18,909
Views
35
CrossRef citations to date
0
Altmetric
Perspective

Fixed dose drug combinations – are they pharmacoeconomically sound? Findings and implications especially for lower- and middle-income countries

ORCID Icon, , ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , , , , ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, , , , ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, , , ORCID Icon, ORCID Icon, ORCID Icon, , , ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, , ORCID Icon & ORCID Icon show all
Pages 1-26 | Received 17 Jan 2020, Accepted 21 Feb 2020, Published online: 01 Apr 2020

ABSTRACT

Introduction

There are positive aspects regarding the prescribing of fixed dose combinations (FDCs) versus prescribing the medicines separately. However, these have to be balanced against concerns including increased costs and their irrationality in some cases. Consequently, there is a need to review their value among lower- and middle-income countries (LMICs) which have the greatest prevalence of both infectious and noninfectious diseases and issues of affordability.

Areas covered

Review of potential advantages, disadvantages, cost-effectiveness, and availability of FDCs in high priority disease areas in LMICs and possible initiatives to enhance the prescribing of valued FDCs and limit their use where there are concerns with their value.

Expert commentary

FDCs are valued across LMICs. Advantages include potentially improved response rates, reduced adverse reactions, increased adherence rates, and reduced costs. Concerns include increased chances of drug:drug interactions, reduced effectiveness, potential for imprecise diagnoses and higher unjustified prices. Overall certain FDCs including those for malaria, tuberculosis, and hypertension are valued and listed in the country’s essential medicine lists, with initiatives needed to enhance their prescribing where currently low prescribing rates. Proposed initiatives include robust clinical and economic data to address the current paucity of pharmacoeconomic data. Irrational FDCs persists in some countries which are being addressed.

1. Introduction

Fixed dose combinations (FDCs) are defined as a combination of two or more active ingredients within a single form of pharmaceutical administration [Citation1Citation6]. They have been shown to appreciably reduce the risk of medication non-adherence, which is particularly important in patients with chronic diseases [Citation7]. However, their rationality for use should be based on sound medical principles as there have been concerns with their irrationality and utility in several countries [Citation1,Citation2,Citation4,Citation5,Citation8].

We will present concerns in more detail in Section 1.1. However, these concerns have to be balanced against the potential advantages of FDCs including their cost-effectiveness in certain situations. These advantages will be discussed in detail in Section 1.2 before debating potential ways forward to enhance the prescribing and funding of valued FDCs. Alongside this, limit authorization, utilization, and funding for medically irrational FDCs and/or FDCs of perceived limited value.

1.1. General concerns with FDCs especially among lower- and middle-income countries (LMICs)

Concerns with FDCs include potentially altering the optimal dosing of one or more of the components due to differences in pharmacokinetic profiles and half-lives of the various constituents [Citation1]. FDCs may also increase the chances of adverse drug reactions or drug:drug interactions due to the different profiles of the medicines in the FDC as well as not fully recognizing the differences that can occur in the pharmacogenetic profiles of patients during the development of FDCs [Citation1,Citation6,Citation9,Citation10]. Pharmacogenetic concerns are particularly important in FDCs when the components are either an essential part of the primary pathway for eliminating the medicines of interest or a critical step in their onset of action [Citation6]. The pharmacokinetic profiles of the constituents in FDCs are also important in patients with infectious diseases as there can be concerns with resistance development due to the combination [Citation11,Citation12]. Additionally, pharmacokinetic and pharmacodynamic considerations of the constituents are important in the elderly where safety profiles may be altered [Citation13]. Evidence has also shown that inappropriately manufactured FDCs can result in their reduced effectiveness or enhanced toxicity in routine clinical care as well as peak effectiveness at different times alongside concerns with their shelf life [Citation14].

Additional concerns with FDCs include potentially higher prices than the sum of the individual components unless justified, higher prices maintained with additional patent protection, difficulty in ascertaining which component is responsible for any side-effects that may arise, and patients may receive too little or too much of a specific ingredient due to challenges with dose adjustments. Besides this, FDCs can encourage an imprecise diagnosis especially for patients with infections and there can be a loss of effectiveness if patients forget to take their FDC as opposed to just one of the individual components [Citation1,Citation5,Citation6,Citation15Citation18]. There are futhermore concerns that FDCs may become too large impeding oral administration [Citation13,Citation19].

