126

5.3 Results .................................................................................... 127

7

5.3.2 Value of Information analysis ........................................ 129

5.4 Discussion .............................................................................. 133

Chapter 6 Conclusion .................................................................... 138

Bibliography ...................................................................................... 141

8

Summary

As defined by World Health Organization (WHO), Health Technology

Assessment is a multidisciplinary process to evaluate the social,

economic, organizational, and ethical issues of a health intervention or

healthcare technology. Comparing a novel technology to the current

standard of care or other alternatives, it provides evidence for healthcare

authorities to make informed policy decisions.

With increasing interest in stratified medicine and multiple drug-gene

associations with proven clinical utility, there is a growing focus on

including pharmacogenomic tests as standard-of-practice in the clinics.

As clinical trials and programs are already evaluating the clinical

implications, the overall economic burden is an equally important

parameter to determine its applicability in routine healthcare. Many

features specific to the population demographics, ethnicity and the

healthcare setup impact the need, willingness and affordability of any

new model of care, like – ethnic makeup, disease prevalence, healthcare

facilities/services availability and costs, awareness among the

physicians and patients etc. With one of the world’s best healthcare

infrastructure in Singapore, clinical translation of pharmacogenomics to

offer effective treatment and/or reduce adverse reactions is of

paramount importance to provide efficient and patient friendly healthcare

experience, and yet manage the increasing expenditure. However, with

a genetically heterogeneous demographic makeup and difference in

9

healthcare services costs, the recommendations of Food & Drug

Administration (FDA) and European Medicines Agency (EMA) may not

always be of the best interest to Singapore. Thus, there is a need of

assessment of these emerging propositions specific to Singapore’s

healthcare and population setup to be able to devise optimal and

sustainable policies.

This PhD project was undertaken to conduct technology assessment,

particularly exploring the cost-effectiveness of implementing new

pharmacogenomic models of healthcare in Singapore. Besides its

academic pursuits, it also aimed to provide scientific evidence and

identify potentially valuable areas of future research to aid in translation

of these technologies in real practice. Working with the NUS Saw Swee

Hock School of Public Health, and public hospitals in Singapore, three

important studies were identified as part of this project, which are at

different stages of discovery and implementation – a technology with

FDA recommendations for clinical implementation (HLA-B*5701

genotyping before Abacavir), a technology with proven clinical utility but

no recommendations by FDA (SLCO1B1 genotyping before Simvastatin

prescription) and a novel technology which is at the stage of clinical

discovery and validation (miRNA serum test for gastric cancer

diagnosis). For the benefit of reader and reviewers, the thesis has been

organized into six chapters as follows:

10

Chapter 1 explores the role of pharmacogenetics in healthcare and

identifies the strengths, weakness, opportunities and threats involved

with its translation in the clinics.

Chapter 2 elaborates on the importance of Health Technology

Assessment for pharmacogenomic implementations and discusses on

the parameters which influence the evaluations and imparts it a

personalized flavor.

Chapter 3 evaluates the cost-effectiveness of implementation of HLA-

B*5701 genetic testing before Abacavir prescription for HIV patients in

Singapore. This genetic test has currently been recommended by FDA.

Chapter 4 evaluates the application of a novel miRNA biomarker serum

blood test to help in detection of gastric cancer in Singapore. The study

further evaluates the benefits of an improved adherence and its future

application for population cancer screening programs in Singapore.

Chapter 5 is focused on cost-effectiveness analysis for SLCO1B1

genotyping (SNP: rs4149056) for safer prescription of Simvastatin

among Chinese AMI patients for secondary prevention in Singapore.

Our study is the first attempt to explore the cost-effectiveness of clinical

translation of this pharmacogene.

Chapter 6 includes the concluding remarks.

11

Publications

1. Kapoor R, Martinez-Vega R, Dong D, et al. Reducing hypersensitivity

reactions with HLA-B*5701 genotyping before Abacavir prescription:

clinically useful but is it cost-effective in Singapore? Pharmacogenetics

and genomics 2014.

2. Kapoor R, Tan-Koi WC, Teo Y-Y. Role of pharmacogenetics in public

health and clinical health care: a SWOT analysis. Eur J Hum Genet

2016;24:1651-7.

3. Tan-Koi WC, Kapoor R, Teo YY. Pharmacogenetics through a public

health lens: from policy to practice. Pharmacogenetics and genomics

2015;25:518-20.

4. Zou R, Zhou L, Too HP, Kapoor R, Zhu F, Goo P, Rha SY, Chung

HC, Yoong J, Yap CT, Rao J, Chia CK, Tsao S, Shabbir A, Lam K. P.,

Yong W-P, Yeoh K-G, So J.B. Serum microRNA Panel Enables Early

Detection of Gastric Cancer. (In-preparation)

5. Kapoor R, Wee HJ. Cost-Effectiveness and Value of Information

Analysis of SLCO1B1 Genotyping (rs4149056) before Simvastatin

Prescription for Secondary Prevention. (In-preparation)

12

List of Tables

Table 1. Model Inputs ......................................................................... 60

Table 2. Cost effectiveness of HIV treatment strategies for newly diagnosed early and late stage HIV patients contra-indicated to Tenofovir ............................................................................................ 63

Table 3. Cost-effectiveness of HIV treatment strategies for newly-diagnosed early and late-stage HIV patients receptive to both Abacavir and Tenofovir ...................................................................................... 64

Table 4. Cost-effectiveness of HIV treatment strategies- Tenofovir as first line (current practice) with ABC as first line (cheapest strategy) for newly-diagnosed HIV patients receptive to both Abacavir and Tenofovir ........................................................................................................... 66

Table 5. Gastric Cancer Statistics for Singapore ................................ 75

Table 6. Base case values and sensitivity ranges for model inputs (Singapore) ......................................................................................... 87

Table 7. Distributions assigned to parameters in the probabilistic sensitivity analysis (Hospital-clinic) ..................................................... 92

Table 8. Base Case Values and Sensitivity Range for model Inputs specific to Korean healthcare setup .................................................... 93

Table 9. Cost-effectiveness results for Singapore Hospital-clinic scenario comparing the two strategies: ‘endoscopy for all’ and ‘miRNA test followed by endoscopy only for test positive patients’ (modelled for 14 years) ............................................................................................ 95

Table 10. Cost and health benefits estimation in hospital-clinic setting in Singapore ........................................................................................... 96

Table 11. Cost-effectiveness results for population screening of Singaporean Chinese from 50-69 years comparing the two strategies: ‘endoscopy for all’ and ‘miRNA test followed by endoscopy only for test positive patients’ (modelled for 20 years) ........................................... 98

Table 12. Results of base-case analysis for population screening for Singapore Chinese Males (50-69 years). ........................................... 99

Table 13. Sensitivity analysis investigating the impact of improvement in patient compliance with miRNA test on cost-effectiveness of endoscopy in the populations screening setup for Singaporean Chinese males (50-69 years) .......................................................................................... 103

13

Table 14. : Cost-effectiveness results for population screening program in Korea for Adults ≥40 years, comparing the two strategies: endoscopy for all and miRNA test followed by endoscopy only for test positive patients (modelled for 24 years) ....................................................... 104

Table 15. Results of base-case analysis for population screening for population screening program in Korea for Adults ≥40 years ........... 104

Table 16. Model estimates and sources ........................................... 121

Table 17. Cost-effectiveness analysis of the cholesterol lowering strategies for secondary prevention in Singapore ............................. 129

Table 18. 1-year Budget Impact of ezetimibe-Simvastatin combination in Singapore in comparison other cholesterol management strategies 131

Table 19. Evaluation of the rate of side effects across different strategies ......................................................................................................... 131

Table 20. Estimates of the value of additional information to resolve uncertainty ........................................................................................ 131

14

List of Figures

Figure 1. Possible treatment therapies considered for the HIV patient subgroups ........................................................................................... 53

Figure 2. Decision tree model for newly diagnosed HIV patients in Singapore who are contra-indicated to Tenofovir. .............................. 55

Figure 3. Decision tree model of the treatment strategy for newly diagnosed HIV patients in Singapore who can be prescribed Tenofovir or Abacavir ......................................................................................... 56

Figure 4. The decrement in utility values due to occurrence of side effect or ABC-HSS ....................................................................................... 59

Figure 5. Sensitivity analysis for early stage Chinese patients receptive to both Abacavir and Tenofovir ........................................................... 67

Figure 6. Sensitivity analysis of cost-effectiveness to variation in PPV and HLA-B*5701 frequency in early stage HIV patients ..................... 68

Figure 7. Cost effectiveness acceptability curve for early-stage Chinese HIV patient receptive to both Abacavir and Tenofovir ......................... 69

Figure 8. Age Specific Incidence rate of Gastric Cancer in Singapore (2008-2012) ........................................................................................ 76

Figure 9. Markov-Decision Tree Model for the strategy 'endoscopy for all' ....................................................................................................... 81

Figure 10. Decision Tree Model for Strategy 'miRNA screening with endoscopy only for test positive patients' and for the current strategy of No-Screening for the population cohort of Singaporean Chinese 50-69 years ................................................................................................... 82

Figure 11. Sensitivity analysis investigating the impact of gastric cancer incidence rate on cost-effectiveness of endoscopy for all at multiple miRNA test cost scenarios in the hospital-clinic setting ...................... 96

Figure 12. Cost-effectiveness acceptability curve for hospital-clinic scenario .............................................................................................. 97

Figure 13. Sensitivity analysis investigating the impact of gastric cancer incidence rate on cost-effectiveness of endoscopy for all at multiple miRNA test cost scenarios in the populations screening setup for Singaporean Chinese males (50-69 years) ...................................... 100

Figure 14. Cost-effectiveness acceptability curve for population screening of Singaporean Chinese Males (50-69 years) .................. 102

15

Figure 15. Sensitivity analysis investigating the impact of improvement in patient compliance with miRNA test on cost-effectiveness of miRNA-based strategy in comparison with endoscopic screening in the population screening program in Korea (for adults ≥ 40 years) ........ 107

Figure 16. Markov-Decision Tree Model for evaluating the cost-effectiveness of cholesterol management strategies for secondary prevention in AMI patients in Singapore ........................................... 120

Figure 17. Selection criteria for determining the Simvastatin side effect Burden .............................................................................................. 121

Figure 18. Possible cholesterol lowering treatment therapies compared for secondary prevention .................................................................. 125

Figure 19. Cost-effectiveness analysis comparing the four strategies on their cost and effectiveness .............................................................. 130

Figure 20. Tornado analysis evaluating the impact on ICER for genotyping strategy vs. Simvastatin for all ....................................... 132

Figure 21. Cost-effectiveness acceptability curve (Probabilistic Sensitivity analysis) evaluating ezetimibe-Simvastatin combination vs. Simvastatin for all ............................................................................. 133

Figure 22. Cost-effectiveness acceptability curve (Probabilistic Sensitivity analysis) .......................................................................... 134

16

List of Abbreviations

3TC Lamivudine

ABC Abacavir

ABC-HSS Abacavir Hypersensitivity Syndrome

ADR Adverse Drug Reaction

AMI Acute Myocardial Infraction

ASR Age Standardized Incidence Rate

AZT Zidovudine

CK Creatinine Kinase

CPIC Clinical Pharmacogenetics Implementation Consortium

CT Computerized Tomography

CX-R Chest X-Ray

EFV Efavirenz

EMA European Medicines Agency

EUR Endoscopic Ultrasound

FDA Food and Drug Administration, US

FTC Emtricitabine

GDP Gross Domestic Product

GST Goods and Service Tax

HAART Highly Active Antiretroviral Therapy

HIV Human Immunodeficiency Virus

HTA Health Technology Assessment

ICER Incremental Cost-Effectiveness Ratio

LDL Low Density Lipoprotein

miRNA micro RNA (ribo nucleic acid)

Mn Million

NNRTI Non-nucleoside Reverse Transcriptase Inhibitor

NPV Negative Predictive Value

NRTI Nucleoside Reverse Transcriptase Inhibitor

NUH National University Hospital, Singapore

PGx Pharmacogenes

PI Protease Inhibitor

PPV Positive Predictive Value

QALY Quality-Adjusted Life Years

QoL Quality of Life

RR Relative Risks

SNP Single Nucleotide Polymorphism

TDF Tenofovir

UGIS Upper-Gastrointestinal Series

17

ULN Upper limit of Normal Range

VOI Value of Information Analysis

WHO World Health Organization

WTP Willingness-to-Pay

18

Chapter 1 Role of Pharmacogenomics in public health and

clinical healthcare: A SWOT Analysis

The success of the Human Genome Project in 20031 marked the

beginning of a new era in medicine. Genomic research has gained

attention in a multitude of areas– understanding of diseases and drugs,

drug development, pre-symptomatic disease diagnosis, future disease

risk prediction, evaluation of drug compatibility and possibility of drug

adverse reactions. The development of large-scale genome-wide

approaches have led to the advent of pharmacogenomics, a more

diverse field of study which combines the traditional pharmaceutical

sciences such as biochemistry with annotated knowledge of genes,

proteins, RNA’s and single nucleotide polymorphism (SNPs) to develop

newer approached for disease diagnosis and stratified treatment with

greater efficacy and safety.

Pre-symptomatic disease diagnosis is of supreme importance to achieve

a better treatment success across diseases. However, in cancers this is

of compelling concern as cancer treatment have a significantly higher

success rates with improved survival if diagnosed in asymptomatic early

stages as compared to advanced stages when the patients begin to

show symptoms. Medical societies around the world have

recommended cancer screening tests for its population with a few high-

risk countries like South Korea and Japan also providing subsidized

cancer screening at national level2-4. However, in intermediate and low

19

risk countries there are still no pre-emptive cancer screening programs

due to concerns of the additional burden on the healthcare budget and

on the healthcare system. This has spurred the development of novel,

and effective pharmacogenomic screening approaches relying on

biomarker identification like disease-linked miRNA (micro ribonucleic

acid) and SNPs(Single nucleotide polymorphism) which can aid in

development of accurate and cheaper screening technologies5-7.

Inadequate response to drugs and adverse drug reactions, resulting

from incompatibility to medicines, are also a leading cause of death and

have grave health and financial implications. On average only 50% (25-

60%) of patients responds adequately to drugs8. Studies report adverse

drug reactions (ADRs) as a fourth to sixth leading cause of death in US

with 0.32% of patients suffering from ADR related deaths8. Genetic

polymorphisms may alter the enzyme synthesis responsible for drug

metabolism leading to either accumulation of unused drug in the body or

a faster breakdown and elimination, restricting the drug to function

appropriately and leading to inactivity and side effects. Some may also

have a genetic predisposition to drug-related immune responses leading

to drug hypersensitivities. As reported by PharmGKB, there are currently

193 biomarkers recognised by US FDA (Food and Drug Administration)

that can directly enhance patient treatment in diseases like HIV, cardiac

ailment, mental illness, neurological disorders, cancers, hepatitis C,

gout, cystic fibrosis etc9,10 . However, only 59 drug-gene associations

have been approved by the US FDA9, stating required/ recommended

20

genetic testing before drug prescription. Though there seems to be an

immediate health benefit for the patients, the uptake of these

advancements in pharmacogenomics in standard clinical practices has

been startlingly slow.

Thus, here we perform a SWOT analysis examining the strengths,

weaknesses, opportunities and threats around the role of

pharmacogenomics in public health and clinical medicine. The aim of our

analysis is to identify the strengths and opportunities of practicing

pharmacogenomics in public health along with the weaknesses and

threats leading to the sluggish uptake of pharmacogenomics in standard

clinical care. The SWOT analysis also identifies existing gaps in the

current situation, and provides a guide to identify the key areas for future

health systems research and developments to enable favourable

policies for public health translation.

1.1 Strengths of Pharmacogenomics in Clinical Medicine

1.1.1 Faster achievement of optimal drug dosages

Conventional clinical medicines and practices mostly rely on one-size fit

all approach followed by routine monitoring to establish a working

dosage of a compatible drug regimen. For many drugs, commercially

available drug dosages are suited for an “average” patient but can be

easily adjusted based on biometric factors such as weight and age. But

a few of pharmaceuticals demand careful dose titration to achieve the

compatible dosage and the appropriate clinical effect within a narrow

21

therapeutic range leading to multiple follow-up tests and monitoring.

Many of these dosages are found to be influenced by genetic variants

which influence the metabolism of the drug in the body. Using genetic

tests to identify the optimal dosage estimate coupled with a few follow

ups eventually helps in faster achievement of optimal dosage with

minimum toxicity. Warfarin11,12 irinotecan13 and atomoxetine14 are

examples of medications where genetic variant combinations strongly

correlates with different dose-response curves.

1.1.2 Minimising toxicity and adverse side effects

The potential to use pharmacogenetics to identify at-risk patients

prospectively poses a very attractive tool, especially when adverse

events from medications constitute as one of the leading causes of

mortality and a huge economic burden15-17. Recent studies have

uncovered numerous genetic linkages with drug-induced toxicity and/or

side effects, where an individual’s genetic profile can provide early

indication of the likelihood of an undesirable outcome and direct the

physician towards prescribing a possible alternative. Abacavir and

carbamazepine are two such befitting examples, especially as patients

who do not carry risk alleles (HLA-B*57:01 and HLA-B*15:02 alleles

respectively) for these two drugs will almost never experience a side

effect, namely, Abacavir-induced hypersensitivity or carbamazepine-

induced –Stevens-Johnson Syndrome (SJS). Conversely, patients who

carry these risk alleles present over hundred-fold increase in risk of

unpredicted potentially fatal side effects and physicians would almost

22

always recommend alternatives as a matter of good clinical practice.

Prospective trials of HLA-B*57:01 screening before Abacavir

prescription have indeed reported striking success with significantly

reduced adverse incidents rates by substituting alternatives for the risk

allele carriers18,19. An assurance of reduced side effects among patients

who are non-carriers of the risk allele also promises improved drug

compliance20, which in itself is a major clinical challenge and of immense

economic value to the healthcare system21.

1.1.3 Identifying efficacious drugs

Using pharmacogenomics to identify efficacious medication for patients

forms the cornerstone of practicing stratified medicine. With a

prospective approach, the Pharmacogenomic Resource for Enhanced

Decisions in Care and Treatment (PREDICT) program by Vanderbilt

University has already illustrated successful examples, for instance one

where physicians could use patients genetic profiles to appropriately

identify the compatible statin with suitable dosage for a heart patient

enduring the aftereffects due to incompatible statin treatments for

years22. Trastuzumab, commonly known as Herceptin®, is also an

effective therapy for cancer patients who are HER2 positive (breast or

gastric), lowering 3-year cancer relapse risk by almost 10% and

improving survival23, but offers no such benefit for HER2 negative

patients. The ability to identify an efficacious treatment, especially for a

debilitating and traumatic disease such as cancer, remarkably increases

patient confidence in both the genetic testing and treatment and the

23

possibility to recover as well, who otherwise get jinxed with a

deteriorating health and the huge bills for potentially harmful or

ineffective treatments.

1.1.4 Reduce overall costs to the healthcare system

Pharmacogenomic testing with an increased upfront cost due to

additional tests are mostly perceived to increase the economic burden

on the treatment. However, many a times genetically-related

medications are cheaper than their counterparts and identification of a

suitable population segment would help to improve not only the

treatment response but also the long-term medication costs. Drugs such

as Abacavir and Simvastatin with related genetic tests to help avoid

undesirable side effects are such examples as they are considerably

cheaper than their respective alternatives (Tenofovir, Alirocumab and

other stating like Atorvastatin, Rosuvastatin etc). In the absence of

genetic tests, assigning expensive alternatives to all incur unnecessary

expenses to the healthcare system along with the risk of alternative

forms of side effects, whereas unguided use of cheaper alternatives is

also not desirable with the risk of unwanted adverse events. Thus, in

such a scenario, genetic stratification of patients could help guide

prescription of costlier alternatives only to those who cannot tolerate the

cheaper medication. This may help to reduce the overall side effect

profile and generate significant cost-savings in the longer run as well. A

similar example is HLA-B*15:02 screening before carbamazepine

prescription in East Asians24-26, delivering overall savings to the

24

healthcare system despite having a low benefit to screening ratio.

However, it should be noted that these cost-savings are specific to

healthcare systems and are not directly transferable, as depicted by

studies concluding cost-effectiveness of HLA-B*57:01 genetic testing

before Abacavir in European populations27 but not being a cost-effective

strategy for Southeast Asian populations28. Patient stratification in risk

categories would also help the physician devise personalised follow-up

plans, ensuring strict monitoring schedule for the high-risk patients and

possibly not-so-strict one for those at low-risk. Along with ensuring an

improved drug risk profile, this might also help optimise the physician’s

workload and possibly save the unsusceptible patients some extra funds

and efforts by avoiding the unnecessary physician visits.

1.2 Weaknesses of Pharmacogenomics in Clinical Medicine

1.2.1 Costs of Pharmacogenomic tests to the individuals

Producing an individual’s pharmacogenomic information is an additional

expense on top of standard clinical care. Although the government/

healthcare agencies reimburse some pharmacogenomic tests, they are

still typically limited to only a handful of cancer-related and HLA-targeted

genetic tests (e.g. HLA-B*15:02 testing in Singapore29,30 and

Thailand31), even in countries like United Kingdom, Singapore and

Thailand with comprehensive universal health coverage (UHC) and

government subsidies. Many other medicines with clinically proven

genetically related potentially fatal side effects (e.g. Allopurinol32,

25

Codeine33 etc) and newer methods of cancer screening like miRNA

testing are not covered with most of the healthcare systems currently

passing on the expense of these pharmacogenetic tests to the

individuals.

1.2.2 Speed of genetic testing to aid in clinical decisions

The generation of test results in a timely manner is as important as the

accuracy of the test itself and its clinical impact. It is important to get the

results timely to contribute in decision making. For example, in Warfarin,

3-7 days are sufficient to adjust the International Normalised Ratio (INR)

to therapeutic range through rigorous monitoring. Testing for HLA alleles

to inform prescription of medications such as Abacavir, carbamazepine

or allopurinol can take even longer, although prescription immediacy is

often relatively less crucial for such patients. Though the turn-around

time of the actual test may be in days, the timeline of the entire

operations from obtaining patient’s samples to delivering the report to

the physician for clinical decision immensely concerns the practical

applicability in clinics, making it a rate determining step in the successful

clinical translation of pharmacogenomics

1.2.3 Imperfect understanding of genetically driven drug

response variability

Except a few of gene-drug associations, mostly HLA alleles-induced

adverse drug reactions and success stories in oncological treatments,

the role of genetics to explain the drug response variability is yet to be

well translated for majority of pharmacogenomic discoveries. For

26

example, the alleles in VKORC1 and CYP2C9, clinically useful for

predicting warfarin dosing explain less than 50% of the INR dosing

variation, with compelling benefits only in a few populations34,35. Rare

variants are also expected to contribute significantly to the functional

variability in pharmacogenes, for instance response variability estimated

to be 17.8% due to rare variants in SLCO1B136 for the drug

methotrexate. This incomplete understanding of the genetic impact on

drug outcomes is likely to affect the confidence of physicians and

patients to genetic testing.

1.3 Opportunities of pharmacogenomics in clinical medicine

1.3.1 Technological innovations and falling prices of genetic tests

The cost of whole genome sequencing has exponentially reduced from

USD3 billion in 2001 to less than USD 1,000 in 2016. Leveraging the

repository of genetic information gathered from thousands of whole

genome sequences, elaborate microarrays which incorporates about

500k -1 million clinically relevant genetic biomarkers have been

designed for relatively lower prices. With such low prices, genetic

information can be synthesized and possibly easily integrated into an

individual’s health records even before the need arises. This thwarts the

weakness of lack of timely procurement of test results to be of practical

usage. Technological innovations have also brought up a direct-to-

consumer market for genetic tests where the decision to genotype is

taken by the individual himself. Rapid test kits are very accessible to

27

order and test for general practitioners and specialists alike. In addition,

multiple initiatives for easy transfer of such data have also been

undertaken, for example - medicare safety code initiative (saving data in

QR code readable easily with a smartphone)37 and pharmacogenomic

IDs38. However, the framework to establish secure and easily accessible

storage services for huge genomic datasets has already begun with the

development of google genomics and its collaboration with Broad

institute. These developments greatly improve the accessibility to

genomic information and provide a platform for pharmacogenomics to

play a bigger role in healthcare.

1.3.2 Next generation sequencing to encourage individualized

pharmacogenomics

With classic pharmacogenomics focusing only on the common variants,

the concept of adapting prescription conforming to individual specific

rare-variants by interpreting individual’s genome is gaining more

importance. Rare variants, have been found to be critical in affecting

drug response, accounting for 30-40% of functional variability, which in-

turn potentially affects multiple drugs pharmacokinetics or

pharmacodynamics39. Thus, research projects (like personal genome

project) and international collaborations have been launched to promote

personal genomics and achieve true treatment personalization.

28

1.3.3 Innovation in healthcare that impacts drug development and

utility

Pharmacogenomics possesses the potential to change the paradigm of

medicine, not only in the prescription of drugs but also in their discovery

and development40. The concept of clinical trials guided with

pharmacogenomics principles is to improve and accelerate drug

development: by correlating patient’s genetic profiles with treatment

outcomes (mainly around safety and efficacy) in early phases of the

clinical trials, and consequently extending Phase III trials to patients with

favourable genotypes. This provides a significantly more precise

approach for drug development leading to a lower attrition of drug

candidates and hence lowering the overall cost of drug development.

Pharmaceutical companies can potentially be the main driver in

promoting use of pharmacogenomics as a standard of care, by

introducing dosage guidelines and safety profiles based on

pharmacogenomic information, thereby compelling the need to genotype

before prescription.

1.3.4 Public pharmacogenomics network on clinical

implementation

Numerous International consortiums have been developed to inspire

application of pharmacogenomics in clinical practice. CPIC, (Clinical

Pharmacogenomics Implementation Consortium, https://cpicpgx.org/) is

an online resource managed collectively by the Pharmacogenomics

Research Network and PharmGKB, with an aim to provide thoroughly

curated evidence and guide on the use of genetic tests in prescribing

29

and optimising drug41. Similar initiative called Dutch Pharmacogenetics

Working Group (DPWG) has been launched by The Royal Dutch

Pharmacist’s Association in Europe42. These consortiums not only focus

on educating on the need to order a genetic test but on the interpretability

of test results to enable genetically guided treatment. IGNITE

(Implementing Genomics in Practice), is an international consortium

funded by NIH and has been started to support in enabling effective

translation, and sustainability of using genomic information in diverse

clinical settings, integration of genomic information in electronic medical

health records and hence provide the much-needed clinical decision

support. These efforts are crucial in translation of pharmacogenomics in

the clinics, boosting the physician’s confidence by providing

internationally accredited portals to supplement and update physician’s

knowledge of new pharmacogenomic guidelines. Also with the rising

popularity of direct-to-consumer genetic tests, such portals can assist to

educate and guide patients as well.

1.3.5 Adoption of pharmacogenomics in outpatient settings

With current implementation of pharmacogenomics witnessed mostly in

specialized, inpatient settings within academic or tertiary medical

centres, integration of pharmacogenomics in outpatient settings like

community pharmacies can substantially help in its widespread adoption

in clinical practice43,44. With these being the most easily accessible

healthcare providers, outpatient settings have substantially contributed

to improve health outcomes in the past45,46 and can play a critical role

30

today in imparting the benefits of pharmacogenomics to the community.

With pilot studies already initiated in this sphere47, effective educational

strategies for pharmacists, strict vigilance to ensure practice of evidence

based approaches and policies to safeguard from commercial financial

interests are inevitable to empower the outpatient settings implement

pharmacogenomics effectively in the community.

1.4 Threats of pharmacogenomics in clinical medicine

1.4.1 Availability of non-genetics alternatives to physicians

There is the tendency that physicians view pharmacogenomics with

scepticism in comparison to the convention approach of an average

prescription, even for medications with genetically related safety and

efficacy concerns is perhaps the greatest threat undermining its clinical

translation. The overall benefits of genetics-based dosing of warfarin has

been challenged by clinical trials illustrating near-similar results using

conventional methods of dose optimization based on clinical factors and

INR monitoring48,49. However, the efficiency of such non-genetic

alternatives specifically that of aggressive monitoring, is hugely

dependent on implementation of a perfect healthcare system that can

diagnose, monitor and circumvent adverse effects or re-adjust drug

dosages in a timely manner. A not-so perfect healthcare system with

less rigorous monitoring due to lack of facilities or patient’s unwillingness

for continued follow-ups may lead to a surge in delayed or missed

diagnosis of adverse events and/or drug failures, for which genetic tests

would act as early preventive measures and help to minimise

31

1.4.2 Misaligned incentives for clinical discovery versus clinical

implementation activities

Currently a disproportionate amount of investment is being done on

pharmacogenomic studies focused on clinical discovery and validation

and less emphasis is being given to the translational phase. The

research studies with the familiar agenda to address the funding

agencies demands are easy to conduct and supervise, whereas the

translational phase studies introduce new complexities. With the need of

systems-level changes for translation in the clinics and incorporation in

national and international level policies/guidelines – a process with

which most clinician scientists are unfamiliar or even averse to. For

example, establishment of an authorized pipeline to certify the use of

pharmacogenomics in the clinic (from the point of blood withdrawal, bio-

informatics or microarray analysis to certified report generation);

provision of manpower training for post-testing genetic counselling;

incorporation of pharmacogenomics in medical schools’ curriculum; and

generation of evidence to guide formulation of national/ international

regulatory legislations. These formative transformations to the

healthcare infrastructure involve the consent of governments, hospital

authorities and universities alike. With the unfamiliarity and intrinsic

complexities of translational programs, the majority of actual research

activities and funding requests have remained focused around clinical

discovery. However, the existing statue of disconnect is unsustainable,

especially when the actual use of the technology in clinics is what both

the research and funding agencies aim to achieve.

32

1.4.3 Lack of healthcare infrastructure

Implementation of pharmacogenomic as a standard of care requires

multiple amendments to the healthcare infrastructure – availability of

data grid, electronic health records to integrate genetic information with

medical records, a platform for data sharing across healthcare

establishment, safety structures to ensure privacy of pharmacogenomic

information, trained manpower for test implementation and

interpretation, genetic counsellors, and the fundamental capital

infrastructure development. However, very few health systems currently

possess a capability to simultaneously accommodate and share genetic

information, leading to concerns over the initial establishment costs

involved.

1.4.4 Mistrust over management and use of genetic information

The flow-over implication of pharmacogenomic information beyond the

individual also adds hesitancy to patient adoption. Pharmacogenomic

information many a times does not solely offer insights into only the

individual-specific biology and disease risks, but also of the family.

Confidentiality of genetic details needs to be secured by the overall

healthcare system, with standard guidelines in place to advice

management of incidental findings especially those related to disease

risks. Insurance agencies can also potentially misuse genomic

information to offer/refuse medical policies over genetic risks50-52. This

fear of misuse of genetic information as a criterion of discrimination in

the society can deteriorate the acceptability of these methods.

33

1.4.5 Lack of buy-in by healthcare financiers

The dilemma with such tests is the tricky predicament faced by

healthcare financiers: to bear tests costs for everyone and sponsor

alternatives among patients incompatible to standard treatment

regimens as compared to the standard treatment for all. Even with

genetically increased risk of a potential drug incompatibility, only a

fraction of such risk-sensitive individuals will eventually end up

experiencing a suboptimal outcome, which many times are already

arrested with routine monitoring. This creates a compelling case to

favour standard treatment especially for chronic diseases with long-term

medications before switching to genetically compatible potentially

costlier alternatives. However, the individuals have a higher motivated

to seek the best possible healthcare. This discrepancy between patient-

centric healthcare and universal healthcare coverage may lead to

unwillingness to cover and reimburse, what may be perceived as an,

unnecessary cost by the financiers.

1.4.6 Higher institutional risk and threats of lawsuits

Though, pharmacogenomic based prescription of alternatives for

patients at high-risk of drug incompatibility/ inefficacy helps physician

provide good clinical care avoiding both - unnecessary adverse events

and any potential legal troubles arising from such complication; there are

scenarios where it may work to the contrary. The interpretability of

pharmacogenomics relies on a complex set of recommendations based

on statistical concepts of positive and negative predictive values.

34

Relegating clinical decision-making to the interpretation of a set of

statistical probabilities inevitably increases institutional risks and the

threats of lawsuits to the healthcare sector, especially when patients are

increasingly risk averse and patient preference for genetic testing may

differ from the physician’s recommendations. This is particularly in the

case of rare adverse outcomes or for low-risk patient groups where

benefits of the medication far outweigh the rare risk of adverse events.

In addition, where such rare occurrence happens independent of

medication or genetic factors, insufficient counselling of such difficult-to-

grasp concepts may mislead patients to assume that the event has

resulted from physician’s oversight and mismanagement. Additionally,

though development of personalised follow-up schedules for patients

based on their risk-groups to improve overall care and save both money

and time seems as a win-win situation, currently drug label inserts with

pharmacogenomic advisories predominantly focus on patients who carry

risk-alleles. There are no recommendations for risk-allele negative

individual and given the incomplete understanding on the role of genetics

in drug response variation, physicians might be reluctant or even scared

of legal consequences to place too much confidence in genotype-

negativity and reduce the level of monitoring.

1.4.7 Widening inequality in healthcare accessibility

One of the unintended after-effects of using pharmacogenomic

techniques in clinics is to widen the inequality in accessibility of

healthcare in the society. It is likely that with the slow incorporation of

35

these techniques in the policies, owing to high cost of establishments

and low acceptability, majority of them may remain uncovered by

insurance companies and as an out-of-pocket expense. This would lead

to a disparity to access among the wealthy in the society. Also, at the

country level, developing countries may consider this advanced

healthcare technology as secondary to the establishment of classical

essential healthcare whereas the developed economies would have the

resources to offer these advanced technologies improving the quality of

care.

1.5 Discussion

Even though today we witness uncertainty about the application of

pharmacogenomics in clinics, no one doubts the enormous public health

benefits pharmacogenomics entails. Multiple efforts are already

underway with numerous organizations and consortiums bringing

researchers, technologists and payer and patient communities together

to help devise ways of successful implementation. Numerous expensive

clinical trials and programs (like PREDICT, UPGRADE etc.,) have begun

to prove the benefits of translation of pharmacogenomics as a standard

of care. However, the healthcare community gravely requires not only

clinical discovery research but also significant efforts and insistence for

translating proven discoveries to impactful solutions in the standard of

care and rectify the implementation hindrances in the health system.

Here, an evidence-based approach would be essential to generate

36

robust and convincing quantitative proof of the value the new healthcare

intervention would add, and this must be performed before regulatory

agencies can be convinced on whether the genetic test ought to be

implemented in the system.

37

Chapter 2 Health Technology Assessment: A logic based

approach to regulatory decision making

As analyzed in the previous chapter, pharmacogenomics accompanies

itself with multiple strengths and opportunities to benefit public health but

also weaknesses and threats, which have led to its sluggish uptake in

actual practice.

Though discovering significant drug-gene associations has always been

a core requirement of pharmacogenomics, its successful clinical

application is the fundamental cornerstone to derive tangible benefits

from all the research. Intuitively a pharmacogenomic (PGx) test seems

like any other medical test but there are intricate complexities underlying

this healthcare proposition. In comparison to a medical test which

immediately helps to diagnose a disease or evaluate overall health,

application of PGx testing before drug prescription claims to prevent a

“potential side effect” or lead to “better treatment efficacy”. However, in

many cases, these may not lead to immediately quantifiable health

benefits like avoiding a life-threatening side effects or guaranteed early

disease recovery. Also, the medical tests have restricted predictive

ability and even if accurate, the genetic associations are able to explain

only limited variability in patient responses. Accompanied with lack of

awareness, low confidence, availability of more familiar alternatives and

many other factors, clinicians tend to be suspicious and prefer traditional

38

methods like trial and error and treatment response tracking to identify

any possible need of therapy change.

However, even with these weaknesses and threats, pharmacogenomic

associations are currently the strongest predictors to realize the lurking

patient-specific drug-response variability. The well documented

evidences of pharmacogenomics9, present a compelling case of its

clinical utility to achieve an efficient stratified system of care.

A fundamental necessity to integrate PGx testing successfully in

standard clinical care is identification of a comprehensive framework of

implementation which offers better health benefits with affordable costs.

Here economic viability stands out as an elemental factor to ensure

sustainability of this intervention. An elemental question rises in the mind

of the patient and health systems alike - Are the test benefits worth the

money? Healthcare systems today are reeling under multiple challenges

– ageing population, increasing burden of chronic diseases, emerging

threats and unknown diseases, innovation and rapid developments in

medical technologies, high user expectation and a worldwide goal to

provide best possible healthcare to all. With a rise in demand and

increasing innovative healthcare solutions and technologies, but limited

resources, the healthcare authorities faces many difficult questions. How

do they decide which new technology, prevention program or healthcare

service delivery model to adopt? How do they determine if their

recommendations have acceptable value for money? With budget

39

constraints there is an inability to afford everything that is clinically

useful. A decision to implement one healthcare activity inevitably

eliminates the resources for some other program, compelling the policy

makers to make important choices. This demands a well-informed

system to support prioritization of resources to be able to provide the

highest “value for money” and help policy makers ensure maximize

health for its population.

These concerns can be addressed by Health Technology Assessment

(HTA) – a multidisciplinary process to evaluate the economic, social,

organizational and ethical issues of a healthcare intervention. Health

technology assessment acts as a tool to compare the current against

new interventions and inform evidence-based, sustainable and

pragmatic policy decisions with budget impact considerations. Such

analysis accounts for long term consequences (both pros and cons) of

health interventions, and helps identify justifiable initial investment on

basis of long term gains and budget impacts. Though such evaluations

do not provide an immediate answer, they act as tools to develop

scenario specific models and provide evidence to identify the key

consequences of allocating resources in different programs. There are

many economic evaluation frameworks to quantify the costs and/or

health benefits – analysis of cost-minimization, cost-benefit, cost-

effectiveness and cost-utility, among which cost-effectiveness analysis,

has particularly gained a lot of popularity among the policymakers.

40

Health technology assessment when broadly applied also includes

social, ethical and legal aspects of health approaches.

Identifying an intervention which helps achieve better health benefits at

a lower cost is the best possible scenario. However, in many cases

betterment of care using an advanced technology involves an additional

cost. Cost-effectiveness evaluation provides a comparative analysis of

two or more proposed interventions based on measurement of their

costs (direct and indirect costs) and achieved health benefits. The health

benefits can be quantified by measuring favourable health outcomes or

adverse events/ mortality avoided or by using a more quantitative

measure called QALY (quality adjusted life years). Used in cost- utility

analysis, a specialised form of cost-effectiveness analysis, a QALY

represents one year in perfect health. It is calculated by using quality-of-

life (QoL) scale - an index measuring health on a 0-1 scale (with death

and perfect health represented by the extremes respectively). QoL

multiplied by the number of years lived in that condition represents the

total QALYs for the patient. A cost-effective strategy helps to validate the

benefit of an intervention’s underlying expenses and identify a superior

healthcare strategy, the population segment where it is most cost-

effective and the preferred cost range and duration of implementation as

well. The less frequently used methodologies include cost-minimization

which helps identify the cheapest strategy and cost-benefit evaluations

which measures the cost and health benefits in monetary terms, which

of increased interest of private payer organisations.

41

Cost-effectiveness analysis involves decision modelling to synthesize

evidence for the different strategies. Along with information about the

clinical effectiveness of the intervention, knowledge about the

epidemiology of the disease, characteristics of the population segment,

natural history of disease, healthcare services availability and overall

acceptability of the proposed interventions all play a role to derive

substantial evidence to identify the best intervention for the target

population segment. Countries with varied characteristics for the above-

mentioned parameters might have different interventions appearing as

most cost-effective, imparting this research methodology a much

personalised flavour. Below we have tried to innumerate five such

important parameters, which make cost-effectiveness analysis a

methodology specific to a country, disease and population segment.

2.1 Perspective of Analysis

Perspective of cost-effectiveness analysis is a very important aspect,

which can generate varying recommendations for the same intervention.

Three main perspectives which cost-effectiveness analysis is performed

with are: patient’s perspective, payer perspective (insurance or

government healthcare system) and policy-maker (societal) perspective.

Provider perspective (perspective of the clinician), is also an additional

perspective which can influence the decisions of the patient and payer.

Every perspective values costs and health benefits from their view point

– patient perspective valuing the individual’s health the most with a

42

concern of out-of-pocket expense, payer perspective evaluating the

public health benefits and contribution to economy over individual’s

health (if government is the payer) or organization’s profit (if the payer is

a private insurance company). Confusions and misunderstandings

unavoidably arises in above two perspectives with conflicts in their

fundamental interests. Thus, there is need of clear distinction in the

evaluation between private healthcare where additional cost will be

borne by the individual who will enjoy the benefits, versus reimbursable

healthcare (by government or insurance), where the patient would

expect the best care irrespective of the cost.

In government subsidized healthcare setting, where the payer aims to

give its population the best healthcare, the unlimited needs of the

population has led to an immense burden on health care budget,

highlighting the need of an evaluation method to prioritize resource

utilization for the benefit of the entire society. This introduces the societal

perspective which analyzes from the society’s viewpoint, the benefits of

a healthcare intervention for public health and contribution to the

economy which would benefit the whole society. This perspective

intends to make all parties aware of the overall effects and forms the

basis to make decisions which is fair for all. This is in line with the goals

of the healthcare systems which aim to utilize the resources in the public

interest.

43

2.2 Allele frequencies, Disease Severity and Prevalence

A new test/ therapy to identify a compatible treatment for a severe illness

and/or avoid a life-threatening or physically debilitating side effects is

welcomed in the clinical settings as they seem cost-effective intuitively.

This mirrors the successful implementation of genetic tests to provide

personalized care to cancer patients53 and in various other serious

illnesses like HIV54 and Epilepsy55. On the contrary, with a lower

prevalence of the risk allele, these clinical practices recommending

screening for all may in turn become economically burdensome as only

a few patients would carry the risk genotype. HLA-B*5701 testing before

Abacavir, a success story of clinical application of genetic testing to

prevent potentially life threatening adverse reaction in HIV patients

carrying the risk-allele, suffers from such a dilemma. While studies in

US27 and UK56 prove its cost-effectiveness, resulting in FDA

recommendation in favor of testing, a similar study in Singapore proves

it to be not cost-effective28 (see Chapter 3). With a lower prevalence of

HIV in Singapore, and a very low risk allele frequency among

Singaporean Chinese and Malays (1% - 2%), the huge cost expenditure

of screening all HIV patients to avoid a handful of adverse events

renders this clinically useful test, economically unfavorable. HLA-B*1502

screening in epilepsy patients for preventing carbamazepine-induced

SJS (Stevens- Johnson syndrome) is another such example which is

proved cost-effective in Singapore24 and Thailand26 but is not an

economically preferred option in the US55.

44

As we note that a low frequency of risk-allele seems to increase the

economic burden of preventive testing, but with a high disease

prevalence, these public health interventions can drive substantial health

benefits and reinforce their importance by achieving health benefits

worth for the overall expense. A well fitted example is cardiovascular

diseases. Currently FDA warning exists for two widely used drugs in this

therapeutic area – Warfarin (anti-coagulant)57 and Clopidogrel (anti-

platelet)58. Simvastatin (a cholesterol lowering drug) also has an FDA

alert limiting its dosage59, with research attributing this risk to genetic

causes60. With about 32% (73.5 million) adults suffering from high

cholesterol61, 29% (70 million) with hypertension62 and approximately

one death every minute due to heart disease related event in US alone63,

cardiovascular diseases poses an enormous public health burden. High

cholesterol, being a prevalent risk factor approximately doubles the risk

of a heart disease64. With a widespread Simvastatin usage, even a low

incidence rate of rhabdomyolysis (3.4 per 100,000 person years with

subsequent 10% mortality)65 - a potential life threatening adverse event

with an existing PGx biomarker60,66, the large absolute patient numbers

and ability to save even some of them using PGx testing stresses on the

benefits of its inclusion as a standard-of-care.

In a scenario where, irrespective of drug’s risk profile based on patient

genetic makeup, it still remains the prescription of choice due to lack of

alternatives, an identification of high-risk patients would help clinicians

in implementing a more aggressive follow-up plan with enhanced patient

45

education measures to help in early diagnosis of adverse events or

treatment inadequacy. This would be instrumental to strengthen the

drug’s benefit profile and reduce the resultant side effects.

2.3 Patients Adherence

Patient adherence is a major parameter governing the success of any

healthcare intervention. Though lot of resources are being invested in

the diagnosis and provision of best treatment to all, the willingness of the

patient to be compliant to the treatment is of vital concern for the overall

success. Medication non-adherence though difficult to quantify is

estimated to be yet another immense economic burden on healthcare,

amounting to more than USD300 billion in the US alone67.

The patient’s non-adherence can have important implications on the

cost-effectiveness of healthcare interventions. Introduction of a new

sophisticated medical technology with significantly better health benefits

but invasive/ logistically demanding procedure may appear very

attractive for its clinical utility but not so cost-effective due to lower

patient willingness. Moderate side effects which are not of major clinical

concern sometimes also inflict an increase in patient discontinuation

rates influencing the cost effectiveness of its prevention strategies. For

example, recently there has been an increasing concern for mild muscle

related side effects of statins like myalgia and myopathy, which though

not guilty of grave medical consequences, contribute as one of the

leading causes of non-compliance among statin- takers68. With non-

46

adherence as high as 75% in the first year itself69,70 it is a major

challenge in practicing effective medicine, leading to poor cholesterol

management among patients and high rate of heart diseases. Thus,

cost-effectiveness of implementation of a SLCO1B1 genetic testing to

stratify Simvastatin users by their side effect risk and achieve safer statin

prescription may be potentially worthwhile predominantly for its ability to

achieve improved adherence.

Currently most of the cost-effectiveness analyses are performed

assuming perfect patient adherence, which is hardly the case. Thus,

along with the improved clinical utility, the willingness to be accepted by

the target population is also vital to identify the cost-effective solutions

and formulate sustainable policy decisions.

2.4 Willingness-To-Pay (Cost-effectiveness threshold)

The patient’s or society’s willingness to pay to accept a solution for a

healthcare problem is critical in health technology assessment. Since

many years the world health organization (WHO) threshold of

USD50,000 or 1-3 GDP (gross domestic product) per capita for gain of

one QALY has been the most commonly used benchmark71. However,

a society’s true willingness-to-pay for health benefits may vary with its

healthcare budget and priorities for public health. Even within a country,

health care priorities may vary with economic settings, especially for a

private or insurance based healthcare system.

47

A clearer idea of willingness to pay of the individual or the target society

would add a lot of value to the actual translation of cost-effective

interventions in policies and practice.

2.5 Healthcare system

A country’s healthcare setup can also influence the benefits which can

be derived from an intervention. An aggressive and well executed

healthcare system would diagnose and cure the side effects or readjust

the doses timely, undermining the benefits of the preventive measures,

whereas a country’s negligent medical network can in-turn lead to

missed diagnosis and consequent adverse events/ mortalities,

increasing the benefits derived from initial preventive tests. Thus, even

for the strategy with the same clinical benefit in two countries, a

pragmatic comparative analysis of the way a healthcare system

responds to a condition and the benefits derived from a proposed

intervention are crucial to determine the actual cost-effectiveness of its

clinical translation.

For example, Warfarin is a drug with a well-established

pharmacogenomic association72 with an FDA approved drug label73 and

an online Warfarin dosing algorithm recommended by CPIC (Clinical

Pharmacogenetics Implementation Consortium)74 which stresses on the

importance of genome-based dosage optimization. Though, not denying

the therapeutic benefits of the PGx based Warfarin dosing, many recent

clinical trials have challenged the overall benefits by illustrating near

48

similar results by dosage optimization based on other clinical factors and

INR monitoring48,49. However, the study performed in the strict setup of

clinical trial is unable to capture the true effectiveness of non-

pharmacogenetic interventions as it fails to account for the

responsiveness of healthcare setups, ease of access of healthcare

facilities and availability of healthcare professionals, patient willingness

for repeated consultations and the overall expenditure due to repeated

visits, consultation and logistics as well.