However, some of the perceived difficulties and concerns with FDCs can potentially be addressed through having multiple formulations available for titration purposes, starting FDCs only when deemed safe to do so, and/or addressing pharmacogenetic and pharmacokinetic concerns during FDC development [Citation5,Citation6,Citation20,Citation21]. Alongside this, look to re-formulate large FDCs with dissolving and other formulations [Citation13,Citation22]. The availability of multiple formulations was particularly important for FDCs containing inhaled corticosteroids (ICS) and long-acting beta-agonists (LABAs) for treating patients with asthma when health authorities, particularly among Western countries, were looking to reduce the doses of steroids in the ICS/LABA FDCs for long-term safety reasons [Citation23]. The lack of different formulations of ICS/LABA FDCs among the pharmaceutical companies promoting cheaper alternatives reduced their uptake initially until this was addressed [Citation23].

A number of these concerns led to the discontinuation of more than 90% of the FDCs marketed in the US in the 1960s and 1970s [Citation5,Citation24]. Following this, the US Food and Drug Administration (FDA) declared that any new FDC required proof of benefits versus the single components before approval, with similar initiatives in Europe [Citation15,Citation24]. In Europe, the revised regulations from the European Medicines Agency (EMA) stated that any proposed FDC should be based on robust and valid therapeutic principles with the potential advantages assessed in studies against potential disadvantages, and where possible for each dose of the medicines included in the FDC [Citation5,Citation15,Citation25]. Typically, the medicines in FDCs should have different mechanisms of action but their pharmacokinetics and/or pharmacodynamics should not be appreciably different as this will impact on their effectiveness in clinical care. In addition, the combination should not be additive in terms of increased toxicity [Citation1,Citation5,Citation25]. Following these regulatory changes, we have seen a growth in the number of FDCs available across countries. For instance, in Europe using 2009 as a baseline, there has been an 8% increase in FDCs approved by EMA in 2011 and a 15% increase in 2013, with this trend continuing [Citation5].

Stringent control measures are typically needed to reduce the availability of irrational FDCs, which has not always been the case [Citation1,Citation14,Citation26,Citation27]. In India, irrational FDCs were often made available by state authorities without prior approval of the Central Drugs Standard Control Organization [Citation28]. However, these concerns have now been recognized resulting in a recent ban on 328 FDCs in India [Citation29]. Whilst there have been concerns with the quality of generic medicines in Pakistan, there appeared to be no concerns with the bioavailability of for instance rifampicin in FDCs in Pakistan to treat patients with tuberculosis (TB) [Citation30,Citation31].

1.2. Potential advantages of FDCs

Potential advantages for FDCs compared with prescribing the components separately or where there are concerns with monotherapy alone include: (i) improved response rates where there is an inadequate response to monotherapy through for instance different mechanisms of action of the medicines in the combination, (ii) the combination of the medicines in the FDC achieves the desired effect more rapidly, (iii) the proposed FDC reduces toxicity with one medicine potentially counteracting the adverse reactions of another and (iv) the potential for combining doses that are sub-therapeutic when used as monotherapy because of issues such as safety as seen for instance with combination medicines for patients infected with human immunodeficiency virus (HIV), with these benefits often translating into lower costs of care [Citation3,Citation15,Citation25,Citation32Citation36].

FDCs also offer the possibility to simplify administration where a combination of active substances is already recognized as clinically important. As a result, seeking to improve adherence as well as targeting multiple disease pathways, improving efficiency, and potentially saving resources for patients and the healthcare system [Citation3,Citation7,Citation15,Citation17,Citation37Citation48]. However, this is not always the case as seen with FDCs for patients with HIV in France and Spain [Citation49].

FDCs can also help clinicians to effectively manage patient outcomes from the perspective of long-term care, allowing them to use combinations of active ingredients that are effective over time and can improve patient safety as seen for instance with FDCs for respiratory conditions and pain-management [Citation23, Citation50Citation56]. In addition, potentially reduce costs and improve the co-operation between physicians and patients [Citation17,Citation57], with savings enhanced by the availability of generic FDCs [Citation58]. Co-payment costs can also potentially be decreased with FDCs versus individual components, which is particularly important in lower- and middle-income countries (LMICs) where there are high co-payments [Citation59].