2.6 Discussion

Thus, cost-effectiveness evaluation is not simply measured by

treatment-related expenses and the health benefits, but involves a

complex approach accounting for the multiple pragmatic parameters

impacting the healthcare availability, implementation and uptake. Failure

to adopt a long-range national-level perspective to cost-effectiveness is

otherwise adopting a myopic view that undermines the development of

sustainable and beneficial national healthcare strategy.

In the next three chapters we have provided three cost-effectiveness

evaluations for different healthcare problems and represent how

beneficial evidence can be generated across disease conditions and

healthcare setups.

49

Chapter 3 Cost Effectiveness Analysis of HLA-B*5701

Genotyping in Newly Diagnosed Persons Living with

HIV/AIDS in Singapore, by Ethnicity

3.1 Introduction

Highly active anti-retroviral therapy (HAART) is an extremely efficacious

three drug combination therapy, credited to progress HIV from a fatal to

a chronic disease. It includes a combination of different classes of drugs,

2 NRTI’s (Nucleoside Reverse Transcriptase Inhibitor) with usually 1

NNRTI (Non- Nucleoside Reverse Transcriptase Inhibitor) or sometimes

a PI (Protease Inhibitor). Tenofovir (TDF), currently the preferred first-

line NRTI is WHO recommended and among the most commonly

prescribed drug (with Lamivudine (3TC)/ Emtricitabine (FTC): NRTI and

Efavirenz (EFV): the NNRTI. Abacavir (ABC) and Zidovudin (AZT) are

the recommended alternatives in case of any Tenofovir related side

effects75.

Though Abacavir is currently used as an alternative 1st line drug, its

comparable in-vitro potency, good bio-availability and tolerability and no

dietary regulations76, makes it a contender for being a first-line drug.

Studies have shown Abacavir and Tenofovir efficacy to be comparable

for patients with viral load <105 copies/ml77. However, for patients with

viral load >105 copies/ml, results are disputed with one study reporting

similar but another reporting a lower efficacy than Tenofovir77,78. Also,

Abacavir combination with lamivudine is ~ 40% cheaper than similar

50

Tenofovir combination in Singapore. Though similar efficacy and lower

cost drives Abacavir as a preferred choice, one of its major drawbacks

is the Abacavir associated hypersensitivity syndrome (ABC-HSS).

Abacavir has been found to cause HSS in approximately 5% of its users’

globally. Characterized initially by general symptoms like rash, fever,

gastro-intestinal issues, constitutional problems, and respiratory

complaints, it initiates mostly within the first 6 weeks after drug

prescription and demands immediate drug cessation which can bring

improvements in symptoms as early as 48-72 hours without any

additional medication. However, with Abacavir re-challenge, the

symptoms tend to worsen and lead to life-threatening reactions or even

sudden death79,80.

Researchers have proved strong genetic association between HLA-

B*5701 allele and the occurrence of Abacavir hypersensitivity

syndrome81. Based on clinical studies proving a statistical decrease in

ABC-HSS cases driven by pre-HLA-B*5701 genetic screening, the US

Food and Drug Administration (FDA) has recommended HLA-B*5701

genetic screening in patients before initiating or re-starting Abacavir or

Abacavir-containing medication and does not recommend it in any form

to test positive patients19,81. Studies conducted in US82 and UK56 has

also concluded its cost-effectiveness. However, with difference in

population dynamics, these studies may not be aptly translated to

Singapore population.

51

The variation of HLA-B*5701 allele frequency across ethnicities governs

the incidence rate of ABC-HSS on Abacavir usage which in turn

influences the effectiveness of the HLA-B*5701 screening test in

preventing ABC-HSS cases. Variations in cost of drugs, genetic

screening tests and medical consultation, medical setup and supervision

system across countries are among other factors governing the potential

harms posed by Abacavir side effects on HIV patients in Singapore.

Thus, this study aims to analyze the cost-effectiveness of HLA-B*5701

genetic screening prior to Abacavir prescription specific to Singapore

healthcare setup. It also focuses to compare all the possible HIV

treatment regimens with Abacavir as a potential first/ second-line drug

with or without HLA-B*5701 screening to be able to weigh the possible

benefits and guide HIV treatment regimen towards a cost-effective

approach.

3.2 Methods

3.2.1 Model Structure

A decision tree model was developed in TreeAge Pro2013 (Figure 2,

Figure 3) to help evaluate the cost-effectiveness of HLA-B*5701 genetic

screening prior to Abacavir prescription separately for the three major

ethnicities in Singapore – Chinese, Malays and Indians. As HIV patients

diagnosed at different disease stages have significant variation in life

expectancies, separate models were developed for patients diagnosed

with early stage HIV and late stage HIV/ AIDS with a further

52

segmentation identifying presence of any contra-indication for Tenofovir.

All possible treatment strategies were compared (Figure 1). The model

considered similar efficacy for all drugs of concern with different side

effect rates based on Singapore HIV patient data obtained from national

university hospital, Singapore. Life-long treatment duration was

modelled, with a life expectancy of 30 years for early stage HIV patients

and 10 years for late stage HIV/ AIDS patients on HAART after

diagnoses (mean diagnosis age ~40 & 50 years respectively)83,84. With

a patient being either compatible to a treatment or facing an intolerable

side effect, all the possibilities were modelled - being successfully on the

first-line treatment life-long or facing intolerable side effects and

successfully adhering to second, third or fourth line. Hypersensitivity

syndrome (ABC-HSS) was considered as an additional outcome among

Abacavir users leading to either full recovery or fatality in 30 days85. If a

patient fails even the 3rd-line, he/she is prescribed a personalized

combination of alternative drugs (stavudine, lamivudine, emtricitabine,

didanosine, raltegravir, darunavir, atazanavir, lopinavir, ritonavir etc)

referred to as hypothetical drug, which continues for the remaining life

tenure. Intolerable side effects were believed to develop within a few

months of drug prescription. Except ABC-HSS, no mortality was

considered for any other side effect. Assumptions of side effect rates

were based on Singapore HIV patient data and clinician inputs.

Intolerable side effects after treatment were estimated to develop after

30, 60 and 90 days of drug prescription for Zidovudin, Abacavir and

53

Tenofovir respectively with patients being immediately switched to the

alternative treatment with the appropriate decrease in quality-of-life.

Attributed to the rigorous follow-ups for HIV patients in Singapore,

patients are expected to be diagnosed with side effects and switched to

alternative even before they experience any substantial health

complication. The major side effect considered were renal dysfunction

and anemia for TDF and AZT respectively, which would add two

additional diagnostic medical tests for these health conditions.

Figure 1. Possible treatment therapies considered for the HIV patient subgroups28

The HIV patients contra-indicated to Tenofovir are considered to be prescribed on only one treatment regimen whereas the patients who are eligible for both Abacavir and Tenofovir can be prescribed one of two possible treatment regimen. Each regimen further considers Abacavir prescription with or without HLA-B*5701 genotyping as different strategies, which are compared for their cost and effectiveness. Abbreviations used are: ABC = Abacavir, TDF = Tenofovir, AZT = Zidovudine, Hyp. Drug = hypothetical drug

54

A sensitivity analysis was conducted to determine the robustness of the

model. The impact of individual variable variation was analyzed using a

one-way sensitivity analysis, whereas a probabilistic sensitivity analysis

was done to understand impact of simultaneous variations. Gamma and

beta distributions was assigned to costs and probability/ utility variables

respectively, except for the probability of other side effects by Abacavir,

progression of early stage HIV to late stage and duration of mortality

after ABC-HSS, which were assigned triangular distributions.

3.2.2 Model Inputs

All inputs variables and sensitivity ranges are shown in Table 1.

Treatment duration

Referencing a Singapore study for the age at diagnoses for early and

late stage HIV patients from 1996 to 2009 and with the help of HIV

statistics data from ministry of health, Singapore, the average age for a

patient diagnosed with early/ late stage HIV can be estimated to be ~40

years / 50 years respectively83,84. Benchmarking with published studies

reporting average life expectancy86, the treatment duration was

calculated to be 30 years and 10 years for early and late stage patients

respectively.

55

Figure 2. Decision tree model for newly diagnosed HIV patients in Singapore who are contra-indicated to Tenofovir28

HIV patients contra-indicated to Tenofovir were modelled for two possible side effects: 1) No HLA-B*5701 genetic testing and assigning Abacavir-based ART to everyone; 2) HLA-B*5701 genotyping and assigning Abacavir-based ART to test negative and Zidovudine-based ART to test positive patients. Three possible outcomes were assigned accordingly: 1) Development of intolerable side effects which leads to treatment switching. Patients on Abacavir based ART are switched to Zidovudine based ART followed by hypothetical drug in case of side effects; 2) No intolerable side effects in which case long term treatment is continued for the entire life tenure; 3) Development of Abacavir induced hypersensitivity which in turn has two possible outcomes – mortality or treatment switch. Abbreviations- ABC: Abacavir, TDF: Tenofovir, AZT: Zidovudine, SE: Side effects: Hypo. Drug: Hypothetical drug, ART: Anti-retroviral treatment, ABC-HSR: Abacavir hypersensitivity syndrome

56

Figure 3. Decision tree model of the treatment strategy for newly diagnosed HIV patients in Singapore who can be prescribed Tenofovir or Abacavir28

HIV patients who can be prescribed ABC or TDF were modelled for three possible strategies: 1) HLA-B*5701 genetic testing and TDF-based ART to test positive and ABC based ART to test negative 2) No HLA-B*5701 genetic testing and ABC-based ART to everyone; 3) TDF-based ART to everyone. Three possible outcomes were

57

assigned accordingly: 1) Development of intolerable side effects which leads to treatment switching; 2) No intolerable side effects in which case long term treatment is continued for the entire life tenure; 3) Diagnosis of Abacavir induced hypersensitivity which in turn can lead to – mortality or treatment switch. Also in 3rd strategy, intolerable side effect can be followed by Abacavir prescription with/ without genotyping. ABC and TDF based ART are the potential first or second line option followed by AZT-based ART and hypothetical drug being the last choice. Abbreviations - ABC: Abacavir, TDF: Tenofovir, AZT: Zidovudine, SE: Side effects: Hypo. Drug: Hypothetical drug, ART: Anti-retroviral treatment, ABC-HSS: Abacavir hypersensitivity syndrome

PPV of HLA-B*5701 genotyping

PREDICT-1 clinical trial reports HLA-B*5701 genetic screening test to

have 45.5% sensitivity and 97.6% specificity and a positive predictive

value (PPV) of 61.2% (46.2–74.8%) and Negative Predictive Value

(NPV) of 95.5% (93.3-96.7%) for clinically suspected cases19. The

cohort consisted of predominantly white population but had representing

patients from other races (African/ Arabic/ others) as well. Though the

HLA-B*5701 genetic screening test is expected to show similar

sensitivity and specificity in Singapore population but the lower allele

frequency and hence lower ABC-HSS incidence rates in Asian than

white population might lead to slightly different PPV and NPV values for

the HLA-B*5701 genetic screening. However, due to lack of availability

of local side effect data, the values reported by Predict-1 trial have been

considered for the study with the impact of variable uncertainty being

evaluated using sensitivity analysis.

58

Cost

The cost for concerned drugs and medical tests were taken from the

National University hospital (NUH) pharmacy and facilities. The

hypothetical drug cost was a weighted average of the several commonly

used alternative drug combinations in NUH. As Singapore has very few

cases of ABC-HSS being diagnosed and no fatal cases has been

reported in the near past, an approximate cost data for ABC-HSS cases

reported in a similar study in US27 has been considered. Every

intolerable side effect is expected to increase 3 consultation visits on an

average for the patient for initial diagnosis and follow-ups. The baseline

costs similar in all strategies (like regular medical checkup costs etc.)

have been ignored and only the cost varying across treatment strategies

have been considered. For ease of exposition all the cost values have

been converted to equivalent cost in US dollars using the exchange rate

of 1.26 SGD to 1 USD in January 2014.

Utility/ Quality of life

Published quality of life (QoL) values have been taken for early and late

stage HIV patients87. The decrements in QoL due to ABC-HSS has been

taken as 0.08 for 3 days for mild HSS88, 0.15 for 7 days for severe HSS89,

0.36 for 15 days for fatal HSS90 (Figure 4). Weighted average decrement

in QoL by ABC-HSS severity has been used for the analysis. Any

intolerable side effect due to TDF or AZT was assumed to cause a

quality-of-life decrement equivalent to mild ABC-HSS. An average early

59

stage HIV patient on has been considered to progress to late stage HIV

after 25 years with the related decrease in quality of life. No gradual

decrement in QoL due to ageing or long-term adherence to the treatment

or any other factor has been considered.

Probabilities

The probabilities of development of intolerable side effects have been

taken as per Singapore HIV data. The side effect rate has been assumed

to be independent of the line of therapy and patient disease stage. Also,

Figure 4. The decrement in utility values due to occurrence of side effect or ABC-HSS28

Perfect health has a utility value of 1. The drop-in utility due to HIV disease stage is throughout the life tenure. However, side effects and ABC-HSS cause a temporary drop in utility which goes back to initial after the SE is successfully treated.

60

only the intolerable side effects occurring in a span of 1-3 months after

drug prescription have been considered. Patients are assumed to be

adherent to the treatment. Although Singapore has not reported any

ABC-HSS related death in the recent past, a 0.7% mortality rate among

ABC-HSS cases has been assumed based on a medical review study91.

Table 1. Model Inputs

Variable Base case Sensitivity

Range Source

Costs (USD)

ABC + lamivudine (monthly) 174 63.2-347.6

NUH pharmacy

TDF + lamivudine (monthly) 296 102-592

AZT + lamivudine (monthly) 132 79-264

EFV (monthly) 79 55-158

Hypothetical drug (monthly) 408 180-824

Clinician consultation 85 42.5-170

HLA-B*5701 genetic test 277 139-554

ABC-HSS cases treatment 1580 790- 3160

Other Intolerable side effects on ABC 32 16-64

Fatal ABC-HSS 31,600 15,800-63,200

Schackman et al.82

TDF intolerable side effects 66 33-132

AZT intolerable side effects 32 16-64

Renal panel & urine analysis 33 16.5-66 NUH

Probabilities

Severity of ABC-HSS

Mild 0.585 Yeni et al.92

Severe, nonfatal 0.408

Fatal 0.007

Yeni et al.92

Hetherington et al.91

Intolerable SE on Tenofovir 6% 3-12% Singapore public hospital data

Intolerable SE on Abacavir 0% 0-5%

Intolerable SE on Zidovudin 24% 12-48%

Mortality among ABC-HSS 0.70% 0-1.4% Hetherington et al.91

HLA-B*5701 genotyping

61

Population frequency

Chinese 1.10% 0.55-2.2% Singapore Genome Variation Project93,94

Malays 1.80% 0.9-3.6%

Indians 6.30% 3.2-12.6%

PPV 61.20% 46.2-74.8% Mallal et al.19

NPV 95.50% 93.3-96.7%

QoL/ utility

Early stage HIV patient 0.781 0.616-0.946 Kauf TL et al.87

Late stage HIV/ AIDS patient 0.746 0.572-0.92

QoL decrements due to side effects

Mild HSS 0.08

(for 3 days) Dodek et al.88

Severe HSS 0.15

(for 7 days) Pepper et al.89

Fatal HSS 0.36

(for 15 days) 2.7 – 10.8 Freedberg et al.90

Avg. QoL decrement for ABC HSS cases (except fatal cases) 0.57 0.24 – 1.05

Other side effects 0.08

(for 3 days) 0 – 0.48 Similar to mild HSS

Abbreviations - ABC: Abacavir, TDF: Tenofovir, AZT: Zidovudine, SE: Side effects: Hypo. Drug: Hypothetical drug, ART: Anti-retroviral treatment, ABC-HSS: Abacavir hypersensitivity, NPV: Negative predictive value, PPV: Positive predictive value

3.3 Results

Based on the commonly cited Incremental cost effectiveness ratio

(ICER) threshold of USD 50,000, HLA-B*5701 genotyping before

Abacavir prescription was not found to be cost-effective for

Singaporean Chinese and Malay HIV patients in both patient

segments irrespective of disease stage. Among Singaporean Indians,

it was found to be cost-effective for early stage HIV patients contra-

indicated to TDF, but not cost-effective for all other segments (Table

2, Table 3). Considering Abacavir-based ART without pre HLA-B*5701

genetic screening (the cheapest treatment strategy) as 1st line, a

comparison with the current prevalent practice in Singapore (TDF-based

62

ART as 1st line followed with ABC-based ART) was made for the early stage

patients who can be prescribed both ABC or TDF. The current practice was

found to be not cost-effective with an ICER of USD 3-6x106/QALY as

compared to the cheapest strategy. Similar results were found for late stage

patients as well (Table 4).

3.3.1 Sensitivity Analysis

Among HIV patients who can be prescribed both ABC and TDF, ABC-

HSS mortality rate was found to be significant for determination of cost-

effectiveness (Figure 5). Among Chinese patients, a mortality rate of

greater than 6.4% for early stage and 13.9% for late stage patient

renders genotyping cost-effective. Similar figures for Malay and Indian

patients were 3.2% / 9.3% and 4.9%/ 3.3% respectively for early/ late

stage patients. For patients who are contra-indicated to Tenofovir, the

mortality rate was again an important parameter with the values being

3.7%/ 10.9% for Chinese, 2.3% / 6.5% for Malay and 0.67% / 1.6% for

Indian early/ late stage patients.

To generalize the results across ethnicities in Singapore, the collective

impact of the two population specific variables - PPV and HLA-B*5701

frequency, were analyzed. Genotyping was found not to be cost-

effective below an allele frequency of 3% for early stage patients and 6%

for late stage patients at any PPV value, irrespective of their contra-

indication to Tenofovir (Figure 6).

63

The probabilistic sensitivity analysis reveals that at USD 50,000/QALY,

genotyping is cost-effective in ~44% of the iterations for Indian HIV

patients contra-indicated to TDF. However, for all the other ethnicities

and patient groups, ABC- based ART as first line with prescription

without HLA-B*5701 genotyping is the most cost-effective strategy for

majority of iterations (Figure 7).

Table 2. Cost effectiveness of HIV treatment strategies for newly diagnosed early and late stage HIV patients contra-indicated to Tenofovir

Strategy Cost Costs QALY's

QALY's

ICER (USD/ QALY)

EARLY-STAGE PATIENTS

Chinese

No Genetic Testing 91167.1 23.2466

HLA-B*5701 Testing 91444.38 277.28 23.2477 0.0011 2,52,350

Malays

No Genetic Testing 91179.42 23.2460

HLA-B*5701 Testing 91457.2 277.78 23.2478 0.0018 1,54,490

Indians

No Genetic Testing 91258.65 23.2418

HLA-B*5701 Testing 91539.63 280.98 23.2481 0.0063 44,649

LATE-STAGE PATIENTS

Chinese

No Genetic Testing 30,449 7.457286

HLA-B*5701 Testing 30,718 269 7.457641 0.0004 757,270

Malays

No Genetic Testing 30,458 7.457076

HLA-B*5701 Testing 30,722 264 7.457656 0.0006 454,223

Indians

No Genetic Testing 30,515 7.455724

HLA-B*5701 Testing 30,746 232 7.457757 0.0020 114,068

64

Table 3. Cost-effectiveness of HIV treatment strategies for newly-diagnosed early and late-stage HIV patients receptive to both Abacavir and Tenofovir

Strategy Cost Costs QALY's

QALY's

ICER (USD/ QALY)

EARLY-STAGE PATIENTS

Chinese

ABC as 1st line without Genetic Testing

93,266 23.247

ABC as 1st line with Genetic Testing

93,723 457 23.248 0.0011 415,845

TDF as 1st line (Genetic test before ABC)

134,255 40,531 23.255 0.0068 5,965,066

TDF as 1st line (No Genetic test done before ABC)

134,295 40 23.255 -0.0001 -647,470

Malays

ABC as 1st line without Genetic Testing

93,441 23.246

ABC as 1st line with Genetic Testing

94,013 572 23.248 0.0018 318,029

TDF as 1st line (Genetic test before ABC)

134,256 40,243 23.255 0.0067 5,964,794

TDF as 1st line (No Genetic test done before ABC)

134,295 40 23.254 -0.0001 -382,561

Indians

ABC as 1st line without Genetic Testing

94,566 23.242

ABC as 1st line with Genetic Testing

95,877 1311 23.248 0.0063 208,231

TDF as 1st line (Genetic test before ABC)

134,263 38,387 23.255 0.0064 5,962,507

TDF as 1st line (No Genetic test done before ABC)

134,300 37 23.254 -0.0004 -99,191

65

LATE-STAGE PATIENTS

Chinese

ABC as 1st line without Genetic Testing

31,143 7.457

ABC as 1st line with Genetic Testing

31,472 329 7.458 0.0004 926,938

TDF as 1st line (Genetic test before ABC)

44,748 13,275 7.460 0.0022 6,162,560

TDF as 1st line (No Genetic test done before ABC)

44,764 13,291 7.460 0.0022 6,120,953

Malays

ABC as 1st line without Genetic Testing

31,205 7.457

ABC as 1st line with Genetic Testing

31,568 363 7.458 0.0006 624,297

TDF as 1st line (Genetic test before ABC)

44,748 13,180 7.460 0.0021 6,200,514

TDF as 1st line (No Genetic test done before ABC)

44,764 13,196 7.460 0.0022 6,120,100

Indians

ABC as 1st line without Genetic Testing

31,607 7.456

ABC as 1st line with Genetic Testing

32,186 579 7.458 0.0020 284,598

TDF as 1st line (Genetic test before ABC)

44,752 12,565 7.460 0.0019 6,471,208

TDF as 1st line (No Genetic test done before ABC)

44,765 12,579 7.460 0.0021 6,114,310

66

Table 4. Cost-effectiveness of HIV treatment strategies- Tenofovir as first line (current practice) with ABC as first line (cheapest strategy) for newly-diagnosed HIV patients receptive to both Abacavir and Tenofovir

Strategy Cost Costs QALY's

QALY's

ICER (USD/ QALY)

EARLY-STAGE PATIENTS

Chinese

ABC as 1st line without Genetic Screening

93,266 23.24663

TDF as 1st line (No genetic test before ABC if SE develops)

134,295 41,028 23.25446 0.0078 5,237,899

Malays

ABC as 1st line without Genetic Screening

93,441 23.24598

TDF as 1st line (No genetic test before ABC if SE develops)

134,295 40,854 23.25442 0.0084 4,839,405

Indians

ABC as 1st line without Genetic Screening

94,566 23.24181

TDF as 1st line (No genetic test before ABC if SE develops)

134,300 39,734 23.25418 0.0124 3,213,980

LATE-STAGE PATIENTS

Chinese

ABC as 1st line without Genetic Screening

31,607 7.455733

TDF as 1st line (No genetic test before ABC if SE develops)

44,752 13,144 7.459710 0.0040 3,305,059

Malays

ABC as 1st line without Genetic Screening

31,205 7.457082

TDF as 1st line (No genetic test before ABC if SE develops)

44,748 13,543 7.459789 0.0027 5,002,848

Indians

ABC as 1st line without Genetic Screening

31,143 7.457292

67

TDF as 1st line (No genetic test before ABC if SE develops)

44,748 13,605 7.459801 0.0025 5,422,356

QALY: Quality adjusted life-years; ICER: Incremental cost-effectiveness ratio (USD/QALY); ABC = Abacavir; TDF = Tenofovir

Figure 5. Sensitivity analysis for early stage Chinese patients receptive to both Abacavir and Tenofovir28

One-way sensitivity analysis (Tornado analysis) of independent variables evaluates the influence of their uncertainty on incremental cost-effectiveness ratio (ICER) of the HLA-B*5701 genotyping strategy in comparison to baseline strategy (Abacavir as 1st line without genotyping). The variables dependent on other variable(s) are not shown. Minus sign at the left side of the bar indicate ICER decrease when variable increases. Abbreviations used are: ABC = Abacavir, TDF = Tenofovir, AZT = Zidovudine, 3TC = lamivudine, SE = side effects, ABC-HSS = Abacavir hypersensitivity syndrome.

68

3.4 Discussion

HLA-B*5701 genetic screening prior to Abacavir prescription though

significantly reduces the incidence of ABC-HSS; the cost-effectiveness

of genotyping varies across ethnicities and healthcare setups. In

Singapore healthcare setup, among early stage patients, genotyping has

been found cost-effective only for patients of Indian ethnicity who are

Figure 6. Sensitivity analysis of cost-effectiveness to variation in PPV and HLA-B*5701 frequency in early stage HIV patients28

(A) Two-way sensitivity analysis to analyze the combined effect of positive prediction value (PPV) of the HLA-B*5701 genotyping and population HLA-B*5701 frequency on the incremental cost-effectiveness ratio (ICER) of the genotyping strategy. Only the cost-effective strategy is shown in the figure. Genetic screening prior to Abacavir prescription is found to be not cost-effective for ethnicities with allele frequency lower than 3%. For allele frequencies higher than 3%, a very high PPV value (>85%) is required to render genotyping cost-effective (B) One-way sensitivity analysis of the influence of positive prediction value (PPV) of HLA-B*5701 genetic screening on the Incremental cost-effectiveness ratio (ICER) of the genotyping strategy, for an early-stage HIV patient receptive to both Abacavir and Tenofovir. A strategy is considered cost-effective for an ICER≤ $50,000/QALY. For this patient group, HLA-B*5701 genotyping before Abacavir prescription was found to be not cost-effective for Chinese and Malay patients. For Indian patients, a PPV > 91.5% was required to achieve cost-effectiveness. Abbreviations used are: ABC = Abacavir, TDF = Tenofovir

69

contra-indicated to TDF. This can be attributed to the high allele

frequency among Indians, which increases the risk of ABC-HSS and

the long-time treatment among early stage patient where avoiding ABC-

HSS saves many QALYs. Among other ethnicity patients, though the

QALY’s saved are similar, the low prevalence of the risk allele lowers the

overall benefit of genotyping. A high cost of alternative drug (TDF) for

early stage patients who are receptive for both ABC and TDF mitigates

the benefits of ABC-HSS prevention by genotyping, rendering the

Figure 7. Cost effectiveness acceptability curve for early-stage Chinese HIV patient receptive to both Abacavir and Tenofovir28

Cost effectiveness acceptability curves from probabilistic sensitivity analysis (PSA) represent the number of iterations for which the genotyping strategy is cost-effective for various willingness to pay values. For early stage HIV patients of Chinese ethnicity, Abacavir without genetic screening was found to be cost-effective for 91% of the iterations, whereas Abacavir with genetic screening was not found to be cost-effective at all (0% iterations). Abbreviations used are: ABC = Abacavir, TDF = Tenofovir, SE = Side effect

70

strategy not-cost-effective for all. Among the late stage HIV patients, as

the life expectancy is low, the overall benefit of preventing ABC-HSS

further diminishes, making it not cost-effective for all late stage patients

irrespective of ethnicity or TDF contra-indication. Thus, the study does

not recommend the policy makers to subsidize HLA-B*5710 test for HIV

patients in Singapore except for Indian HIV patients.

The present study has a few limitations. The study assumes no co-

infections or other death causes among HIV patients as modelling the

complexity of treatment and disease outcomes in such cases was

beyond the scope of this study. We assume perfect patient compliance

leading to absolute treatment compatibility throughout the modelled

duration and that no drug resistance to develop on long-term treatment.

With limited Abacavir usage and almost no ABC-HSS related mortality

observed in Singapore, relevant local data was unavailable, and

literature based assumptions have been made. All drugs are assumed

to have similar efficacy.

With limited government subsidies for HIV drugs in Singapore and an

expensive life-long treatment, costs is of extreme importance to the

patients and their families. Abacavir - a cheap, efficacious and safe

alternative for majority of Singaporean patients can immensely help to

achieve considerable cost-savings for the patient and their families. As

currently the possibility of a side effect and the FDA recommendation of

an expensive HLA-B*5701 screening before Abacavir raises many

71

questions in the minds of clinicians and patients alike, this study

highlights that unlike Caucasians, the risk of ABC-HSS is very low for

two of the three ethnicities in Singapore, and that the expensive

genotyping test is not cost-effective in Singapore, except for a subset of

early stage Indian patients who are contra-indicated to Tenofovir. In

addition, Singapore’s excellent healthcare system further mitigates the

benefits of prior genotyping by providing a strong framework for quick

diagnosis. Thus, except for early stage Indian HIV patients, Abacavir as

a first-line without genotyping appears to be the most cost-effective

strategy for all other HIV patients in Singapore.

72

Chapter 4 Cost-Effectiveness Analysis for including a Serum-

microRNA Biomarker Panel as a Screen before

Endoscopy, for Detection of Gastric Cancer among

High Risk Patients in Singapore

4.1 Introduction

Gastric Cancer is among the top four cancers by prevalence and the

third leading cause of death worldwide. Starting from the inner lining of

the stomach, with no symptoms in the early stages, the cancer silently

progresses across the stomach walls and/or to the other organs.

However, with advancement in medical science, highly efficacious

treatments have been developed for treatment of early stage gastric

cancer, late presentation in the clinic due to delayed manifestation of

symptoms and limited treatment success in advanced stages is the

biggest challenge in its successful treatment.

The world witnessed almost 1 million gastric cancer cases in 2012 with

0.7 million deaths every year due to this malignancy95. Although both

genders have distressing incidence rates, males are at double the risk

than females. Highest incidence of gastric cancer is reported in Eastern

Asia, Latin America and the Caribbean with majority of cases globally

happening in China alone (42%). The Republic of Korea had the highest

risk of gastric cancer in 2012, followed by Mongolia and Japan with an

age-standardized incidence rate of 41.8, 32.5 and 29.9 per 100,000

73

respectively. The countries with the lowest risks are in Africa and North

America95. Gastric cancer is prevalent in mostly elderly patients, with the

median age at presentation found to be greater than 65 years for majority

of the countries96-100

Gastric cancer overall has a very poor survival rate and currently account

for ~9% of all cancer death worldwide101. The poor prognosis is mostly

attributed to its late presentation in the clinics when the tumor has

already locally advanced or metastasized. The survival rates are

influenced greatly by the stage at diagnosis with the 5-year survival rates

varying from 70-90% in stage 1 to 5-10% for stage 4 cancers102-104.

However due to late presentation, most of the patients diagnosed die

within 1- 2 years of treatment.

In western countries about two-third of the gastric cancer patients are

diagnosed with locally advanced or metastatic cancer with a median

survival rate of 10 months105. The 5-year relative survival for diagnosed

patients is reported to fall in the range of 15-32% in UK106 , Scotland107

and US108. However, Korea and Japan with an effective screening

program have achieved 5-year survival as high as 60-65% with more

than half of its patients diagnosed in early stages109,110. Both countries

recommended national level screening for adults ≥40 years, annually in

Japan and biennially in Korea using upper-gastrointestinal series (UGIS)

or endoscopy. These screening programs have been proven cost-

74

effective and have resulted in significant health benefits in these high-

incidence countries2,103.

Endoscopy and upper-gastrointestinal series (UGIS) are the two major

modalities for gastric cancer screening. In endoscopy, a thin flexible

lighted tube with a small video camera on the end is passed into the

stomach through the throat and helps in direct visualization of the gastric

mucosa. In the case of abnormal areas, samples can be collected using

the endoscope and sent for biopsy. Endoscopic ultrasound using a small

transducer attached on the endoscope tip helps doctors take a closer

look at the layers of the stomach wall and nearby lymph nodes to study

the suspicious area more closely and obtain biopsy samples. However,

UGIS is an x-ray technique to look at the inner lining of the esophagus,

stomach and first part of the small intestine. It includes the patient

drinking a barium solution, which lines the stomach, esophagus and

small intestine. As x-rays cannot pass through the barium lining, the

images obtained would outline any abnormalities of the lining of these

organs. A double-contrast technique, which involves pumping air using

a thin tube passed into the stomach, makes barium coating very thin to

help in diagnosis of even small abnormalities common in early gastric

cancer. Although, both these techniques are commonly used for

screening, endoscopy is the gold standard. It has good accuracy and is

also the follow up diagnosis test for suspicious cancer cases identified

through UGIS. But because of the invasive procedure and high cost of

endoscope, there are concerns about patent willingness limiting its

75

outreach as a screening tool. A compliance rate as low as ~30% has

been observed for endoscopic screening in national screening programs

even in high incidence country like Korea111.

4.1.1 Gastric Cancer statistics in Singapore

Singapore being a multi-racial country comprises of Chinese, Malay and

Indian ethnicities as its majority of residents. With a 5.4 million

population, Singapore witnesses an overall gastric cancer incidence rate

of 10 per 100,000 residents. However, the risk is seen to rise sharply

above the age of 50 years, with an incidence rate as high as 60 to 160

per 100,000 for residents aged 65 to 75 years (Figure 8). Chinese, which

comprise ~ 75% of Singapore’s population, are at the highest risk among

the ethnicities, accounting for 90% of the total gastric cancer burden of

the country. Basic Singapore-specific gastric cancer statistics are

presented in Table 5.

Table 5. Gastric Cancer Statistics for Singapore (Singapore Cancer Registry Committee 2015)112

Incidence Data

(2008-2012)

Males Females Total

ASR RR ASR RR ASR RR

All residents

11.9 - 7.1 - 9.2 -

Chinese 13.3 1.0 7.7 1.0 10.2 1.0

Malay 4.5 0.4

(0.3-0.5) 4.0

0.5 (0.4-0.7)

4.3 0.4

(0.3-0.5)

Indian 8.3 0.6

(0.5-0.8) 3.9

0.5 (0.4-0.7)

6.1 0.6

(0.5-0.7)

ASR: age standardized incidence rate; RR: relative risk

76

Although the 5-year survival rates for gastric cancer in Singapore is

among the best in the world (5-year survival by stages 1:2:3:4::

90%:70%:40%:10%), the overall survival among cancer patients

continues to be poor due to the diagnosis of patients at advanced stages.

With 320 deaths every year, gastric cancer is among the top five causes

of cancer associated deaths98, raising concerns about the need for

strategies to encourage early diagnosis.

Figure 8. Age Specific Incidence rate of Gastric Cancer in Singapore (2008-2012)

(Singapore Cancer Registry Committee 2015)98

Although population level screening in Korea and Japan has led to

significant health benefits with proven cost-effectiveness, its replication

in Singapore with a low to intermediate gastric cancer risk profile may

appear to be unnecessary and an additional cost burden. With half of the

population >40 years, 5500-6000 persons would need to be screened to

77

identify just one case of gastric cancer creating immense economic

burden and enormous stress on the healthcare service setup. Thus, with

an aim to achieve early diagnosis, currently clinicians in Singapore

recommend endoscopic screening to the patients presenting in the

specialist/ hospital clinics with any form of persistent gastric distress

especially for Chinese patients.

Even though endoscopic screening in this high-risk patient group has

improved the cancer diagnosis in Singapore, there are many challenges

yet to overcome. Firstly, this still leads to an immense cost burden as

the absolute incidence remains low and many negative endoscopies are

being done. Although the cancer incidence in the hospital-clinic patient

group for Singaporean Chinese patients is more than 30 times higher

than the population incidence overall (0.7% vs. 0.01% respectively), the

absolute value of incidence remains low, implying that majority of

endoscopies are still being done on non-cancer cases. Currently only 1

in 150 patients being screened in this high-risk group are diagnosed with

cancer which leads to an estimated expenditure of ~ SGD100,000 on

endoscopy alone to identify just one gastric cancer case. Secondly, most

of the cancers diagnosed in this target group are already at advanced

stages. About >80% of cancer cases diagnosed in hospital-clinics are

already stage 2 or higher with ~20% of cases being terminal cancers.

With most of the patients presenting in hospital-clinics with some

symptoms and the poor outcomes associated with late stage cancers,

the potential benefits from the abovementioned intervention are

78

fundamentally limited. Thirdly, endoscopy is an invasive and costly

procedure limiting patient’s willingness and adherence for follow up

procedures. Even though local studies have proved cost-effectiveness

of endoscopic screening in intermediate risk population group in

Singapore, the implementation has been restricted, potentially due to

budgetary impacts and concerns about patients’ willingness.

With these challenges, scientists are focusing on cheaper and more

efficient screening methods to achieve patient risk stratification cost-

effectively and in a patient-friendly manner. There have been multiple

efforts with methodologies like nano-array analysis of breath

composition for VOC’s (volatile organic compounds)113 and microarray

analysis of microRNA (miRNA) samples in blood, to name a few to

achieve this aforementioned goal.

miRNA are small non-coding RNAs that regulate gene-expression post-

transcriptionally. Altered miRNA expressions are suspected in the

pathogenesis of many diseases including cancers with research

correlating their presence in body fluids (serum, plasma etc) with disease

onset and progression. The scientists in Singapore have successfully

developed and validated a miRNA panel specific to gastric cancer, which

can be utilized as a minimally invasive biomarker panel for detection of

gastric cancer. Using a case-cohort study model, the novel miRNA panel

has been formulated in a large Singapore Chinese study cohort and

validated blindly in two independent cohorts: a Korean and a Singapore

79

cohort. With favorable results, a novel microRNA biomarker panel has

been designed, which would help to stratify patients based on their

cancer risks and achieve an efficiently targeted population for

endoscopic screening, reducing the false endoscopies without

compromising substantially on the diagnosis accuracy. In addition, this

simple blood test with the merit of its non-invasiveness is believed to

improve the follow-up adherence and further enhance the overall

diagnosis accuracy.

Thus, this study aims to analyze the cost-effectiveness of implementing

the newly developed miRNA biomarker panel as a screen before

endoscopy in the current framework of Singapore’s hospital-clinic setting

and evaluate the potential cost-savings. Further, with an aim to enhance

gastric cancer diagnosis strategies through mass screenings, we also

evaluate the performance of translating miRNA panel for population

screening of intermediate risk population cohort in Singapore (Chinese

male, age 50-69 years) and analyze benefits achieved through an

enhanced coverage and its cost-implications. Furthermore, we also

explore the cost-effectiveness of translating miRNA-based strategy in

high-risk country setup by replicating the analysis for existing national

screening program for gastric cancer in Korea.

80

4.2 Methods

4.2.1 Target Population:

The study analyses the translation of miRNA blood test for gastric cancer

screening in the following scenarios:

Patients at specialist hospital-clinic in Singapore: This subgroup consists

of patients who visit the hospital clinic with gastric distress and are

screened using endoscopy as per the current practice. The annual

incidence of gastric cancer in this group is >30 times the population

incidence98, making it a favorable high-risk group for screening.

Singaporean Chinese aged 50-69 years: Chinese ethnicity carries 90%

of gastric cancer disease burden in Singapore with males at a 30%

higher risk of gastric cancer than females112. With the cancer incidence

rising sharply after the age of 50 years, this subgroup with intermediate

gastric cancer risk has a 4 times higher annual incidence rate than the

general population98.

National screening program for gastric cancer in Korea: With gastric

cancer among the top 2 most prevalent cancers in Korea114, currently

screening is recommended for adults ≥40 years under the national

screening program for cancer.

81

Figure 9. Markov-Decision Tree Model for the strategy 'endoscopy for all'

The tree structure divides for the distribution of patients by stages, accounting for true positives and false negative test outcomes among the patients and false positive and true negative among the healthy people. The disease progression among the missed cancer cases are evaluated specific to the stage, with all missed stage 1 cancers expected to be diagnosed after 3 years as stage 3 or stage 4 cancers and all missed stage 2,3,4 cancers diagnosed later as stage 4. Patients who were non-compliant are expected to follow a similar disease journey as the missed cancer patients. Abbreviations used are: TP: True Positive, FP; False positive, TN: True Negatives, FN: False Negatives

82

Figure 10. Decision Tree Model for Strategy 'miRNA screening with endoscopy only for test positive patients' and for the current strategy of No-Screening for the population cohort of Singaporean Chinese 50-69 years

83

4.2.2 Strategies Compared

The two strategies compared are:

1. Screen for cancer using the miRNA test, followed by upper-endoscopy

for test-positive patients

2. Current clinical practice of upper-endoscopy for all

As Singapore does not have any national cancer-screening program for

the population, the analysis for the population screening for Chinese

males (50 – 69 years) involves comparison with no screening as the

current practice.

4.2.3 Methodology

A Markov decision model was built in TreeAgePro2017 to compare the

two strategies and assess their cost-effectiveness in the two

aforementioned scenarios (Figure 9, Figure 10). The model was

populated using local and published data with any required technical

assumptions drawn with a conservative bias i.e. in favor of current

practice, universal endoscopy. With a healthcare system perspective,

model was built with a lifetime horizon accounting for the remaining life

expectancy of each population subgroup. As mean patient age in

hospital clinic setting (discovery cohort) is 68 years, model was built for

14 years (average life expectancy: 82 years)115 and 20 years for

population screening cohort (mean age 60 years, life expectancy of

males: 80 years)115. Patients were modelled in five progressive health

84

states – healthy (cancer-free), TNM Stage 1, TNM Stage 2, TNM Stage

3 and untreatable terminal (Stage 4). Early or advanced stage patients

(stage 1, 2, and 3) received curative treatment with a stage specific

recurrence probability after a mean duration of 2 years116,117, while

terminal cancer patients (stage 4) received only palliative care with a

conditional life expectancy of 1 year116,117. As prognosis of the cancer

recurrence is poor, patients diagnosed with recurrence were assumed

untreatable (equivalent to stage 4) and were given palliative care.

The patients not diagnosed with gastric cancer would include both

healthy and missed cancer cases. The model admits the possibility of

missed cancer cases at early as well as advanced stages in both the

strategies as per the relevant sensitivity. The healthy cases are expected

to remain healthy for the complete duration of the analysis, while the

missed cancer patients were expected to experience the consequences

of treatment delays – disease progression, impact on cost and quality-

of-life and an increased mortality, as the cancer would progress in them

undiagnosed and untreated. Missed stage1 patients are considered to

progress to advanced-stages in an average of 3 - 4 years as per the

natural course of progression of gastric cancer118 at which they are

assumed to be diagnosed due to presentation in the clinics with

symptoms (by endoscopy). With a conservative assumption, we have

assumed all the missed stage 1 cancers to progress to stage 3 or 4

cancer in 3 years (in ratio observed at hospital clinics) and get

diagnosed. After diagnosis, the appropriate treatment is started with

85

possibility of potential recurrence for stage 3 cancers as discussed

above. Patients initially missed at stage 2 & 3 are believed to be

diagnosed after they have advance to stage 4 in 12 months. They are

expected to return to the clinic with symptoms where they are diagnosed

by endoscopy and treated with palliative care. Missed stage 4 patients

are experienced to be diagnosed after a mean time of 2 months followed

by palliative treatment, with their life expectancy reduced to half due to

initial misdiagnosis. No other causes of mortality are considered.

The hospital clinic scenario expects 100% compliance among the

patients for both the strategies. On the contrary, as the general

population is not always fully compliant, an additional compliance rate

among the population-screening cohort (Chinese males 50-69 years),

has been considered to account for the unwillingness for screening

among the population. In the base-case, the compliance for endoscopy

has been assumed to be 45%, based on the overall screening

participation rate in Korean national programs. miRNA test was

expected to improve the compliance by 50% in comparison to

endoscopy due to its lower costs and minimally invasive testing

procedure. However, with a miRNA test positive, 100% compliance is

assumed for the follow-up endoscopy. Performance across a range of

compliance rates has been evaluated. All costs quoted in US dollars

have been calculated based on the exchange rate of 1.41 SGD to 1 USD

as per exchange rates in March 2017.

86

4.2.4 Model Inputs

Treatment Protocols for Cancer Treatment and Related Costs

Stage-specific treatment protocols and average medical expenditures

for gastric cancer were obtained from the National University Hospital

and expert opinions of clinicians based on current practices in Singapore

(Table 6). Patients diagnosed with gastric cancer undergo staging

investigation, which includes Computerized Tomography (CT), Chest X-

Ray (CX-R), Endoscopic Ultrasound (EUR) and a specialist consultation

(including the cost of nurse counseling and an estimated round-trip

transport). Curative treatment administered to stage 1, 2, and 3 cancer

patients includes surgery (total/ partial gastrectomy) and hospital stay of

12 days. Stage 3 patients undergo an additional chemo-radiotherapy (5

follow-ups) and radiotherapy sessions (5 sessions/ week for 5 weeks).

Palliative care for stage 4 patients includes bypass surgery (30%),

endoscopic stenting (6%), palliative chemotherapy-5 sessions (16%)

and conservative treatment (2x specialist visits) (48%) with an

appropriate hospital stay (12 days in surgery cases and 2.5 days for

cases with no surgery). Patients are also expected to adhere to follow

up visits (average 2.2 visits/ 5 years) and repeat CT and CXR (average

1.5per year for 5years after the diagnosis). The miRNA panel test cost

in Singapore has been assumed to be SGD100 (~ USD70), which is

~15% of endoscopy cost, an approximate estimation with the locally

prevalent cost of RNA – based diagnostic test (~SGD100-150), which

follows a testing procedure similar to the developed miRNA test.

87

However, a sensitivity analysis has been performed on the miRNA test

cost to evaluate its impact. In the hospital clinic model, a biopsy rate of

40% is assumed to probe on suspicious cases (as obtained from local

hospital) whereas in population screening model the biopsy rate is

expected to be only 15%. Total costs have been evaluated inclusive of

GST and without considering any subsidy. A discount rate of 3% has

been considered for cost and health benefits.

Quality of life values

Stage-specific EQ-5D quality-of-life (QoL) index measures were

obtained from a previous local study119 performed on Chinese gastric

cancer patients in National University Hospital, Singapore (Table 6). A

diagnosed patient is expected to be immediately started on treatment,

and experience the diagnosed stage-specific QoL for 1 year with a 6-

months additional decrease in QoL due to the initial surgery referred as

disutility120. After one year of treatment, the patient is expected to enjoy

a QoL equivalent to an asymptomatic patient (similar to stage 1 cancer)

for the remaining time until faced with any recurrence, which would

subsequently drop the QoL to the stage 4 equivalent.

Test Characteristics

Test characteristics for diagnostic endoscopy with biopsy for the

suspected cases (sensitivity: 93%, specificity: 100%) have been

Table 6. Base case values and sensitivity ranges for model inputs (Singapore)

88

Variable name Base case value Sensitivity

Range Source

Costs (SGD)

MiRNA test 100 (USD70) 0 – 300 Assumed

Upper -endoscopy (EGD) 605 (USD432) 300 – 1,210

National University Hospital, Singapore (NUH)

Biopsy 150 (USD106) 75 - 300

Bypass Surgery 4,520 2,260 – 9,040

Curative Chemotherapy

(5 sessions/week x 5

weeks)

22,432 11,216 – 44,865

Curative Surgery

(partial or total

gastrectomy)

6,227 3,115 – 12,000

Endoscopic Stenting 1,280 640 – 2,560

CT & CXR (follow up

tests) 485 242 – 970

Hospital stay (per day) 471 235 – 942

Palliative Chemotherapy

(5 sessions) 3,000 1,500 – 6,000

Specialist Consultation 104 52 – 208

Staging Investigation (EUS + CT+ CXR+ follow-up)

1,856 925 – 3,700

Probabilities

Diagnosis by Stages in

Singapore

Stage1 : 2 : 3 : 4

30% : 15% : 23% :

32%

(Hospital-Clinic)

Discovery cohort statistics

40%: 32% : 18%:

10%

(population

screening)

Calculated

Diagnosis Rate in

hospital clinic by

endoscopy (annual)

(per 100,000)

0.67% 0.3-1.4% NUH

Prevalence of

undiagnosed gastric

cancer in Chinese

Males (50-69 years)

(per 100,000)

130

89

resourced from a study evaluating diagnostic accuracy through a

retrospective study among gastric cancer patients121. Biopsy is believed

to be perfect with 100% sensitivity and specificity. The true incidence of

gastric cancer in Singapore hospital-clinics has been estimated based

on the aforementioned sensitivity values. The miRNA test characteristics

Recurrence in Stage 1

patients (early gastric

cancer)

11% 0-20%

Roukos et al.116

Recurrence in Stage 2

patients 53% 11-83%

Recurrence in Stage 3

patients 83% 53-100%

Utility Values (disutility*)

Stage 1 0.88 (0.28) 0.6–1

Zhou HJ et al.119

Stage 2 0.86 (0.29) 0.62–0.99

Stage 3 0.77 (0.31) 0.58–0.95

Stage 4 0.68 0.51–0.84

Test Characteristics

Endoscopy Sensitivity

93% 84–100% Voutilainen

et al.121

88.6% 70–97.6% Hamashima

et al122

Endoscopy Specificity 100% Voutilainen

et al.121

miRNA sensitivity 81% 70–100%

Current

Study

miRNA specificity 89% 75–100%

miRNA Sensitivity by

Stages (Stage 1:2:3:4)

63% : 75% : 89% :

93%

*Disutility refers to temporary reduction in QoL during first 6 months of treatment. Note: Assumed treatments are based on observed practice in Singapore. Gastric cancer patient on diagnosis undergoes staging investigation (CT, CXR, EUS & specialist consultation). Curative treatment includes surgery (total/ partial gastrectomy) & hospital stay (12days). Stage 3 patients undergo additional chemo-radiotherapy. Follow ups include: visits (2.2/year), repeat CT, CXR (1.4/year). Palliative care includes bypass surgery (30%), endoscopic stenting (6%), palliative chemotherapy (16%) & conservative treatment (2x specialist visits) (48%) with an appropriate hospital stay (12 days - on surgery, 2.5 days on average - if no surgery is performed). Abbreviations used: CT: Computerized Tomography; CXR: Chest X-Ray; EUS: Endoscopic Ultrasound

90

estimated from the Singapore Discovery Cohort have been considered

as the base case value in the hospital-clinic model with an overall

sensitivity and specificity of 81% and 89% respectively.