The World Health Organization (WHO) endorses FDCs particularly for infectious diseases such as HIV, malaria, tuberculosis (TB), and Hepatitis B to improve the effectiveness of treatments especially given the toxicity that can exist with antiretrovirals as well as help prevent resistance from developing [Citation15,Citation25,Citation60]. This is particularly important in sub-Saharan Africa with a high prevalence of both non-communicable diseases (NCDs) such as hypertension and diabetes along with infectious diseases including HIV, TB and malaria, and a high prevalence of patients with joint co-morbidities versus other continents [Citation15,Citation61Citation69]. Having said this, a recent Cochrane review suggested that there was no difference in outcomes with FDCs versus single-drug formulations combined in managing patients with newly diagnosed pulmonary TB [Citation70]. However, others have published different findings (Section 2. 6). In the case of new FDCs for patients with HIV, the dogma of effective antiretroviral therapy (ART) containing at least three active substances is being challenged by new data showing effectiveness with FDCs containing just two medicines, with costs helped by patent expiry and more generic formulations becoming available [Citation71]. Treatment costs can also increase in some settings with the prescribing of multiple medicines over patented FDCs due to higher rates of adverse effects [Citation72].

1.3. Aims and objectives

Given the controversies surrounding the use of FDCs, there is an urgent need to discuss both their advantages and disadvantages including their cost-effectiveness. This is particularly important in LMICs in view of their high prevalence of both infectious diseases and NCDs, their considerable resource pressures, and the continued growth in both morbidity and mortality from NCDs [Citation73Citation80]. However, different conclusions concerning the clinical and economic value of FDCs can be drawn from the different disease areas as well as within disease areas depending on the FDCs available within a country. There is also a perceived paucity of published data regarding the costs, value, and cost-effectiveness of FDCs across different disease areas in LMICs versus high-income countries, which needs to be addressed. Consequently, the principal focus of the findings and suggestions in this perspective paper is on the costs, value, and pharmacoeconomics of current and future FDCs aimed mainly at governments and their advisers in LMICs. However, patients also play a key role especially in LMICs where there can be high co-payments and predominant ‘out of pocket’ payments, and patients’ illness can have a catastrophic effect on the rest of the family [Citation81,Citation82].

An iterative process was used to develop this review paper building on pertinent publications known to the coauthors in both infectious diseases and NCDs. These publications were supplemented by suggested activities from the senior level coauthors from across countries and continents on potential ways forward to enhance the prescribing of valued FDCs. This reflects, as mentioned, the envisaged paucity of published pharmacoeconomic data on FDCs in LMICs.

We are aware of ongoing initiatives among several LMICs to strive toward universal health care (UHC) recognizing the challenges. Besides this, ongoing initiatives across LMICs to achieve agreed Sustainable Development Goals (SDGs). The SDGs include a reduction in morbidity and mortality associated with NCDs such as cardiovascular and respiratory diseases [Citation74,Citation83Citation86]. The availability of pertinent and affordable FDCs can potentially play a key role in achieving these goals alongside educational and lifestyle changes.

1.4. Methodology

We were aware that there have only been a limited number of publications assessing the value of FDCs in LMICs, with most publications typically involving higher-income countries We are also aware that the potential role and value of FDCs also vary between and within disease areas and populations as well as across countries. Consequently, we did not undertake a formal systematic review; however, based this perspective paper, including suggested future activities, on pertinent publications known to the senior level coauthors across multiple LMICs combined with their extensive experiences with FDCs to contextualize the findings.

This perspective paper will be divided into three parts to provide future direction. Firstly, we will briefly review the role and value of FDCs within and across key infectious and noninfectious diseases. Infectious diseases include HIV, TB, and malaria and noninfectious diseases include cardiovascular diseases (CVD) such as hypertension and type 2 diabetes mellitus (T2DM) as well as pain management and respiratory diseases. This also addresses the polypill for patients with CVD [Citation38,Citation40,Citation87].

The senior-level personnel involved in this paper come from a wide variety of backgrounds including government groups, academia, rational use medicine personnel, clinicians, and patient representative groups. A wide variety of LMICs have been included in this perspective paper in terms of their geography, population size, GDP per capita, and progress toward universal health care. We have used such approaches before to stimulate debate in priority disease areas to provide future guidance [Citation88Citation100]. The 2018 World Bank classification has been used to categorize countries into LMICs or upper-income countries [Citation101] wherever pertinent.