As in the population-screening model, the diagnosed gastric cancer

tends to have a different mix by stages with an increase in proportion of

early cancers, a population study from Japan reporting a sensitivity of

88% for endoscopy has been referenced122. As all suspicious cases are

followed-up with biopsy, the specificity of endoscopy (and biopsy) is

considered 100%. miRNA test stage specific sensitivity and specificity

values have been used.

Estimating population prevalence of gastric cancer in Chinese

Males 50-69 years

As the study aims to evaluate a one-time population screening in the

cohort of Chinese males, 50-69yrs in Singapore, it is essential to

calculate the population prevalence of undiagnosed gastric cancer

cases in this target group. The current annual age-specific incidence rate

is 43 cancers per 100,000 in this group98. All these cases diagnosed

currently (in absence of any screening program) are expected to have a

stage specific distribution of stage 1:2:3:4:: 30%:15%:23%:32%

equivalent to that observed in current hospital clinic setting. As all

advanced cancers diagnosed in next 2-3 years are expected to be

present as early cancers in the current year, an approximation of

91

prevalence of undiagnosed gastric cancer prevalence was calculated

based on following assumptions:

Time to progress from stage 1 gastric cancer to stage 2 is 1-2 years

and to advanced stages (stage 3,4) is 3 years

Stage 2 cancers progress to stage 3 cancers in 1-2 years and to

stage 4 in 2 years

Stage 3 cancer progresses to stage 4 cancers in 1 year

Thus, the prevalence of undiagnosed gastric cancer in the 50-69 years

Chinese male population cohort was estimated to be 0.13% with

expected stage distribution as stage 1:2:3:4 :: 40%: 32%: 18% 10%.

4.2.5 Sensitivity Analysis

Both deterministic and probabilistic sensitivity analysis (100,000

iterations) has been done to evaluate the robustness of the model. The

deterministic sensitivity analysis evaluates the impact of varying one

variable at a time in a specified range (Table 6) whereas probabilistic

analysis varies all the variables simultaneously across 100,000

iterations, with each variable value been determined by a random draw

from an assigned distribution as specified (Table 7). Cost-effectiveness

of miRNA strategy was also analyzed with varying miRNA test cost,

compliance rates and gastric cancer incidence rates.

92

Table 7. Distributions assigned to parameters in the probabilistic sensitivity analysis (Hospital-clinic)

Input Variables Type of Distribution

Mean (S.D)

Costs

miRNA test Gamma 100 (50)

Biopsy Gamma 150 (37.5)

Curative Chemotherapy Gamma 4520 (1130)

Bypass Surgery Gamma 22432 (5608)

Curative Surgery (partial or total gastrectomy)

Gamma 6227 (1481)

Upper -endoscopy (EGD) Gamma 605 (152)

Endoscopic Stenting Gamma 1280 (320)

Palliative Chemotherapy Gamma 3000 (750)

Probabilities

Incidence of gastric cancer (hospital clinic) Beta 0.72 (0.18)

Prevalence of undiagnosed gastric cancer in Chinese males (50-69 years)

Beta 0.13 (0.036)

Recurrence in Stage 1 patients (early gastric cancer)

Beta 0.11 (0.07)

Recurrence in Stage 2 patients Beta 0.53 (0.12)

Recurrence in Stage 3 patients Beta 0.83 (0.08)

Utilities

Stage 1 Beta 0.88 (0.05)

Stage 2 Beta 0.86 (0.07)

Stage 3 Beta 0.77 (0.1)

Stage 4 Beta 0.68 (0.08)

Test Characteristics

Endoscopy Sensitivity

hospital-clinic Beta 0.93 (0.03)

population –screening) Beta 0.86 (0.05)

miRNA Sensitivity

Overall Beta 0.81 (0.05)

Stage 1 Beta 0.63 (0.08)

Stage 2 Beta 0.75 (0.06)

Stage 3 Beta 0.89 (0.045)

Stage 4 Beta 0.93 (0.027)

miRNA specificity Beta 0.89 (0.04)

93

4.3 Analysis for the Korean population screening program

Analysis for the Korean population screening cohort (age ≥40 years) was

done for a time horizon of 24 years in accordance with the median

patient age (57 years)123 and life expectancy (81 years)124. Cost of

endoscopy (USD35), biopsy (USD25)125 and stage–specific treatment

cost for gastric cancer126, age specific incidence rates (130 per

100,000)97 and stage at diagnosis (stage 1:2:3:4:: 63%:14%:14%:9%)

was obtained specific to Korea from published literature109 (Table 8).

Benchmarking with the healthcare services costs in Korea, the miRNA

test cost was assumed to USD20.

Table 8. Base Case Values and Sensitivity Range for model Inputs specific to Korean healthcare setup

Variable name Base case

value Sensitivity

Range Source

Costs (USD)

MiRNA test 20 0 – 40 Assumed

Upper -endoscopy (EGD) 35 12 - 70 Lee et al.125

Biopsy 25 12 - 50

Treatment Costs (for first-year after diagnosis)

Kim et al.126

Stage 1 3318 6000 – 22,000

Stage 2 12891 6000 – 24,000

Stage 3 16464 16000 – 68000

Stage 4 31759 15000 – 60000

Specialist Consultation 4 2 – 10

Probabilities

Diagnosis by Stages

Stage1 : 2 : 3 : 4 63% : 14% : 14% : 9%

Choi et al.109

Age specific incidence rate (per

100,000) 130 0.3-1.4% Shin et al.97

94

4.4 Results

4.4.1 Hospital-clinic setting

Analyzing in a hospital- clinic setup with 100% compliance for both

strategies, the universal endoscopy was found to have a mean cost per

patient of SGD807 (SGD573 additional dollars as compared to miRNA

strategy) for an incremental health benefit of 0.0035 QALY only. This

leads to an ICER of SGD162,000/QALY (USD 116,000/QALY) for

universal endoscopy, making it unfavorable and miRNA-based strategy

(screening the patients by miRNA test followed by endoscopy only for

the test positive patients) cost-effective than universal endoscopy (Table

9). Thus, for every 800 patients screened in the hospital-clinic, miRNA-

based strategy would create a cost-savings of 0.45 million SGD (USD

0.32 million) but at expense of 1 additional missed gastric cancer case

(Table 10). With endoscopy used as the confirmatory diagnostic test

among miRNA positive patients, both the strategies have a perfect PPV

of 100%, with miRNA-based strategy having a NPV of 99.82% in

comparison of an NPV of 99.95% for universal endoscopy.

Sensitivity Analysis

Evaluating the model for sensitivity to the value of variables, none of the

variables affected the model outcomes up to willingness-to-pay (WTP)

of SGD 100,000/QALY in the hospital clinic setting for the specified

sensitivity ranges (Table 6). The strategy of endoscopy for all is not cost-

effective even at a very high incidence rate of 2000 (age-specific crude

95

incidence rate/ 100,000) across miRNA price range from SGD30-

SGD100, highlighting the huge cost-burden of this strategy on the

Singapore healthcare system (Figure 11).

Probabilistic sensitivity analysis also identifies miRNA-based strategy as

the cost-effective strategy for majority of the iterations (>80% iterations)

up to WTP of SGD 100,000/QALY (USD 70,000/QALY). 92% of

iterations support miRNA cost-effectiveness for a WTP of SGD

71,000/QALY (USD 50,700/QALY) with these numbers rising to >97%

iterations for WTP lower than SGD 50,000/QALY (WTP: USD

35000/QALY) (Figure 12).

Table 9. Cost-effectiveness results for Singapore Hospital-clinic scenario comparing the two strategies: ‘endoscopy for all’ and ‘miRNA test followed by endoscopy only for test positive patients’ (modelled for 14 years)

As endoscopy for all has an ICER >SGD71,000, it is not cost-effective, making miRNA test a cost-effective strategy for hospital clinic scenario

Strategy Cost

(SGD) Costs (SGD)

Efficiency (QALY)

Efficiency (QALY)

ICER (SGD/Q

ALY)

miRNA test followed by Endoscopy for test positive patients

234 (USD167)

8.7232

Endoscopy for all

807 (USD576)

573

8.7267 0.0035 162,497

(USD 116,069)

96

Table 10. Cost and health benefits estimation in hospital-clinic setting in Singapore

Endoscopy for all

miRNA-based strategy

Sample Cohort Size 1000 1000

Cost of testing (SGD) 807,000 234,000

Additional cost spent for endoscopy (SGD) 573,000 -

Number of cancers diagnosed* 6.7 5.4

Additional cancers detected by endoscopy 1.3 -

Expense per additional cancer detected (SGD)

450,387 (USD321,705)

-

Figure 11. Sensitivity analysis investigating the impact of gastric cancer incidence rate on cost-effectiveness of endoscopy for all at multiple miRNA test cost scenarios in the hospital-clinic setting As ‘endoscopy for all’ has a high ICER (> SGD71,000), miRNA based strategy is the cost-effective strategy for gastric cancer incidence rates from 200-2000 per 100,000 and for miRNA test cost from SGD30-100. miRNA-based strategy is the cheaper strategy in hospital-clinic scenario and is hence used as the baseline strategy

97

4.4.2 Population screening for Chinese males (50-69 years)

As currently there are no population screening programs in Singapore,

current practice of no-screening has been used as the baseline to

compare both the strategies – endoscopy for all and miRNA based

strategy (miRNA test followed by endoscopy for test positive patients).

Figure 12. Cost-effectiveness acceptability curve for hospital-clinic scenario

Cost-effectiveness acceptability curve represents the number of iterations for which the two strategies are cost-effective for various willingness-to-pay (WTP) values. miRNA-based strategy was found to be cost-effective for 92% of iterations at a WTP of SGD71,000 (1GDP per capita). WTP represents the amount of an organization is willing to pay to gain 1 QALY of health benefits and helps to develop the cost-effectiveness threshold.

98

Analyzing a one-time population screening for gastric cancer among

Singapore Chinese males (50-69 years) in comparison to no-screening,

miRNA-based strategy would lead to a mean cost per patient of SGD146

(USD127) leading to a gain of 0.0028 QALY and a resultant ICER of

SGD 45,821/QALY (USD 32,533/QALY). Thus, at a WTP of SGD

71,000/QALY (threshold of 1 GDP/capita), miRNA-based strategy is

cost-effective in comparison to current practice of no screening whereas

endoscopic screening is not cost-effective with an ICER of SGD

112,772/QALY (USD 80,068/QALY). Comparing with endoscopic

screening for all, miRNA-based screening would save SGD160 per

patient with an average gain of 0.0002 QALY (Table 11). Evaluating over

a range of miRNA cost SGD30 – 60 (USD 21-43), the resultant ICER

would lie between SGD 28,000-35,700/QALY (USD 19,806-

25,253/QALY) at a baseline prevalence of 0.13% (Figure 13).

Table 11. Cost-effectiveness results for population screening of Singaporean Chinese from 50-69 years comparing the two strategies: ‘endoscopy for all’ and ‘miRNA test followed by endoscopy only for test positive patients’ (modelled for 20 years)

Strategy Cost

(SGD)

Costs (SGD)

Efficiency

(QALY)

Efficiency

(QALY)

ICER (SGD/Q

ALY)

Current Practice: No screening

20 (USD14)

12.091

miRNA test followed by Endoscopy for test positive patients

146 (USD104)

126 12.0937 0.0027 45,821 (USD

32,729)

Endoscopy for all 306

(USD219) 160 12.0935 -0.0002

112,772 (USD

80,551)

99

miRNA-based strategy also appears to be the most cost-effective

screening strategy for gastric cancer prevalence as high as 0.7% in the

population cohort (Figure 13). With the population cohort size for

Singaporean Chinese 50-69 years estimated to be ~400,000 subjects in

Singapore127, a one-time screening with miRNA-based strategy

(compliance: 70%) would lead to an incremental cost of SGD~51 million

and diagnose 240 cancer cases in comparison to the current practice of

no screening. On the other hand, endoscopy for all (compliance: 45%)

would lead to an incremental cost of SGD~114 M but lead to diagnosis

of only 207 gastric cancer cases in comparison to the current practice of

no screening (Table 12).

Table 12. Results of base-case analysis for population screening for Singapore Chinese Males (50-69 years).

Screening ~400,000 people will lead to an incremental cost of SGD~51 million with miRNA-based strategy and diagnose 240 gastric cancer cases as compared to the current practice of no screening. This is cost-effective with an ICER of SGD 45,821/QALY, which is lower than the threshold of SGD 71,000. Comparing with endoscopy for all, miRNA-based strategy will result in cost savings of SGD 63 million along with 16% better diagnosis rate for cancer cases and an 85% decrease in the endoscopes performed. Baseline used: No screening | GC: Gastric Cancer

Endoscopy

for all miRNA-based

strategy

Cohort size 400,000

Compliance 45% 75%

Incremental cost for screening 114,448,000 (USD 81Mn)

50,660,000 (USD 36Mn)

Number of cancers diagnosed 207 240

To diagnose 1 gastric cancer

Number needed to screen 868 1,165

Number of miRNA tests done - 1,165

Number of endoscopes done 868 129

Cost of diagnosing 1 GC case 552,025 210,794

Incremental Cost per QALY saved 112,772 45,821

100

Figure 13. Sensitivity analysis investigating the impact of gastric cancer incidence rate on cost-effectiveness of endoscopy for all at multiple miRNA test cost scenarios in the populations screening setup for Singaporean Chinese males (50-69 years)

Endoscopy for all has an ICER > 71,000 SGD/QALY for all scenarios’, making it not cost-effective. Whereas, miRNA based strategy always have an ICER lower than the threshold ( < SGD 71,000/QALY) identifying its cost-effectiveness

Sensitivity Analysis

Evaluating the model for sensitivity to the value of variables (variable

range mentioned in (Table 6), cost of miRNA and endoscopy were found

to be significant variables at a WTP of 71,000 SGD/QALY in the

population screening scenario for Singaporean Chinese (50-69 years).

101

SGD200 (USD141) was estimated as the threshold cost for miRNA to

remain cost-effective after which no screening seems as the preferred

approach. In addition, endoscopy with a cost lower than SGD133

(USD94) would drive universal endoscopy to cost-effectiveness.

Although the miRNA test has a lower sensitivity than endoscopy,

achieving a 45% or greater improvement in patient coverage for testing

than endoscopy would result in better health gains, making miRNA-

based strategy a superior strategy for both health and cost benefits

(Table 13).

Probabilistic sensitivity analysis identifies miRNA-based strategy as the

cost-effective strategy for majority of the iterations at a WTP >SGD

60,000/QALY (>80% iterations). 90% of iterations support miRNA cost-

effectiveness for a WTP of SGD 71,000/QALY (WTP: USD

50,700/QALY) (Figure 14).

4.4.3 Population screening in Korea

Replicating the analysis for population screening program for Korea,

miRNA-based strategy was found to be cost-effective with an ICER of

USD 1600/QALY in comparison to endoscopic screening at the base

case scenario with cost of miRNA assumed USGD20 and a patient

compliance of 45% for endoscopy and 70% for miRNA test (Table 14).

miRNA based strategy is expected to improve gastric cancer diagnosis

rate by 10%, identifying 1209 additional cancer cases with an

incremental expenditure of 4 million USD (USD 3,315 per cancer case

102

diagnosed) considering the population size of 22 million adults in this

cohort123 (Table 15).

Sensitivity analysis

Sensitivity analysis reveals three significant variables influencing the

cost effectiveness results, which include the cost of miRNA, improved

patient compliance and specificity of miRNA test. A miRNA cost < USD

24, a 50% or greater improvement in patient compliance and miRNA

Figure 14. Cost-effectiveness acceptability curve for population screening of Singaporean Chinese Males (50-69 years)

miRNA-based strategy was found to be cost-effective for 90% of iterations at a WTP of SGD71,000 (1GDP per capita). WTP represents the amount of an organization is willing to pay to gain 1 QALY of health benefits and helps to develop the cost-effectiveness threshold.

103

specificity higher than 79% would each drive miRNA-based strategy for

cost-effectiveness in the base case scenario with a WTP of USD

26,000/QALY (1 GDP/capita). However, even with a higher miRNA test

cost (USD30-40), an 80% or more improvement in compliance by using

miRNA would drive miRNA-based strategy to cost-effectiveness with its

higher health gains for the population in comparison to endoscopic

screening for all (Figure 15).

Table 13. Sensitivity analysis investigating the impact of improvement in patient compliance with miRNA test on cost-effectiveness of endoscopy in the populations screening setup for Singaporean Chinese males (50-69 years)

Even with a lower sensitivity and specificity than endoscopy, miRNA-based strategy achieves better overall health benefits for a 50% and higher improved patient compliance in comparison to endoscopy.

Improvement in

Compliance with miRNA

Strategies Cost

(SGD) Efficiency

(QALY)

ICER (SGD/QALY)

0%

No Screening 20 12.0909 0

miRNA test followed by Endoscopy (for test positive patients) 101 12.0927 45821

Endoscopy for all 306 12.0935 269255

20%

No Screening 20 12.0909 0

miRNA test followed by Endoscopy (for test positive patients) 117 12.0931 45821

Endoscopy for all 306 12.0935 465377

40%

No Screening 20 12.0909 0

miRNA test followed by Endoscopy (for test positive patients) 134 12.0934 45821

Endoscopy for all 306 12.0935 3478169

60% No Screening 20 12.0909 0

104

miRNA test followed by Endoscopy (for test positive patients) 150 12.0938 45821

Endoscopy for all 306 12.0935 -509496

80%

No Screening 20 12.0909 0

miRNA test followed by Endoscopy (for test positive patients) 166 12.0941 45821

Endoscopy for all 306 12.0935 -211058

100%

No Screening 20 12.0909 0

miRNA test followed by Endoscopy (for test positive patients) 183 12.0945 45821

Endoscopy for all 306 12.0935 -121263

Table 14.: Cost-effectiveness results for population screening program in Korea for Adults ≥40 years, comparing the two strategies: endoscopy for all and miRNA test followed by endoscopy only for test positive patients (modelled for 24 years)

miRNA-based test with an ICER < USD 1,600/QALY is the cost-effective strategy for this cohort

Strategy Cost

(USD) Costs (USD)

Efficiency (QALY)

Efficiency

(QALY)

ICER (USD/QALY)

Endoscopy for all 51.47

14.0143

miRNA test followed by Endoscopy for test positive patients

51.65 USD 0.18

14.01441 0.00011 1600

Table 15. Results of base-case analysis for population screening for population screening program in Korea for Adults ≥40 years

Screening ~22 million people with miRNA-based strategy will lead to an incremental cost of USD ~4 million but diagnose 1209 additional gastric cancer cases as compared to the practice of endoscopic screening. This is cost-effective with an ICER of USD 1600/QALY, which is lower than the threshold of US 26,000 in comparison to endoscopic screening

105

Endoscopy for

all miRNA-based

screening

Cohort size 22,272,389

Compliance 45% 70%

Total cost for screening (USD) 1,146,359,862 1,150,368,892

Number of cancers diagnosed 11544 12753

To diagnose 1 gastric cancer

Number needed to screen 868 1222

Number of miRNA tests done 0 1222

Number of endoscopes done 868 135

Cost of diagnosing 1 GC case (USD) 99,304 90,202

Incremental cost per QALY saved (USD/ QALY)

- 1600

Baseline: Endoscopy for all| GC: Gastric Cancer

4.5 Discussion

In this study, we analyze the cost-effectiveness of translating miRNA

panel as the preferred screening test for gastric cancer in two scenarios

for Singapore – hospital clinic and population screening for adult

Singaporean Chinese males (50-69 years). Our study suggests that

miRNA has a favorably high potential as a cheaper and accurate test for

screening gastric cancer and that long term clinical trials should be

undertaken to assess the benefits. Also, efforts must be made to improve

the sensitivity of the screening specially in specialist clinic setting

through technical modifications or follow-up testing to identify the false

negatives early.

The hospital-clinic scenario, which represents the patients presenting in

specialist clinic with chronic gastric distress, are currently made to

undergo endoscopic screening. Although this has improved the overall

diagnosis, the expenditure is immense with still a low diagnosis rate

106

leading to suboptimal utilization of healthcare services and resources.

miRNA-blood test with a cheaper and simpler testing procedure is much

less resource intensive and easier to integrate in the existing testing

structures and skill set. With its comparable negative predictive value to

endoscopic screening (99.82% vs 99.95%), the projected cost savings

of SGD 450k per 800 persons screened would be a significant asset with

the concerns about current increasing healthcare expenses but at the

expense of 1 missed gastric cancer. However, there is a need to

evaluate the benefits of extending follow up testing for miRNA test

negative patients, which could potentially help in improving the overall

efficacy and safety of this strategy.

With the majority of the gastric cancer cases diagnosed in advanced

stages in the hospital-clinic, the proposed population-screening

scenario, aims to improve the early diagnosis that is favorable for

improving the health and overall survival and reducing the expenditures

on gastric cancer treatment. With the globally recognized approach of

early diagnosis for better survival among cancer patients, there have

been many studies, with a few specific to Singapore evaluating

endoscopic screening in population cohort for achieving this goal.

However, to persuade older populations to undergo an expensive

invasive endoscopy procedure without any immediate health care

concerns is a daunting task. The willingness to undergo a screening test

is of crucial importance for any preventive national program to succeed

which has prominently been missing from earlier studies. Thus, in our

107

analysis, we have tried to forecast the health benefits and budget impact

of using both miRNA-based screening test

Figure 15. Sensitivity analysis investigating the impact of improvement in patient compliance with miRNA test on cost-effectiveness of miRNA-based strategy in comparison with endoscopic screening in the population screening program in Korea (for adults ≥ 40 years)

If miRNA test is unable to improve compliance by 50%more than endoscopy, the ICER of miRNA-based strategy is negative due to higher cost but lower health benefits. However, if compliance improves greater than 80% than that on endoscopy, the miRNA-based strategy becomes cost-effective even at a higher cost than endoscopy.

-60000

-40000

-20000

0

20000

40000

60000

80000

0% 20% 40% 60% 80% 100%

ICE

R (

US

D/

QA

LY

)

(Base

lin

e: E

nd

osc

op

y f

or

all

)

Improvement in Compliance

compared to baseline with miRNA

cost of miRNA = USD 30

cost of miRNA = USD 35

cost of miRNA = USD 40

Threshold

ICER = 26,000

108

benefits and budget impact of using both miRNA-based screening test

and endoscopic screening for the population cohort incorporating

quintessential willingness among the population to be compliant take the

test at the first place. With the miRNA test being a cheaper and simple

blood test, the compliance is expected to be much higher enabling the

health care systems to enforce this preventive care effort and achieve

improved diagnosis

Endoscopy stands as the gold standard for gastric cancer screening,

with an assumed sensitivity of 93% for hospital-clinic and 89% for

population screening in our analysis, whereas miRNA tests sensitivity is

expected to be less accurate with the sensitivity for early gastric cancer

calculated to be 63%. As the population screening is expected to

encounter mostly early gastric cancers, sensitivity of the screening test

is a major concern. However, the improved compliance using the miRNA

blood test in comparison to endoscopy is found to offset the loss due to

lower sensitivity, making it a superior strategy for both costs and health

benefits. The sensitivity of endoscopy is also overly dependent on the

skill and experience of the attending clinician who undertakes this

complicated procedure along with the accuracy of equipment leading to

a varied sensitivity reported in literature ranging 60-70% from Korean

national screening programs to 70-90% from Japanese screening

programs. Whereas, miRNA-test with a comparably standard testing

procedure is expected to have less variability in these test characteristics

109

across countries. An evaluation in Korean healthcare setup also

validates the benefits of using a simpler miRNA test than endoscopy for

national screening program. However, there is a need of detailed cost

evaluations to obtain estimates that are more accurate.

The study has a few limitations. Firstly, the analysis in the hospital-clinic

setup does not consider the endoscopies recommended for other health

conditions and believe all the endoscopies are being performed to

screen for gastric cancer. This was based on the premise that without

the fear of gastric cancer, any other condition like peptic ulcer would

include initial medicine prescription with endoscopes recommended only

on no-response on treatment. Secondly, our evaluation does not

consider indirect costs for the family due to gastric cancer and considers

only the direct costs and implications. Thirdly, the assumption of

willingness-to-pay is based on the WHO guidelines, which was the best

estimate in absence of locally published guidelines. In addition, the

improvement in compliance with miRNA test is based on an estimate.

Although sensitivity analysis evaluates for all the values across a range,

there is a need to validate these parameters locally in the population to

evaluate the benefits more accurately. Lastly, an evaluation measuring

the cost-effectiveness for an extended strategy consisting annual or

biennial follow-up for persons with negative miRNA test would be able

to explore detailed screening programs and evaluate the cost and health

implications more accurately.

110

The future of cancer prevention relies on timely detection of cancer and

appropriate treatment, making higher survival rates and lower healthcare

cost feasible. With the ageing population of Singapore and the world, the

population at risk is increasing. Effective screening or prevention

programs can be efficient tools to check the rise of cancer and lead to

reductions in incidence and subsequent mortality. The national cancer

screening programs in Korea and Japan have exemplified this claim.

However, cancers with its low incidence but grave health and economic

consequences calls for a need to optimize screening programs and find

innovative, effective and cheaper methods of cancer prevention and

early detection to achieve the goal of improved care, better survival and

manageable costs.

111

Chapter 5 Cost-Effectiveness and Value of Information Analysis

of SLCO1B1 Genotyping (rs4149056) before

Simvastatin Prescription for Secondary Prevention of

Acute Myocardial Infraction in Singapore

5.1 Introduction

The burden of cardiovascular disease is immense worldwide. It is the

number one killer globally with a death every minute from a heart

disease-related event in United States (US)128. With greater emphasis

to promote good cardiac health, reduction of high cholesterol is one of

the significant risk factors being addressed. In US about 32% of the

population has high cholesterol129. Singapore also faces 17-25% of its

population to suffer from high cholesterol (aged 18-69 years and

above)130. This has led to a substantial increase in the use of statins –

the most commonly prescribed cholesterol lowering drugs globally with

studies proving its cost-effectiveness both for primary and secondary

prevention131. US alone consumed 17 billion worth of statins related

drugs in 2012132.

Simvastatin has long been the preferred statin for cholesterol lowering

in the world133. With good efficacy, it is cheaper than most of the other

statins and has been proven to decrease the risk of heart attack, stroke

and cardiovascular morbidity and mortality134-137. However, there has

been an increasing concern for its side effects138-142 – myalgia, myopathy

112

and rhabdomyolysis, shifting the prescriptions to other costlier

alternative drugs – Atorvastatin, Rosuvastatin, Ezetimibe-Statin

combinations and other statins.

Many of these side effects, which are muscle related, are characterized

by debilitating muscular pain, muscle breakdown, increasing creatinine

levels in the blood and in severe cases of myopathy and rhabdomyolysis

may cause kidney damage or even death. Though clinical trials report

negligible side effect rates, observational and behavioral studies are

headlining the high prevalence of side effects which usually go

unreported in formal dossiers68,140,143. The USAGE survey

(Understanding Statin Usage in America and Gaps in Patient

Education), the largest survey conducted to understand the behavior

and attitude of cholesterol users in US68, highlights a 30% prevalence of

side effects among the statin-users with a non-compliance rate as high

as 25%. It further concludes side effects as a major reason of non-

compliance accounting for 60% of such cases. With high discontinuation

rate among statin-users, non-compliance is emerging as a major

challenge to effectively lower cholesterol and prevent cardiovascular

diseases.

Researchers have identified significant genetic linkages with the

potential to stratify patients by side effect risks144,145 and prescribe

Simvastatin with higher confidence. A significant genetic association has

been found between a SNP (rs4149056) in SLCO1B1 gene and side

113

effects on Simvastatin144,145. A carrier of one or two copies of the risk

allele carrier are estimated to be at 4.5 times to 17-times increased risk

for developing significant myopathy respectively and 2-fold increased

risk of myalgia in comparison to non-risk allele carriers144,145. Based on

multiple study reports, the Clinical Pharmacogenetics Implementation

Consortium (CPIC) has issued an update to Simvastatin Dosage

Guideline in 201436 warning about this genetic linkage and

recommending a lower dosage or an alternative statin to the risk-allele

carriers36. Though currently, FDA does not recommend SLCO1B1

screening before Simvastatin prescription, many clinical studies like

Vanderbilt university’s PREDICT initiative & Icahn School of Med.’s

CLIPMERGE program are testing patients for this variant and a success

story has already been published22.

The objective of this study is to evaluate the cost-effectiveness of

rs4149056 genetic screening before Simvastatin prescription in

comparison to the current and other plausible management options for

acute myocardial infraction (AMI) patients in Singapore. The study

focusses on Chinese male patients, who carry the majority of the disease

burden in the country146, addressing secondary prevention, where the

absolute benefit of efficient statin treatment is expected to be among the

highest. To the best of our knowledge, this is the first study evaluating

the cost-effectiveness of SLCO1B1 genetic testing in a clinical setting.

We further aim to assess the significant uncertainties in the current

114

knowledgebase and identify priority areas of future research using value

of information analysis (VOI).

5.2 Methods

5.2.1 Model Structure

A markov model was developed with a healthcare perspective in

TreeAge Pro 2017 for secondary prevention of AMI patients in Singapore

with an aim to quantify the health and cost implications of all strategies

and their impact on side effect cases and resultant non-adherence

among patients (Figure 16). Strategies compared were: 1) Simvastatin

as 1st line for all; 2) Atorvastatin as 1st line for all; 3) Genotyping and

Atorvastatin to risk allele carriers and Simvastatin to risk allele non-

carriers; 4) Ezetimibe/ Simvastatin combination as 1st-line for all. The

prescribed patient was expected to be either compatible to the drug or

develop side effects. The model considered only muscle-related side

effects – myalgia (Creatine Kinase (CK) < 3 ULN)147, myopathy (3 < CK

< 10 ULN)147 and rhabdomyolysis (CK >10 ULN)147 (ULN: upper limit of

normal range). Patients with side effects were managed as per clinical

recommendations (explained below) with some myalgia and myopathy

patients expected to become non-adherent69. Non-adherence would not

have an immediate impact on the patient’s health but lead to an increase

in risk of recurrent AMI (in comparison to those adherent to statins

>80%)137,148. All strategies have been illustrated in Figure 18.

115

Safer statin prescription after genotyping is expected to reduce the side

effect rates, improving patient adherence to simvastatin and a

subsequent reduction in recurrent AMI events. As majority of side effects

and ~50% of recurrent AMI’s happens in the first year of statin

treatment137,149, the model has been developed with a one year time

horizon with an aim to compute the side effect cases avoided with

confidence and identify parameters critical for cost-effectiveness.

Side effects on Cholesterol Lowering Drugs

Only muscle-related side effects were considered as they form most of

the clinical complaints. Myalgia leads to muscle pains, myopathy causes

serious muscle weaknesses, degradation and pain and rhabdomyolysis,

a severe and debilitating side effect, leads to degradation of skeletal

muscle, potential kidney damage and even death in rare cases.

All cholesterol lowering drugs were prescribed in comparable medium

intensity dosages (based on LDL lowering capacity) with non-statin

drugs having a low-intensity dosage prescription59 (Table 16).

Simvastatin and Atorvastatin could result in side effect events of myalgia,

myopathy or rhabdomyolysis after 1 month140,147 whereas rosuvastatin,

a safer and well tolerated statin, was expected to cause only myalgia

after a mean time of 5 months150. Ezetimibe/ Simvastatin combination is

reported to be very safe with negligible side effects. However, a 2%

myalgia rate occurring after 1 month of prescription was assumed to

account for the rare occurrences151. Though in current practice,

116

intolerance of a patient to all statins – Simvastatin, Atorvastatin,

Rosuvastatin and Ezetimibe-Simvastatin combinations is a rare

possibility, but our model considers this event and such patients are

prescribed on non-statin alternative. Once switched to alternatives, the

patients were expected to be compatible and adherent to them for the

rest of the duration of the analysis. The non-statin alternatives

considered were Ezetimibe alone, Fibrates or Gemfibrozil.

Clinical Management of Side effects

If patients develop myalgia, they were expected to be immediately

switched to the next in-line treatment – Simvastatin, Atorvastatin,

Rosuvastatin, Ezetimibe/ Simvastatin combination or non-statins in the

given order of preference with myalgia symptoms persisting for 2

months147. For myopathy and rhabdomyolysis cases, patients were

immediately stopped from the current medication and treated for the side

effect with an estimated time to recover of 2 and 4 months

respectively147. Only after complete recovery, the patients were re-

initiated on drugs- myopathy patients on either Rosuvastatin or

Ezetimibe/ Simvastatin combination, whereas rhabdomyolysis patients

were started on non-statin alternatives only, as they were considered

contra-indicated to statins after the side effect. Patients developing

myalgia on the Ezetimibe-Simvastatin combination administered as 1st

line (4th strategy), were expected to switch to Rosuvastatin followed by

non-statin alternatives only if myalgia re-appears. Non-statin alternatives

117

with a lower efficacy than medium-intensity statins were also considered

to increase the AMI risk152.

The model considers only additional side effect cases, AMI cases and

consequent mortality and expenses and did not account for the identical

baseline costs, death rates and other health conditions associated with

high cholesterol. Only 30-day mortality cases were considered for

recurrent AMI as it accounts for >75% of AMI related mortality146. Based

on WHO guidelines, the willingness-to-pay (WTP) was fixed at

SGD71,000 (~ equivalent to 1GDP/ capita)71,153.

5.2.2 Sources of Information

Rate of Side effect

The analysis was performed with the perspective of Singapore

healthcare systems and evidence obtained from literature, Singapore

registry and hospital data from National University Hospital (NUH),

Singapore (Table 16). As the literature does not report the side effect

rates for Simvastatin 40mg, a retrospective analysis was performed

using electronic medical records of diabetes mellitus patient admitted in

National University Hospital from January 2010 – December 2011. The

selection criteria have been illustrated (Figure 17). The dataset was

curated for a separate study and comprised of patients with Type 2

diabetes mellitus (T2DM, identified through a combination of anti-

diabetes drugs, International Classification of Diseases (ICD) diagnosis

118

codes and laboratory tests) who were managed in NUH. We further

extracted a subset of Chinese male patients with an earlier episode of

1A

The model compares the four strategies with the preferred first-line drugs: 1) Simvastatin for all; 2) Atorvastatin for all; 3) Genotyping and Simvastatin for allele negative patients and Atorvastatin for allele positive patients; 4) Ezetimibe-

119

Simvastatin for all. Abbreviations used: Simva40: Simvastatin 40 mg, Atorva 20: Atorvastatin 20 mg, Rosuva 5 mg: Rosuvastatin 5mg. Non-statin alternatives considered are ezetimibe, fenofibrate or gemfibrozil.

1A) Decision tree model for the strategy 1: Simvastatin for all. The strategy begins with Simvastatin for all as first line which could potentially lead to side effects namely-myalgia, myopathy, and rhabdomyolysis or long term treatment with no side effects. Patients on myalgia would be immediately switched to the next line with side effect symptoms continuing for next 2 months. Patients with myopathy and rhabdomyolysis will be first treated for the side effect and then switched to the next-in line treatment. Myalgia and myopathy patients are expected to become non-adherent which increases their risk of recurrent AMI. The decision tree only considers additional AMI cases and does not account of baseline events and costs similar to all strategies.

Strategy 2: Atorvastatin for all can be identified in the tree structure as a subset of the first strategy, starting from “atorva 20mg (2nd line)” as the first line. Strategy3: Genotyping strategy involves SLCO1B1 genetic testing for all patients with Simvastatin to allele non-carriers and Atorvastatin for allele carriers.

1B

Decision Tree Model for Strategy 4: This strategy considers ezetimibe-Simvastatin combination (10/10 mg) for all as first-line drug with myalgia as a possible side effect. Myalgia patients are then immediately switched to the next line of treatment i.e. rosuvastatin 5 mg recovering with myalgia symptoms for the next 2 months. Patients on rosuvastatin are expected to be tolerant with a small fraction facing myalgia, who would be switched immediately to non-statin alternatives. This strategy does not consider prescription of Simvastatin or Atorvastatin to patients.

120

Figure 16. Markov-Decision Tree Model for evaluating the cost-effectiveness of cholesterol management strategies for secondary prevention in AMI patients in Singapore

AMI and those being treated on Simvastatin 40mg after the AMI episode

were identified. We further used the serum CK levels to identify the

patients with side effects due to Simvastatin excluding those patients

with CK/ CKMB ratio >2.5 to remove potential CK elevations due to

cardiac events themselves. CKMB is the bound combination of two

variants (isoenzyme CKM and CKB) of the enzyme phosphocreatine

kinase which is used as a biomarker to diagnose acute myocardial

infraction. Though diabetes mellitus patients can be expected to be at a

higher risk of side effect, literature reports mixed reviews with some

studies reporting significant differences whereas others identifying no

difference in the risk profile142,154,155. In our current analysis, we assume

a similar risk profile overall for patients under secondary prevention.

Costs

Cost of genotyping was assumed to be SGD200 in accordance with the

existing cost of similar tests (HLA-B*1502 screening) and unsubsidized

costs for cholesterol lowering treatments were obtained from hospital

pharmacy. The treatment expenses for the side effects were estimated

by accounting the health services expenses used for side effect

management based on expert opinion. Cost of treating AMI was

estimated from average of hospital bills across six public hospitals in

Singapore, which was adjusted to account for the subsidy.

121

Table 16. Model estimates and sources

Parameter Value Sensitivity

Range Distribution Source

Costs

Genetic Test Cost 200 0.0 to 500.0 Gamma(α = 7.11, λ = 0.036) Assumed

Simvastatin 40mg cost (monthly) 7.2 3.6 to 21.6

Gamma(α = 36, λ = 5)

NUH

Atorvastatin 20 mg cost (monthly) 40.2 40.0 to 160.0

Gamma(α = 74.74, λ = 1.86)

Rosuvastatin 5 mg cost (monthly) 46.5 10.0 to 50.0

Gamma(α = 1465.8, λ = 36.46)

Expense (fatal Rhabdomyolysis) 40675

0.0 to 80,000.0

Gamma(α = 16, λ = 0.0004)

NUH (expert opinion)

Consultation cost 80 40.0 to 160.0 Fixed NUH

Routine test cost (for patients with side–effect)* 40 20.0 to 80.0 Fixed NUH

Expense (Myopathy) 3944

1,000.0 to 8,000.0

Gamma(α = 9.0, λ = 0.002)

NUH (based on expert opinion) Expense( Myalgia) 134 50.0 to 500.0

Gamma(α = 3.19, λ = 0.024)

Figure 17. Selection criteria for determining the Simvastatin side effect Burden The data was analysed form the diabetes mellitus patient database for Chinese patients from NUH, Singapore (1/2010- 12/2011)

122

Expense (Rhabdomyolysis) 13923

0.0 to 25,000.0

Gamma(α = 16, λ = 0.001)

Ezetimibe/ Simvastatin (10/10 mg) cost (monthly) 87 40.0 to 160.0

Gamma(α = 5256.25, λ = 60.42) NUH

Cost of non-statin alternatives (last line of treatment)** 47 4.0 to 80.0

Gamma(α = 2.16, λ = 0.05) NUH

Hospital expense on AMI (1st month)^ 18353

0.0 to 18,353.0

Gamma(α = 10.80, λ = 0.0006)

ref156

Allele frequency

Allele frequency (C/C: C/T:T/T)

0.021 :0.177 :0.80

2 0.6 to 1.0 Fixed ref94

Non Adherence

Non adherence (on myalgia ) 27% 0.0 to 1.0

Beta(α = 5.05, β = 13.66)

ref69

Non adherence (on myopathy) 40% 0.0 to 1.0

Beta(α = 2.92, β = 4.38)

Probabilities

Myalgia (Simvastatin) 22% 0.0 to 0.3

Beta(α = 11, β = 39)

NUH (DM patient database)

Myopathy (Simvastatin) 10% 0.0 to 0.1

Beta(α = 17, β = 134)

Rhabdomyolysis (Simvastatin) 4.50% 0.0 to 0.05

Beta(α = 6, β = 134)

Death rate (Rhabdomyolysis) 10% 0.0 to 1.0 Fixed ref65,157

Recurrent AMI rate 19% 0.0 to 0.3 Beta(α = 32.3, β = 137.7) ref146,148

Death rate (AMI: 30-days mortality) 14% 0.07 to 0.28

Beta(α = 6.60, β = 40.56) ref146

Myalgia (Rosuvastatin) 11% 0.0 to 0.2

Beta(α = 11.86, β = 95.92) ref150

Myalgia (Ezetimibe/ Simvastatin: 10/10) 2% 0.01 to 0.2

Beta(α = 0.34, β = 16.66) assumed

Quality of life

QoL on Rhabdomyolysis p 0.4 0.0 to 0.61

Beta(α = 0.30, β = 0.45)

ref158

QoL on Myopathy q 0.61 0.4 to 0.85 Beta(α = 2.68, β = 1.72) ref159

QoL in patients with an index AMI 0.85 0.73 to 1.0

Beta(α = 1.86, β = 0.33) ref160

QoL (Myalgia) r 0.73 0.61 to 0.85 Beta(α = 3.71, β = 1.37) ref159

QoL after AMI (immediate) 0.66 0.0 to 0.73 Fixed

ref160

123

QoL (after recurrent AMI)

0.73 (0.66 - 0.85) 0.66 to 0.85

Beta(α = 1.57, β = 0.58)

Relative Risks/ Odds Ratio

OR (Myalgia on Atorvastatin) a 0.72 0.45 – 1.15 - ref140

RR of myalgia on Atorvastatin b 0.77 0.77 to 1.1

ln N(µ = -0.26, σ = 0.1) ref140

RR of AMI (statin adherence > 80%) 0.19 0.08 to 0.47

Beta(α = 2.06, β = 8.78) ref137

RR of AMIc (on non-statin alternatives/ pravastatin) 1.29 1.06 to 1.53

ln N(µ = 0.25, σ = 0.09) ref152

RR of Rhabdomyolysis on Atorvastatin 1 0.5 to 1.5

ln N(µ = 0, σ = 0.28) Assumed

Notes: All drugs except alternatives are prescribed in comparable medium intensity dosage (LDL lowering capacity- Simvastatin 40mg, Atorvastatin 20 mg, Rosuvastatin-5mg, Ezetimibe-Simvastatin combination 10/10mg)59. Simvastatin and Atorvastatin can cause all three side effects – myalgia, myopathy and rhabdomyolysis, Rosuvastatin and Ezetimibe-Simvastatin combination with only side effect of myalgia. *Routine tests in such cases include: Urine analysis + CK + LDH (lactate dehydrogenase) + AST (aspartate transaminase). **Alternatives include non-statins with low-intensity dosage in terms of LDL lowering capacity (Ezetimibe alone -10mg, fenofibrate – 150mg, gemfibrozil-600mg x twice daily) and Pravastatin (40mg). ^Approximated to the expense in angioplasty+ stent (without complications) as they represent majority of treatment cases in Singapore. Average of hospital bill size across public hospitals is considered. aOR was reported for high dosage of Atorvastatin (20,40mg) and Simvastatin (40,80mg). bRR were calculated using formula from ref157. cRR of AMI on alternatives (low intensity dosage) was considered equal to low intensity statins with medium intensity statins as reference152. Ezetimibe-Simvastatin combination is very safe for use but a 2% side effect rate has been assumed accounting for rare cases161. QoL due to side effects have been approximated to similar health conditions: p acute kidney injury, q chronic widespread pain, rfibromyalgia.

124

A) Strategy 1: Simvastatin as 1st line followed by 2nd line drugs based on treatment outcomes: Atorvastatin (myalgia), Rosuvastatin or Ezetimibe-Simvastatin combination (myopathy) and alternative drugs (rhabdomyolysis). Please refer to the key (with the figure caption) to read the figure below

B) Strategy2: Atorvastatin as 1st line followed by 2nd line drugs based on treatment outcomes: rosuvastatin (myalgia), rosuvastatin or ezetimibe-Simvastatin combination (myopathy) and alternatives as 2nd line (rhabdomyolysis)

Strategy3: Genotyping and A) for risk allele non-carriers and B) for risk allele carriers Patients with myalgia on rosuvastatin are prescribed ezetimibe-Simvastatin combination, followed with alternatives if myalgia persists on ezetimibe-Simvastatin combination (rare possibility)

A)

B)

125

Figure 18. Possible cholesterol lowering treatment therapies compared for secondary prevention

Quality-of-life (QoL) value

Baseline QoL of patients for secondary prevention was assumed based

on published literature160. As statin side effects are not very widely

studied with negligible literature reporting their quality of life values,

these estimates were obtained for the health condition with similar health

outcomes, namely, widespread body pain for myalgia, fibromyalgia for

C) Strategy4: Ezetimibe-Simvastatin combination as 1st line followed by rosuvastatin as 2nd line in case of myalgia and alternatives as 3rd line if myalgia occurs again.

Key to the figure: SE outline colors represent the causative drug (for ex: Myalgia with green outline represents myalgia caused due to Simvastatin). As all the side effects are expected to happen within one month, the text boxes are accordingly

placed with the vertical lines with a t=1 mentioned below. Vertical lines represent time points. The next-in line treatment follow the side effect, which is atorvastatin for myalgia (due to simvastatin in strategy A) and rest and treatment lag for myopathy and rhabdomyolysis cases. The period of treatment lag in myopathy and rhabdomyolysis cases is represented as recovering. Due to space constraints, SE corresponding to the 1st time a drug appears in the strategy is shown. All other occurrences can have similar outcomes | Alternatives refer to drugs like – fenofibrate, gemfibrozil, ezetimibe alone or Pravastatin at low intensity dosage. All other drugs are prescribed at medium intensity (based on LDL lowering capacity)

Abbreviations used: Simva: Simvastatin 40mg; Atorva: Atorvastatin 20mg; Rosuva5: Rosuvastatin 5mg, Eze/Simva combination: Ezetimibe 10mg + Simvastatin 10mg; Myo.: Myopathy; Rhabdo: Rhabdomyolysis; SE: side effect

C)

126

myopathy and acute kidney injury for rhabdomyolysis. AMI incidence is

expected to cause an immediate decrease in QoL for 2 months after

which the health is expected to return to a baseline value lower than that

of initial patient cohort. All values have been reported in Table 16.

Other variables

As reported by literature, Atorvastatin was considered to have a safer

profile for myalgia than Simvastatin140, but have similar rates for

myopathy and rhabdomyolysis. Reduced risk of AMI recurrence among

patients adherent to statins was estimated from a six-year follow up

study investigating impact of statin adherence (>80% adherence) on

secondary prevention patients in Scotland, UK137. The patient non-

adherence rates were estimated from a retrospective cohort study

evaluating the discontinuation of statins in routine care settings in the US

with all patients with >80% adherence to enjoy the health benefits of full

compliance69.