We will start with NCDs including CVD, diabetes, respiratory diseases, and pain management, before discussing the potential role and value of FDCs in patients with high priority infectious diseases including HIV, malaria, and TB. These disease areas are included as they are the subject of most publications regarding the pharmacoeconomics of FDCs across countries and they are the major source of morbidity and mortality within LMICs. We have not included FDCs for patients with Hepatitis C despite being listed in the WHO Essential Medicine List (EML) as a result of their considerable effectiveness and safety versus previous medicines since their prices can be prohibitive for countries and citizens without substantial discounts. This is exacerbated by pharmaceutical companies making up to 99.9% gross profit in some countries adding to the overall cost of medicines [Citation102Citation106]. Having said this, expenditure on new medicines for patients with Hepatitis C has been helped by the increasing availability of low-cost generics [Citation107], as well as treatments provided free or for limited costs in some countries; however, this is not universal among LMICs [Citation108,Citation109]. Similarly, we have not included topical FDCs for use in dermatology although we are aware that there are concerns with a number of these in LMICs such as India [Citation4,Citation110]. Lastly, we have excluded combination antibiotics such as amoxicillin with an enzyme inhibitor as these medicines should now be reserved under the recent WHO AWaRe list of antibiotics and there can be considerable concerns with their availability [Citation11,Citation12,Citation111Citation114]. We have though included sulfamethoxazole and trimethoprim FDC to help prevent Plasmodium falciparum (Pf) malaria and other infections despite initial concerns that this FDC would impact on the effectiveness of treatments such as sulfadoxine-pyrimethamine due to shared mechanisms of action and resistance pattern development [Citation115]. These concerns, however, have not materialized and this combination is now widely used in malaria-endemic countries as prophylaxis in both HIV-infected children and adults as well as those without HIV [Citation116,Citation117].

Secondly, we will document FDC availability within public health-care systems among a range of LMICs covering multiple countries and continents versus the latest WHO EML. This is because the WHO EML is recognized as a guide to the development of national and institutional EMLs with medicines selected for national lists with due regard to disease prevalence and public health relevance in a country as well as evidence of clinical efficacy and safety, comparative costs and cost-effectiveness in a country [Citation111,Citation118,Citation119].

We will also try and explain any variability in the availability of FDCs between countries as a prelude to lastly discussing their advantages and disadvantages as well as potential ways forward to enhance their prescribing where pertinent. These deliberations will be based on the perceived value, or lack of it, of FDCs across the chosen disease areas among the senior level coauthors. We will also seek to guide key stakeholder groups in LMICs going forward given the lack of pharmacoeconomic data to date to make future policy decisions. We will also describe potential measures to reduce the prescribing of FDCs where there are concerns. We have undertaken this approach to address the paucity of published studies addressing the pharmacoeconomics of FDCs in LMICs versus high-income countries despite the high prevalence of both infectious and noninfectious diseases in these countries.

2. Role and value of FDCs across disease areas including health economic evaluations

We will start with a review of the role and value of FDCs in patients with NCDs followed by high priority infectious disease areas including HIV, malaria, and TB. This will include a consolidation of studies that have been published in LMICs regarding the pharmacoeconomics of FDCs in these chosen disease areas.

2.1. Cardiovascular diseases (CVD)

Improved management of CVD is seen as critical with CVD a leading cause of morbidity and mortality globally as well as causing a high economic burden to health-care systems [Citation120,Citation121]. Reducing the morbidity and mortality due to CVD is particularly relevant in lower-income countries and areas with under-developed/equipped health-care systems where rates have increased in recent years exacerbated by changes in lifestyle, diet, and urbanization [Citation122Citation128].

Management strategies for CVD and hypertension largely involve encouraging changes in lifestyle as well as prescribing medicines from several pharmacological classes. Consequently, CVD and hypertension may be good candidates for the development and use of FDCs, with a number of FDCs currently in use globally.

Overall, FDCs are thought to be a potential solution to the high pill burden seen in some patients with CVDs, including those with hypertension, increasing adherence, and the clinical effectiveness of prescribed medicines [Citation20,Citation35,Citation37,Citation45,Citation57,Citation126,Citation129Citation139]. A key highlight of the 2018 European Society of Cardiology and European Society of Hypertension (ESC/ESH) guideline on the treatment of hypertension is the single-pill treatment strategy with the preferred use of a single pill combination for most patients to improve their blood pressure (BP) control [Citation135,Citation140]. FDCs also offer the potential to combine the additive effects of different treatment approaches without having to appreciably increase the dose of individual medicines, which could increase their side-effects and potentially decrease adherence in routine care [Citation37,Citation141,Citation142].