5.2.3 Sensitivity analysis

Both discrete and probabilistic sensitivity analysis (100,000 iterations)

were performed to test the model robustness with gamma, beta and

lognormal distributions being assigned to cost, probabilities and relative

risks variables respectively (Table 16). A discrete 2-way and 3-way

sensitivity analysis was also done to explore the impact of uncertainty

surrounding side effect rates, relative risk of side effects and non-

adherence among patients on overall decision making. Monte Carlo

127

simulations were used to estimate net benefits (NB) and calculate value

of information (VOI) measures (1000 inner and 1000 outer loops)

reporting the worth of future research for those parameters. A budget

impact evaluation was done to analyze the affordability of clinical

translation.

5.3 Results

For secondary prevention of AMI in Singapore using cholesterol lowering

strategies, our analysis indicated Simvastatin for all as 1st-line as the

cheapest strategy and Ezetimibe-Simvastatin combination as the most

cost-effective strategy (Table 17, Figure 19). Both genotyping strategy

and Atorvastatin for all as 1st-line were dominated by Ezetimibe-

Simvastatin combination (Figure 19) which adds an average of 0.2 QALY

per patient, achieving a 100% reduction in myopathy and

rhabdomyolysis cases and ~90% reduction in myalgia cases. Sensitivity

analysis identifies rate of rhabdomyolysis on Simvastatin, quality of life

values for rhabdomyolysis and AMI patients in the first month to be of

critical importance with threshold values of 0.6% or greater and below

0.09 or 0.4 respectively to favor Ezetimibe-Simvastatin combination

(Figure 20). Probabilistic sensitivity analysis also ensures the robustness

of the results with this strategy being cost-effective for 87% of iterations

(Figure 21). However, the average additional cost this strategy incurs is

estimated to be SGD520 with a potential budget impact of 1.2 - 2.2

million dollars in the first year for the limited cohort of Singaporean

128

Chinese for secondary prevention (4327 patients annually)146 (Table 18).

With this huge cost-burden and long term treatment requirement, there

are compelling concerns about the affordability of this intervention.

Evaluating for more affordable treatment strategy, we compare the

remaining treatment approaches. Comparing with Atorvastatin for all as

1st line, which is currently the most commonly followed practice for

secondary prevention in Singapore, the genotyping strategy was found

to a cheaper (SGD97 cost savings per patient) with better health benefits

(an average gain of 0.01 QALY per person) and hence, the cost-effective

strategy with an ICER of 33,645 SGD/QALY. Simvastatin for all as 1st-

line without genotyping was a cheaper but less effective strategy (Table

17).

For the genotyping strategy, along with the cost benefit, the major benefit

was a decrease in myopathy cases by 8% and rhabdomyolysis by 10%

as compared to the current practice but a possible 44% increase in

myalgia cases (Table 19). However, as the resultant expense and

consequences of non-adherence among the myalgia patients are less

consequential than myopathy and rhabdomyolysis, the myalgia losses

were outweighed by the benefits of a lower myopathy and

rhabdomyolysis rate.

5.3.1 Sensitivity analysis

The one-way sensitivity analysis reveals the genotyping strategy to be

cost-effective compared to Simvastatin for all if the rate of

129

rhabdomyolysis on Simvastatin is 1.6% or higher. In comparison to the

current practice, Atorvastatin having a relative risk of rhabdomyolysis of

0.8 or greater than Simvastatin or a QoL value on rhabdomyolysis lesser

than 0.17 favors genotyping. The model robustness was reaffirmed by

probabilistic sensitivity analysis, with 65% iterations supportive of the

genotyping strategy followed by 23% in favor of Atorvastatin as 1st line

and 11% for Simvastatin for all (Figure 22).

5.3.2 Value of Information analysis

Value of information (VOI) analysis identifies overall model uncertainty

to risk SGD1.2 million in treatment decision making in the first year with

the relative risk of side effects and the absolute side effect rates to be

significant in driving cost-effectiveness. The RR of myalgia and

rhabdomyolysis on Atorvastatin in comparison to Simvastatin was

identified as a potentially significant uncertainty amounting to 50% of the

overall risk (Table 20). The probability of non-adherence, and cost-

variables were not found to be of significant importance.

Table 17. Cost-effectiveness analysis of the cholesterol lowering strategies for secondary prevention in Singapore

No. Strategies Cost

(SGD) Δ

Cost Efficacy (QALY)

ΔEfficacy ICER (SGD/QALY)

1 Simvastatin to all 517 Ref 0.8295 Ref Ref

2 Genotyping and Simvastatin to risk allele non-carriers and Atorvastatin 20mg to risk allele carriers

653 136 0.8335 0.004 33645

3 Atorvastatin to all 750 233 0.8325 0.0029 79255

130

Figure 19. Cost-effectiveness analysis comparing the four strategies on their cost and effectiveness

Evaluating cost-effectiveness graphically, the cost-effectiveness frontier connects Simvastatin for all, which is the cheapest option with Ezetimibe-Simvastatin combination strategy which is the optimal choice. The strategy Atorvastatin for all is dominated with genotyping strategy being cheaper and more effective. Also, genotyping strategy is being extendedly dominated as a combination of a combination of other two strategies on the cost-effectiveness

frontier could potentially be a cheaper and more effective alternative.

4 Ezetimibe-Simvastatin combination to all

1037 520 0.8495 0.02 26055

Abbreviations used: QALY: Quality adjusted life years; ICER: Incremental cost-effectiveness ratio; Ref: Reference

131

Table 18. 1-year Budget Impact of Ezetimibe-Simvastatin combination in Singapore in comparison other cholesterol management strategies

No. Strategies compared with ezetimibe-Simvastatin for all

Δ Cost

Δ QALY

Budget Impact (population)

1 Simvastatin 40 mg for all 520 0.02 2.2 M

2

Genotyping and Simvastatin 40mg to

risk allele non-carriers and Atorvastatin

20mg to risk allele carriers

384 0.19 1.6 M

3 Atorvastatin 20 mg to all 287 0.2 1.2 M

Note: Every year 4237 Chinese experience AMI for the first time in Singapore

Table 19. Evaluation of the rate of side effects across different strategies

Simvastatin

for all

Genotyping & atorva to risk

allele +ve

Atorva for all (no simva.

ever)

Ezetimibe & Simva. comb. for

all

Myalgia 32.47% 26.74% 18.51% 2.08%

Myopathy 11.59% 9.16% 9.97% 0.00%

Rhabdomyolysis 5.31% 4.08% 4.55% 0.00%

Recurrent AMI (overall)*

2.03% 1.63% 1.58% 0.09%

Table 20. Estimates of the value of additional information to resolve uncertainty

Per-patient Value (SGD)

Population Value (SGD)

EVPI* 287 1.2 million

EVPPI^

RR (Rhabdomyolysis on Atorvastatin) RR (Myalgia on Atorvastatin) Baseline: Simvastatin

145.62 616k

Side effect probabilities 6.57 27.8k

RR_AMI on non adherence 0 -

RR_AMI on a lower dosage 0 -

Non Adherence values 0 -

Cost variables 0 -

Note: Every year 4237 Chinese experience AMI for the first time in Singapore | *100,000 iterations are done for EVPI calculations| ^ 1000 iterations were run for both inner (PSA) and outer loops (Value-of-information) for EVPPI calculations.

132

EVPI: Expected value of perfect information | EVPPI: Expected Value of perfect parameter information| RR: Relative risk

Figure 20. Tornado analysis evaluating the impact on ICER for genotyping strategy vs. Simvastatin for all

Using one-way sensitivity analysis (Tornado analysis), we are analysing the influence of uncertainty of independent variables on Incremental cost-effectiveness ratio of Ezetimibe-Simvastatin combination strategy in comparison to Simvastatin for all which is the cheapest strategy. Rate of rhabdomyolysis on Simvastatin, quality of life values for rhabdomyolysis and AMI patients in the first month are found to be of critical importance with threshold values of 0.6% or greater and below 0.09 or 0.4 respectively to favour Ezetimibe-Simvastatin combination. These variables have been highlighted in red.

133

Figure 21. Cost-effectiveness acceptability curve (Probabilistic Sensitivity analysis) evaluating Ezetimibe-Simvastatin combination vs. Simvastatin for all

Cost-effectiveness acceptability curve represents the number of iterations for which genotyping strategy is cost-effective for varying willingness-to-pay (WTP). This study found ezetimibe-Simvastatin combination to be cost-effective in 87% of the iterations and Simvastatin for all strategy as cost-effective only for 13% of iterations.

5.4 Discussion

Our study identified Ezetimibe-Simvastatin combination as the safest

drug options for cholesterol management aimed at secondary prevention

of AMI in Singaporean Chinese males with an ICER of SGD

26,055/QALY in the first year of treatment. Although this strategy offers

the best health benefit profile, it has very high costs (12x expensive

134

than Simvastatin) leading to concerns of additional economic burden to

the healthcare system in the long run. An average patient with a survival

of 16 years after the index AMI (mean age of index AMI: 68 years), is

expected to spend approximately an additional 15,000 SGD on

treatment costs during the lifetime based on these estimates. However,

with cheaper generic versions of Ezetimibe expected to enter the market

soon, long term analysis of cost-implications would be valuable.

Figure 22. Cost-effectiveness acceptability curve (Probabilistic Sensitivity analysis)

Cost-effectiveness acceptability curve represents the number of iterations for which genotyping strategy is cost-effective for varying willingness-to-pay (WTP). This study found genotyping strategy to be cost-effective in 65% of the iterations and Atorvastatin for all strategy as cost-effective only for 23% of iterations. This evaluation was done with the genotyping cost assumed to vary between SGD5 – SGD500.

135

Further analyzing with atorvastatin for all, the most prevalent clinical

practice, genotyping before Simvastatin prescriptions was found to be

cost-effective when evaluated for the first year of treatment. Although

with an incremental cost of SGD200 for genetic test, genotyping strategy

is eventually cost-savings for the patient with better health benefits (cost-

saving of SGD100 and incremental QALY of 0.01) in the first year itself.

As statins are prescribed for longer durations, cost-savings are expected

to increase with Simvastatin being 5x times cheaper than Atorvastatin in

Singapore. We also identify side effect rates as a critical driving factor

and recognize the need of future research to quantify population specific

side effect rates corresponding to statin dosages. Thus, our study

suggests that it may be worthwhile to prioritize a long term evaluation

but with more certain estimates of relative risks and absolute measures

of statins’ side effects, specific by their dosage levels.

Our study has a few limitations. With the lack of literature reporting statin

side effect rates, our side effect estimates are based on a limited patient

cohort seen in National University Hospital. Although we have been

conservative in identifying side effect cases, excluding patients with CK

elevations due to cardiac events, the rates identified are higher than the

published values available. This may be due to the event of sampling

errors or an increased risk in our cohort due to age, an earlier AMI event

or Chinese ethnicity. Also, only CK elevations have been used to identify

side effect cases. This is specifically of concern among myalgia patients,

as many cases with statin associated muscle pain but without any CK

136

elevations have been reported. Also, with only voluntary reporting of side

effect cases, it was a practical limitation to identify these events and

could have potentially lead to underestimation of actual myalgia

concerns in the society. Value-of-information analysis identifies

uncertainty in side effect rates of significant value and future studies

evaluating the statin side effect rates in Singapore would be potentially

worthwhile to drive decision making.

Also, non-adherence rates among patients with side effects have been

estimated from a study done in the US. With patient behavior varying in

Singapore, these estimates may change. However, sensitivity analysis

and value-of-information analysis have revealed it to be not a critical

parameter of concern. Also, quality-of-life values for side effects have

been estimated based on similar health conditions, which however are

the best estimate based on the available literature. Our model considers

only AMI events and does not account for stroke and other health

complications. However, as prevention of only one outcome i.e. AMI

favors cost-effectiveness, the actual benefits which can be derived from

using genotyping to achieve a safer statin prescription profile and better

cholesterol management are expected to be higher. Finally, our model

evaluates only for a 1-year time horizon. Though, longer time duration

would have been ideal, but with limited available evidence on dosage

specific side effect rates and impact on quality of life, a 1-year study still

enables to validate potential health and cost benefits of a safer statin

prescription practice and identify uncertainties which are critical to

137

perform future long-term evaluations with confidence. As majority of side

effects and recurrent AMI happen in the first-year of secondary

prevention, our study intends to capture the majority of side effects

avoided by using a safer statin prescription method and with the results

showing cost-savings for genotyping even in this short timeline, it

provides strong evidence of the cost-effectiveness in real practice.

Lastly, though our study has been based on strong clinical evidence of

the clinical benefits offered by statins in secondary AMI prevention, there

have been scientific speculations on the benefits. Thus, remaining

mindful of the scientific progress is advisable to ensure the applicability

of this evaluation.

With an increase in concern over statin side effect rates in observational

studies and clinically useful genetic associations, there is a need to

identify novel methods to implement stratified medicine in clinics for the

benefit for all. Our study is an attempt to evaluate the cost-effectiveness

of application of SLCO1B1 genotyping in actual practice in Singapore.

Our study has identified that among all parameters relative risk of side

effects on Atorvastatin in comparison to Simvastatin and the availability

of lower cost generics are critical for efficient decision making. Further,

internationally recognized clinical guidelines to manage statin side effect

cases and analysis evaluating the implications of long term costs

impacts would help to evaluate across ethnicities and health care

systems.

138

Chapter 6 Conclusion

The healthcare market is expected to reach USD8.7 trillion dollars by

2020 with an expected increase of 2.4-7.5% between 2015 to 2020

worldwide162. With increasing life expectancy, ageing population and

immense burden of chronic and other diseases, the world is reeling

under an enormous pressure to provide effective healthcare to all.

Innovation is being sought in every aspect - healthcare practices,

operations, social engagement, financial and care delivery models to

achieve improved patient outcomes cost-effectively.

Development of efficacious and compatible treatment practices to

improve patient outcomes is a fundamental goal to reduce clinical and

financial inefficiencies in the system. With the advent of genomics and

many success stories, the future medicine promises to be much more

customized to patient’s needs. It involves focus on prescription of

genomic compatible drugs, a personalised drug development approach

as well as pharmaco-vigilance checks. Here cost-effectiveness analysis

would work as an impetus for the network, aiding in successful

incorporation in standard care with proved economic gains. Convincing

the health systems to make the necessary changes to enhance clinical

practice must stem from an evidence-based approach that rigorously

interrogates whether a pharmacogenomic test genuinely improves the

quality of care in a cost-effective manner, prior to convincing regulatory

agencies on its implementation. Implementation of pharmacogenomic

139

IDs in Thailand serves as a commendable example where even in a

resource-limited setup; the country equipped with its pharmacogenomic

strategy is on the verge of eradicating the severe SJS/TEN side effect

(arising from carbamazepine usage) cost-effectively, whereas the

developed countries are more dependent on the expensive alternative

for all approaches. More comprehensive phenotype specific chips which

could potentially test for multiple mutations leading to an adverse drug

reaction phenotype, or a co-morbidity testing platform can offer one stop

test to clinicians to be able to prescribe an effective treatment right away,

looking attractive on the cost-effectiveness parameter as well.

In recent years, the greater focus on effective application of health

technology and budget constraints has led to an increase in its

application. Along with countries in Europe, North America & Britain,

where traditionally HTA has been used, there has been increased

emphasis in Asian and other developing countries on its applications.

The recent HTA pilot program started from April 2016 in Japan, inclusion

of HTA system developments under the Healthy China 2030 Plan with a

focus of development of hubs and national policy documents for HTA,

incorporation in PhilHealth in Philippines (a national health insurance

which covers >92% of the population), increased interest by South Korea

and Japan to include it for evaluation of medical devices are some of the

many such initiatives.

140

Even though health technology assessment is one of the best scientific

tools at the service of decision-makers, it faces many challenges163-165.

Lack of understanding for the establishment’s need and structure among

the senior authorities, need of co-ordination among the many

stakeholders, absence of a systematic system of collecting evidence to

compare and an inherent shortage of experts and academic excellence

centres are among the prominent few. Different stakeholders may hold

different views regarding the specific nature of health problems,

interpretation of our moral commitments and also methodologies to

collect reliable information. The available evidence for health benefits

may not always be straight-forward. Though randomized clinic trials are

considered the gold standard for evidence of clinical efficiency, they may

sometimes be inconclusive or susceptible to biases and other sources

like observational studies, routine data, reference sources and expert

opinions may provide important contextual information. In addition, an

enhanced understanding of HTA processes and improved co-ordination

at the policy-making level are significant to facilitate its application in

practice.

The advancements and innovation in healthcare, presents this decade

with enormous opportunities and challenges and Health Technology

Assessment would play a significant role to inform and drive decision-

making and assist to achieve an improved sustainable healthcare setup

for all.

141

Bibliography

1. Finishing the euchromatic sequence of the human genome. Nature

2004;431:931-45.

2. Kim Y, Jun JK, Choi KS, Lee HY, Park EC. Overview of the National

Cancer screening programme and the cancer screening status in Korea. Asian

Pacific journal of cancer prevention : APJCP 2011;12:725-30.

3. Yamaguchi K. Overview of Cancer Control Programs in Japan.

Japanese Journal of Clinical Oncology 2002;32:S22-S31.

4. Monden M. [The basic plan to promote cancer control in Japan]. Gan

to kagaku ryoho Cancer & chemotherapy 2013;40:559-64.

5. Sanjay ST, Fu G, Dou M, et al. Biomarker detection for disease

diagnosis using cost-effective microfluidic platforms. The Analyst

2015;140:7062-81.

6. Lee JW, Figeys D, Vasilescu J. Biomarker assay translation from

discovery to clinical studies in cancer drug development: quantification of

emerging protein biomarkers. Advances in cancer research 2007;96:269-98.

7. Erichsen HC, Chanock SJ. SNPs in cancer research and treatment.

British journal of cancer 2004;90:747-51.

8. Squassina A, Manchia M, Manolopoulos VG, et al. Realities and

expectations of pharmacogenomics and personalized medicine: impact of

translating genetic knowledge into clinical practice. Pharmacogenomics

2010;11:1149-67.

9. Whirl-Carrillo M, McDonagh EM, Hebert JM, et al.

Pharmacogenomics knowledge for personalized medicine. Clinical

pharmacology and therapeutics 2012;92:414-7.

10. Drug Labels. Pharmgkb.org, 2017. (Accessed 20th April, 2017, at

https://www.pharmgkb.org/view/drug-labels.do.)

11. Klein TE, Altman RB, Eriksson N, et al. Estimation of the warfarin

dose with clinical and pharmacogenetic data. The New England journal of

medicine 2009;360:753-64.

12. Chan SL, Suo C, Lee SC, Goh BC, Chia KS, Teo YY. Translational

aspects of genetic factors in the prediction of drug response variability: a case

study of warfarin pharmacogenomics in a multi-ethnic cohort from Asia. The

pharmacogenomics journal 2012;12:312-8.

13. Marsh S, Hoskins JM. Irinotecan pharmacogenomics.

Pharmacogenomics 2010;11:1003-10.

142

14. Brown JT, Bishop JR. Atomoxetine pharmacogenetics: associations

with pharmacokinetics, treatment response and tolerability.

Pharmacogenomics 2015;16:1513-20.

15. Sultana J, Cutroneo P, Trifirò G. Clinical and economic burden of

adverse drug reactions. Journal of Pharmacology & Pharmacotherapeutics

2013;4:S73-S7.

16. Shepherd G, Mohorn P, Yacoub K, May DW. Adverse drug reaction

deaths reported in United States vital statistics, 1999-2006. The Annals of

pharmacotherapy 2012;46:169-75.

17. Park WB, Choe PG, Song KH, et al. Should HLA-B*5701 screening

be performed in every ethnic group before starting abacavir? Clinical

infectious diseases : an official publication of the Infectious Diseases Society

of America 2009;48:365-7.

18. Rauch A, Nolan D, Martin A, McKinnon E, Almeida C, Mallal S.

Prospective genetic screening decreases the incidence of abacavir

hypersensitivity reactions in the Western Australian HIV cohort study.

Clinical infectious diseases : an official publication of the Infectious Diseases

Society of America 2006;43:99-102.

19. Mallal S, Phillips E, Carosi G, et al. HLA-B*5701 Screening for

Hypersensitivity to Abacavir. New England Journal of Medicine

2008;358:568-79.

20. Charland SL, Agatep BC, Herrera V, et al. Providing patients with

pharmacogenetic test results affects adherence to statin therapy: results of the

Additional KIF6 Risk Offers Better Adherence to Statins (AKROBATS) trial.

The pharmacogenomics journal 2014;14:272-80.

21. Fung B. The $289 Billion Cost of Medication Noncompliance, and

What to Do About It. The Atlantic 2012 Sep12, 2012.

22. Whitney K. PREDICT helps pinpoint right statin for patient. Reporter

2012.

23. Moja L, Tagliabue L, Balduzzi S, et al. Trastuzumab containing

regimens for early breast cancer. The Cochrane database of systematic reviews

2012:CD006243.

24. Dong D, Sung C, Finkelstein EA. Cost-effectiveness of HLA-B*1502

genotyping in adult patients with newly diagnosed epilepsy in Singapore.

Neurology 2012;79:1259-67.

25. Rattanavipapong W, Koopitakkajorn T, Praditsitthikorn N,

Mahasirimongkol S, Teerawattananon Y. Economic evaluation of HLA-

143

B*15:02 screening for carbamazepine-induced severe adverse drug reactions

in Thailand. Epilepsia 2013;54:1628-38.

26. Tiamkao S, Jitpimolmard J, Sawanyawisuth K, Jitpimolmard S. Cost

minimization of HLA-B*1502 screening before prescribing carbamazepine in

Thailand. International journal of clinical pharmacy 2013;35:608-12.

27. Schackman BR, Scott CA, Walensky RP, Losina E, Freedberg KA,

Sax PE. The cost-effectiveness of HLA-B*5701 genetic screening to guide

initial antiretroviral therapy for HIV. Aids 2008;22:2025-33.

28. Kapoor R, Martinez-Vega R, Dong D, et al. Reducing hypersensitivity

reactions with HLA-B*5701 genotyping before abacavir prescription:

clinically useful but is it cost-effective in Singapore? Pharmacogenetics and

genomics 2014.

29. Nieves Calatrava D, Calle-Martin Ode L, Iribarren-Loyarte JA, et al.

Cost-effectiveness analysis of HLA-B*5701 typing in the prevention of

hypersensitivity to abacavir in HIV+ patients in Spain. Enfermedades

infecciosas y microbiologia clinica 2010;28:590-5.

30. Health sciences authority MoH, Singapore. Dear Healthcare

Professional Letter: Recommendations for HLA-B*1502 genotype testing

prior to initiation of carbamazepine in new patients. 2013.

31. Shotelersuk V, Limwongse C, Mahasirimongkol S. Genetics and

genomics in Thailand: challenges and opportunities. Molecular Genetics &

Genomic Medicine 2014;2:210-6.

32. Hershfield MS, Callaghan JT, Tassaneeyakul W, et al. Clinical

Pharmacogenetics Implementation Consortium guidelines for human

leukocyte antigen-B genotype and allopurinol dosing. Clinical pharmacology

and therapeutics 2013;93:153-8.

33. Crews KR, Gaedigk A, Dunnenberger HM, et al. Clinical

Pharmacogenetics Implementation Consortium (CPIC) guidelines for codeine

therapy in the context of cytochrome P450 2D6 (CYP2D6) genotype. Clinical

pharmacology and therapeutics 2012;91:321-6.

34. Anderson JL, Horne BD, Stevens SM, et al. Randomized trial of

genotype-guided versus standard warfarin dosing in patients initiating oral

anticoagulation. Circulation 2007;116:2563-70.

35. Pilote L, Lauzon C, Huynh T, et al. Quality of life after acute

myocardial infarction among patients treated at sites with and without on-site

availability of angiography. Archives of internal medicine 2002;162:553-9.

36. Ramsey LB, Johnson SG, Caudle KE, et al. The clinical

pharmacogenetics implementation consortium guideline for SLCO1B1 and

144

simvastatin-induced myopathy: 2014 update. Clinical pharmacology and

therapeutics 2014;96:423-8.

37. The Medication Safety Code Initiative. 2013. at http://safety-

code.org/.)

38. Sukasem C, Chantratita W. A success story in pharmacogenomics:

genetic ID card for SJS/TEN. Pharmacogenomics 2016;17:455-8.

39. Kozyra M, Ingelman-Sundberg M, Lauschke VM. Rare genetic

variants in cellular transporters, metabolic enzymes, and nuclear receptors can

be important determinants of interindividual differences in drug response.

Genet Med 2017;19:20-9.

40. Surendiran A, Pradhan SC, Adithan C. Role of pharmacogenomics in

drug discovery and development. Indian Journal of Pharmacology

2008;40:137-43.

41. Caudle KE, Klein TE, Hoffman JM, et al. Incorporation of

pharmacogenomics into routine clinical practice: the Clinical

Pharmacogenetics Implementation Consortium (CPIC) guideline development

process. Current drug metabolism 2014;15:209-17.

42. Relling MV, McDonagh EM, Chang T, et al. Clinical

Pharmacogenetics Implementation Consortium (CPIC) guidelines for

rasburicase therapy in the context of G6PD deficiency genotype. Clinical

pharmacology and therapeutics 2014;96:169-74.

43. Johnson JA, Weitzel KW. Advancing Pharmacogenomics as a

Component of Precision Medicine: How, Where, and Who? Clinical

Pharmacology & Therapeutics 2016;99:154-6.

44. Kisor DF, Bright DR, Conaway M, Bouts BA, Gerschutz GP.

Pharmacogenetics in the Community Pharmacy. Journal of Pharmacy Practice

2014;27:416-9.

45. Bunting BA, Smith BH, Sutherland SE. The Asheville Project: Clinical

and economic outcomes of a community-based long-term medication therapy

management program for hypertension and dyslipidemia. Journal of the

American Pharmacists Association 2008;48:23-31.

46. Kalista T, Lemay V, Cohen L. Postdischarge community pharmacist-

provided home services for patients after hospitalization for heart failure.

Journal of the American Pharmacists Association : JAPhA 2015;55:438-42.

47. Ferreri SP, Greco AJ, Michaels NM, et al. Implementation of a

pharmacogenomics service in a community pharmacy. Journal of the

American Pharmacists Association : JAPhA 2014;54:172-80.

145

48. Kimmel SE, French B, Kasner SE, et al. A Pharmacogenetic versus a

Clinical Algorithm for Warfarin Dosing. New England Journal of Medicine

2013;369:2283-93.

49. Baker WL, Chamberlin KW. New oral anticoagulants vs. warfarin

treatment: no need for pharmacogenomics? Clinical pharmacology and

therapeutics 2014;96:17-9.

50. Prince AER, Roche MI. Genetic Information, Non-Discrimination, and

Privacy Protections in Genetic Counseling Practice. Journal of genetic

counseling 2014;23:891-902.

51. Hall MA, Rich SS. Patients' fear of genetic discrimination by health

insurers: the impact of legal protections. Genet Med 2000;2:214-21.

52. Hall MA, McEwen JE, Barton JC, et al. Concerns in a primary care

population about genetic discrimination by insurers. Genet Med 2005;7:311-6.

53. Rodriguez-Antona C, Taron M. Pharmacogenomic biomarkers for

personalized cancer treatment. Journal of internal medicine 2014.

54. FDA. Information for Healthcare Professionals: Abacavir (marketed as

Ziagen) and Abacavir-Containing Medications. 2008.

55. FDA. Information for Healthcare Professionals: Dangerous or Even

Fatal Skin Reactions - Carbamazepine (marketed as Carbatrol, Equetro,

Tegretol, and generics).

http://wwwfdagov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPat

ientsandProviders/ucm124718htm 2007.

56. Hughes DA, Vilar FJ, Ward CC, Alfirevic A, Park BK, Pirmohamed

M. Cost-effectiveness analysis of HLA B*5701 genotyping in preventing

abacavir hypersensitivity. Pharmacogenetics 2004;14:335-42.

57. Hughes S. Black Box for Warfarin. Medscape Medical News 2006.

58. FDA. FDA safety information for Plavix (clopidogrel bisulfate) tablet.

http://wwwfdagov/Safety/MedWatch/SafetyInformation/ucm225843htm 2010.

59. FDA Drug Safety Communication: New restrictions, contraindications,

and dose limitations for Zocor (simvastatin) to reduce the risk of muscle

injury. December 15, 2011. at

http://www.fda.gov/Drugs/DrugSafety/ucm256581.htm.)

60. Wilke RA, Ramsey LB, Johnson SG, et al. The Clinical

Pharmacogenomics Implementation Consortium: CPIC Guideline for

SLCO1B1 and Simvastatin-Induced Myopathy. Clinical pharmacology and

therapeutics 2012;92:112-7.

146

61. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke

statistics--2015 update: a report from the American Heart Association.

Circulation 2015;131:e29-322.

62. Nwankwo T, Yoon SS, Burt V, Gu Q. Hypertension among adults in

the United States: National Health and Nutrition Examination Survey, 2011-

2012. NCHS data brief 2013:1-8.

63. Roger VL, Go AS, Lloyd-Jones DM, et al. Heart disease and stroke

statistics--2012 update: a report from the American Heart Association.

Circulation 2012;125:e2-e220.

64. High Cholesterol Facts. (Accessed September 19, 2015, at

http://www.cdc.gov/cholesterol/facts.htm.)

65. Law M, Rudnicka AR. Statin safety: a systematic review. Am J

Cardiol 2006;97:52C-60C.

66. Stewart A. SLCO1B1 Polymorphisms and Statin-Induced Myopathy.

PLoS currents 2013;5.

67. NEHI 2011. Thinking Outside the Pillbox: A System-wide Approach

to Improving

Patient Medication Adherence for Chronic Disease.

68. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin

Use in America and Gaps in Patient Education (USAGE): an internet-based

survey of 10,138 current and former statin users. Journal of clinical lipidology

2012;6:208-15.

69. Zhang H, Plutzky J, Skentzos S, et al. Discontinuation of statins in

routine care settings: a cohort study. Annals of internal medicine

2013;158:526-34.

70. The USAGE Survey Fact Sheet. 2014. at statinusage.com.)

71. Cost effectiveness and strategic planning (WHO-CHOICE). (Accessed

11 Jun, 2015, at http://www.who.int/choice/costs/CER_thresholds/en/.)

72. Johnson JA, Gong L, Whirl-Carrillo M, et al. Clinical

Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and

VKORC1 genotypes and warfarin dosing. Clinical pharmacology and

therapeutics 2011;90:625-9.

73. Coumadin® tablets (Warfarin sodium tablets, USP) Crystalline

coumadin® for injection (Warfarin sodium for injection, USP) [package

insert]. 2011 at http://dailymed.nlm.nih.gov/dailymed/lookup

.cfm?setid=c0f238ff-2804-4150-9766-22a5111b4e74.)

147

74. CPIC Dosing Guideline for warfarin and CYP2C9, VKORC1. at

https://www.pharmgkb.org/guideline/PA166104949.)

75. World Health Organization. Consolidated guidelines on the use of

antiretroviral drugs for treating and preventing HIV infection. In: HIV AIDS

program, ed. June 2013 edJune 2013.

76. Clay PG. The abacavir hypersensitivity reaction: A review.

2002;24:1502-514.

77. Sax PE, Tierney C, Collier AC, et al. Abacavir/lamivudine versus

tenofovir DF/emtricitabine as part of combination regimens for initial

treatment of HIV: final results. The Journal of infectious diseases

2011;204:1191-201.

78. Tan DH, Chan K, Raboud J, et al. Comparison of abacavir/lamivudine

and tenofovir/emtricitabine among treatment-naive HIV-infected patients

initiating therapy. Journal of acquired immune deficiency syndromes (1999)

2011;58:38-46.

79. Hewitt RG. Abacavir hypersensitivity reaction. Clinical infectious

diseases : an official publication of the Infectious Diseases Society of America

2002;34:1137-42.

80. Hughes CA, Foisy MM, Dewhurst N, et al. Abacavir hypersensitivity

reaction: an update. The Annals of pharmacotherapy 2008;42:387-96.

81. Mallal S, Nolan D, Witt C, et al. Association between presence of

HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1

reverse-transcriptase inhibitor abacavir. Lancet 2002;359:727-32.

82. Schackman BR, Gebo KA, Walensky RP, et al. The lifetime cost of

current human immunodeficiency virus care in the United States. Medical care

2006;44:990-7.

83. Tey JS, Ang LW, Tay J, et al. Determinants of late-stage HIV disease

at diagnosis in Singapore, 1996 to 2009. Annals of the Academy of Medicine,

Singapore 2012;41:194-9.

84. Ministry of Health Singapore. HIV Statistics. 2012.

85. Escaut L, Liotier JY, Albengres E, Cheminot N, Vittecoq D. Abacavir

rechallenge has to be avoided in case of hypersensitivity reaction. Aids

1999;13:1419-20.

86. Guidry JA UL, Yi O. HIV and aging: emerging issues in the HAART

era. LGBTQ policy journal at harvard kennedy school

87. Kauf TL, Roskell N, Shearer A, et al. A predictive model of health

state utilities for HIV patients in the modern era of highly active antiretroviral

148

therapy. Value in health : the journal of the International Society for

Pharmacoeconomics and Outcomes Research 2008;11:1144-53.

88. Dodek P, Phillips P. Questionable history of immediate-type

hypersensitivity to penicillin in Staphylococcal endocarditis: treatment based

on skin-test results vers-us empirical alternative treatment--A decision

analysis. Clinical infectious diseases : an official publication of the Infectious

Diseases Society of America 1999;29:1251-6.

89. Pepper PV, Owens DK. Cost-Effectiveness of the Pneumococcal

Vaccine in Healthy Younger Adults. Medical Decision Making 2002;22:S45-

S57.

90. Freedberg KA, Losina E, Weinstein MC, et al. The Cost Effectiveness

of Combination Antiretroviral Therapy for HIV Disease. New England Journal

of Medicine 2001;344:824-31.

91. Hetherington S, McGuirk S, Powell G, et al. Hypersensitivity reactions

during therapy with the nucleoside reverse transcriptase inhibitor abacavir.

Clinical therapeutics 2001;23:1603-14.

92. Eron J, Jr., Yeni P, Gathe J, Jr., et al. The KLEAN study of

fosamprenavir-ritonavir versus lopinavir-ritonavir, each in combination with

abacavir-lamivudine, for initial treatment of HIV infection over 48 weeks: a

randomised non-inferiority trial. Lancet 2006;368:476-82.

93. Pillai NE, Okada Y, Saw WY, et al. Predicting HLA alleles from high-

resolution SNP data in three Southeast Asian populations. Human molecular

genetics 2014;23:4443-51.

94. Teo YY, Sim X, Ong RT, et al. Singapore Genome Variation Project: a

haplotype map of three Southeast Asian populations. Genome research

2009;19:2154-62.

95. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and

mortality worldwide: sources, methods and major patterns in GLOBOCAN

2012. International journal of cancer 2015;136:E359-86.

96. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015.

CA: a cancer journal for clinicians 2016;66:115-32.

97. Shin A, Kim J, Park S. Gastric Cancer Epidemiology in Korea. Journal

of Gastric Cancer 2011;11:135-40.

98. Office NRoD. Cancer incidence and mortality 2003 - 2012 and

selected trends 1973 - 2012 in Singapore2015.

99. Lin Y, Ueda J, Kikuchi S, et al. Comparative epidemiology of gastric

cancer between Japan and China. World Journal of Gastroenterology : WJG

2011;17:4421-8.

149

100. Kim Y, Park J, Nam B-H, Ki M. Stomach cancer incidence rates

among Americans, Asian Americans and Native Asians from 1988 to 2011.

Epidemiology and Health 2015;37:e2015006.

101. GLOBOCAN 2012: Estimated Cancer Incidence, Mortality, and

Prevalence Worldwide in 2012. 2012. (Accessed 7 July, 2015, at

http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx.)

102. Koong HN, Chan HS, Nambiar R, et al. Gastric cancers in Singapore:

poor prognosis arising from late presentation. The Australian and New

Zealand journal of surgery 1996;66:813-5.

103. Tsubono Y, Nishino Y, Tsuji I, Hisamichi S. Screening for Gastric

Cancer in Miyagi, Japan: Evaluation with a Population-Based Cancer

Registry. Asian Pacific journal of cancer prevention : APJCP 2000;1:57-60.

104. Lello E, Furnes B, Edna TH. Short and long-term survival from gastric

cancer. A population-based study from a county hospital during 25 years. Acta

oncologica (Stockholm, Sweden) 2007;46:308-15.

105. Chau I, Norman AR, Cunningham D, Waters JS, Oates J, Ross PJ.

Multivariate prognostic factor analysis in locally advanced and metastatic

esophago-gastric cancer--pooled analysis from three multicenter, randomized,

controlled trials using individual patient data. Journal of clinical oncology :

official journal of the American Society of Clinical Oncology 2004;22:2395-

403.

106. Age-Standardised One-, Five- and Ten-Year Net Survival, Adults

(Aged 15-99), England and Wales, 2010-2011. Cancer Research UK, 2014.

(Accessed 15th March, 2017,

107. Trends in Cancer Survival 1983-2007. ISD Scotland 2010, 2010.

(Accessed 15th March 2017,

108. Howlader N NA, Krapcho M, Miller D, Bishop K, Altekruse SF,

Kosary CL, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS,

Feuer EJ, Cronin KA (eds). SEER Cancer Statistics Review, 1975-2013:

National Cancer Institute; 2015.

109. Choi KS, Jun JK, Suh M, et al. Effect of endoscopy screening on stage

at gastric cancer diagnosis: results of the National Cancer Screening

Programme in Korea. British journal of cancer 2015;112:608-12.

110. Cancer Statistics in Japan-2015 Tokyo: Foundation for Promotion of

Cancer Research; 2015.

111. Lee S, Jun JK, Suh M, et al. Gastric Cancer Screening Uptake Trends

in Korea: Results for the National Cancer Screening Program From 2002 to

2011: A Prospective Cross-Sectional Study. Medicine 2015;94:e533.

150

112. Trends in Cancer Incidence in Singapore 2009-2013. 2014. (Accessed

7 July, 2015, at https://www.nrdo.gov.sg/docs/librariesprovider3/Publications-

Cancer/cancer-trends-report-2009-

2013.pdf?sfvrsn=0&AspxAutoDetectCookieSupport=1.)

113. Amal H, Leja M, Funka K, et al. Detection of precancerous gastric

lesions and gastric cancer through exhaled breath. Gut 2016;65:400.

114. Jung K-W, Won Y-J, Kong H-J, Oh C-M, Seo HG, Lee J-S. Cancer

Statistics in Korea: Incidence, Mortality, Survival and Prevalence in 2010.

Cancer Res Treat 2013;45:1-14.

115. Life expectancy at birth. 2017,

116. Roukos DH, Lorenz M, Karakostas K, Paraschou P, Batsis C, Kappas

AM. Pathological serosa and node-based classification accurately predicts

gastric cancer recurrence risk and outcome, and determines potential and

limitation of a Japanese-style extensive surgery for Western patients: a

prospective with quality control 10-year follow-up study. British journal of

cancer 2001;84:1602-9.

117. Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance

from the median, range, and the size of a sample. BMC medical research

methodology 2005;5:13.

118. Tsukuma H, Oshima A, Narahara H, Morii T. Natural history of early

gastric cancer: a non-concurrent, long term, follow up study. Gut

2000;47:618-21.

119. Zhou HJ, So JB, Yong WP, et al. Validation of the functional

assessment of cancer therapy-gastric module for the Chinese population.

Health and quality of life outcomes 2012;10:145.

120. Zhou HJ. Health Services Research on Endoscopic Surveillance for

Gastric Cancer in the Singapore Chinese Population - Experience of the

Gastric Cancer Epidemiology Clinical and Genetic Program: National

University of Singapore; 2013.

121. Voutilainen ME, Juhola MT. Evaluation of the diagnostic accuracy of

gastroscopy to detect gastric tumours: clinicopathological features and

prognosis of patients with gastric cancer missed on endoscopy. European

journal of gastroenterology & hepatology 2005;17:1345-9.

122. Hamashima C, Okamoto M, Shabana M, Osaki Y, Kishimoto T.

Sensitivity of endoscopic screening for gastric cancer by the incidence

method. International journal of cancer 2013;133:653-9.

123. (2012) OfEC-oaD. Total population by age and sex. OECD.stat2012.

151

124. Bank TW. Life expectancy at birth, total (years). The World Bank;

2016.

125. Lee HY, Park EC, Jun JK, Choi KS, Hahm MI. Comparing upper

gastrointestinal X-ray and endoscopy for gastric cancer diagnosis in Korea.

World J Gastroenterol 2010;16:245-50.

126. Kim SS, Kim J-H, Cheung DY, Park S-H. Analysis of medical

expenses according to the stage of gastric cancer during the first year after

diagnosis. Journal of Clinical Oncology 2014;32:4-.

127. Department of Statistics S. Population Trends 20162016.

128. WHO. Cardiovascular disease (CVDs) factsheet.

http://wwwwhoint/mediacentre/factsheets/fs317/en/ 2015.

129. Ohsfeldt RL, Gandhi SK, Fox KM, McKenney JM. Statin cost-

effectiveness comparisons using real-world effectiveness data: formulary

implications. Value in health : the journal of the International Society for

Pharmacoeconomics and Outcomes Research 2008;11:1061-9.

130. Ministry of Health Singapore. National Health Survey 2010. 2010.

131. Ward S, Lloyd Jones M, Pandor A, et al. A systematic review and

economic evaluation of statins for the prevention of coronary events. Health

technology assessment (Winchester, England) 2007;11:1-160, iii-iv.

132. Christensen K, Doblhammer G, Rau R, Vaupel JW. Ageing

populations: the challenges ahead. Lancet 2009;374:1196-208.

133. Fernandez G, Spatz ES, Jablecki C, Phillips PS. Statin myopathy: a

common dilemma not reflected in clinical trials. Cleveland Clinic journal of

medicine 2011;78:393-403.

134. Zhou Z, Rahme E, Abrahamowicz M, et al. Effectiveness of statins for

secondary prevention in elderly patients after acute myocardial infarction: an

evaluation of class effect. CMAJ : Canadian Medical Association Journal

2005;172:1187-94.

135. Sakamoto T, Kojima S, Ogawa H, et al. Effects of early statin

treatment on symptomatic heart failure and ischemic events after acute

myocardial infarction in Japanese. Am J Cardiol 2006;97:1165-71.

136. Kadota S, Matsuda M, Izuhara M, et al. Long-term effects of early

statin therapy for patients with acute myocardial infarction treated with stent

implantation. Journal of Cardiology 2008;51:171-8.

137. Wei L, Wang J, Thompson P, Wong S, Struthers AD, MacDonald TM.

Adherence to statin treatment and readmission of patients after myocardial

infarction: a six year follow up study. Heart 2002;88:229-33.

152

138. Joy TR, Hegele RA. Narrative review: statin-related myopathy. Annals

of internal medicine 2009;150:858-68.

139. Wagner M, Muhldorfer-Fodor M, Prommersberger KJ, Schmitt R.

Statin-induced focal myositis of the upper extremity. A report of two cases.

European journal of radiology 2011;77:258-60.

140. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild to moderate

muscular symptoms with high-dosage statin therapy in hyperlipidemic

patients--the PRIMO study. Cardiovascular drugs and therapy / sponsored by

the International Society of Cardiovascular Pharmacotherapy 2005;19:403-14.

141. Kuncova K, Sedlackova M, Vencovsky J, et al. Inflammatory

myopathy associated with statins: report of three cases. Modern rheumatology

/ the Japan Rheumatism Association 2014;24:366-71.

142. Rallidis LS, Fountoulaki K, Anastasiou-Nana M. Managing the

underestimated risk of statin-associated myopathy. International journal of

cardiology 2012;159:169-76.

143. Tomlinson SS, Mangione KK. Potential adverse effects of statins on

muscle. Physical therapy 2005;85:459-65.

144. Voora D, Shah SH, Spasojevic I, et al. The SLCO1B1*5 genetic

variant is associated with statin-induced side effects. Journal of the American

College of Cardiology 2009;54:1609-16.

145. Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-

induced myopathy--a genomewide study. The New England journal of

medicine 2008;359:789-99.

146. NRoD. O. Trends in Acute Myocardial Infarction in Singapore 2007 -

2013. Ministry of Health, Singapore.

147. Hansen KE, Hildebrand JP, Ferguson EE, Stein JH. Outcomes in 45

patients with statin-associated myopathy. Archives of internal medicine

2005;165:2671-6.

148. Smolina K, Wright FL, Rayner M, Goldacre MJ. Long-term survival

and recurrence after acute myocardial infarction in England, 2004 to 2010.

Circulation Cardiovascular quality and outcomes 2012;5:532-40.

149. Suzuki S, Takaki H, Yasumura Y, et al. Assessment of quality of life

with 5 different scales in patients participating in comprehensive cardiac

rehabilitation after acute myocardial infarction. Circulation journal : official

journal of the Japanese Circulation Society 2005;69:1527-34.

150. Meek C, Wierzbicki AS, Jewkes C, et al. Daily and intermittent

rosuvastatin 5 mg therapy in statin intolerant patients: an observational study.

Current medical research and opinion 2012;28:371-8.

153

151. Florentin M, Liberopoulos EN, Elisaf MS. Ezetimibe-associated

adverse effects: what the clinician needs to know. International journal of

clinical practice 2008;62:88-96.

152. (UK) NIfHaCE. Lipid Modification: Cardiovascular Risk Assessment

and the Modification of Blood Lipids for the Primary and Secondary

Prevention of Cardiovascular Disease. London: National Clinical Guideline

Centre (UK); 2014 Jul.

153. Annual GDP at Current Market Prices. (Accessed Jun 11, 2015, at

http://www.singstat.gov.sg/statistics/browse-by-theme/national-accounts.)

154. Derosa G, D'Angelo A, Franzetti IG, et al. Efficacy and safety of

ezetimibe/simvastatin association on non-diabetic and diabetic patients with

polygenic hypercholesterolemia or combined hyperlipidemia and previously

intolerant to standard statin treatment. Journal of clinical pharmacy and

therapeutics 2009;34:267-76.

155. Magni P, Macchi C, Morlotti B, Sirtori CR, Ruscica M. Risk

identification and possible countermeasures for muscle adverse effects during

statin therapy. European journal of internal medicine 2015.

156. Ministry of Health S. Total Hospital Bills - By condition/procedure.

www.moh.gov.sg: Ministry of Health, Singapore; 2016.

157. Xu Cong Ruan YHK, Joanne Chang. Prevalence of statin-induced

rhabdomyolysis in Asian populations. ACCP Annual Meeting.

Pharmacotherapy 2012;10.

158. Johansen KL, Smith MW, Unruh ML, Siroka AM, O'Connor TZ,

Palevsky PM. Predictors of health utility among 60-day survivors of acute

kidney injury in the Veterans Affairs/National Institutes of Health Acute Renal

Failure Trial Network Study. Clinical journal of the American Society of

Nephrology : CJASN 2010;5:1366-72.

159. Schaefer C, Mann R, Masters ET, et al. The Comparative Burden of

Chronic Widespread Pain and Fibromyalgia in the United States. Pain Practice

2015:n/a-n/a.

160. Henriksson C, Larsson M, Herlitz J, Karlsson JE, Wernroth L, Lindahl

B. Influence of health-related quality of life on time from symptom onset to

hospital arrival and the risk of readmission in patients with myocardial

infarction. Open heart 2014;1:e000051.

161. Bays HE, Ose L, Fraser N, et al. A multicenter, randomized, double-

blind, placebo-controlled, factorial design study to evaluate the lipid-altering

efficacy and safety profile of the ezetimibe/simvastatin tablet compared with

ezetimibe and simvastatin monotherapy in patients with primary

hypercholesterolemia. Clinical therapeutics 2004;26:1758-73.

154

162. ablBirkemeyer R, Dauch A, Müller A, et al. Short term cost

effectiveness of a regional myocardial infarction network. Health Economics

Review 2013;3:10-.

163. Olyaeemanesh A, Doaee S, Mobinizadeh M, Nedjati M, Aboee P,

Emami-Razavi SH. Health technology assessment in Iran: challenges and

views. Medical Journal of the Islamic Republic of Iran 2014;28:157-.

164. Simard C, Pharm B, Poirier P. Ezetimibe-associated myopathy in

monotherapy and in combination with a 3-hydroxy-3-methylglutaryl

coenzyme A reductase inhibitor. The Canadian journal of cardiology

2006;22:141-4.

165. Abrams HL, Hessel S. Health technology assessment: problems and

challenges. AJR American journal of roentgenology 1987;149:1127-32.