FDCs can also transform the management of patients with CVD including hypertension by reducing the need for titration and adding in different classes of medicines to help control patients’ BP [Citation143]. This is particularly important in LMICs where the costs of transport and loss of income attending health-care clinics can adversely affect their attendance and goal attainment [Citation144,Citation145], with medication follow-up visits among patients with CVDs already a major issue particularly in Africa [Citation144Citation146]. There are ongoing programs across LMICs to address concerns with adherence to medicines including instigating adherence clubs as well as educational and other programs for patients given concerns with the educational level of patients with CVD in a number of LMICs [Citation144,Citation145,Citation147Citation150]. FDCs can help with this as well as help reduce the potential for stock-outs by limiting the number of medicines that need to be available among primary health-care centers [Citation151]. Consequently, FDCs can be viewed positively by all key stakeholder groups including physicians, pharmacists, and patients [Citation143,Citation152,Citation153]. Other factors positively influencing the perception of FDCs include health authority policies toward FDCs and their costs if these are reduced especially when there are high co-payments [Citation152]. Their use, however, does vary between health-care systems for many reasons including clinician/patient preferences, their costs, and healthcare system approvals [Citation41,Citation154,Citation155]. There have also been differences in outcomes between FDCs for CVDs as well as when doses are missed [Citation156,Citation157].

The renin-angiotensin system (RAS) blockers are seen as a core component of FDCs for patients with hypertension except where they are contraindicated [Citation158,Citation159]. Typically, thiazide diuretics or calcium channel blockers (CCBs) should be used as first-line treatment especially in sub-Saharan Africa with RAS blockers – angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) – potentially added in as a single FDC [Citation35,Citation159Citation161]. However, ACE inhibitors have a more limited effect in reducing blood pressure in the black population and their prescribing needs to be more carefully managed in this population [Citation160,Citation162]. Diuretics can though enhance the effect of RAS blockers whilst minimizing their undesirable metabolic effects, whilst CCBs and RAS blockers have synergistic protective effects on the vascular wall and have been shown to be effective FDCs when combined improving adherence and outcomes [Citation159,Citation163,Citation164]. ACE inhibitors may also offset one of the major side effects associated with CCBs which is pedal edema [Citation165]. In any event, the management of patients with hypertension is a priority area in LMICs including sub-Saharan Africa with often patients needing two or more medicines following titration [Citation144,Citation160,Citation166,Citation167].

Studies assessing the costs and cost-effectiveness of FDCs in CVD have principally been performed in high-income countries. These include studies by Sherrill et al. (2011) demonstrating health-care costs were appreciably lower in the FDC group compared with those patients taking the medicines separately [Citation168]. A study in Canada also showed yearly medicine cost savings with FDCs [Citation169], and a study in Japan also showed significant medicine cost savings with FDCs versus patients taking multiple tablets [Citation170]. Other published studies involving high-income countries have also shown significantly lower costs for FDCs in CVD versus multiple tablets [Citation45,Citation171Citation174]. However, Deshmukh et al. (2017) in the US found the acquisition costs for FDCs were higher among patients being treated for their hypertension versus free-pill combinations although the higher costs were more than offset by lower inpatient costs [Citation43].

There can though be concerns if FDCs include combination drugs with similar mechanisms of action such as combining an ACE inhibitor with an ARB, which increases adverse events and costs without any obvious additional clinical benefits [Citation175]. In addition, we have seen some LMICs flooded in recent years with multiple FDC anti-hypertensive medicines not listed in the WHO EML and concerns with their rationality [Citation1,Citation36,Citation176]. Having said this, combinations of anti-hypertensive medicines are typically needed in LMICs, especially in sub-Saharan Africa, given the high prevalence of hypertension as well as resistant hypertension that can exist in these countries [Citation144,Citation149,Citation160,Citation167,Citation177]. However, the nature of the anti-hypertensive medicines in the various combinations can be important especially given, as mentioned, concerns with ACE inhibitors and ARBs among the black population [Citation160,Citation162]. A recent meta‐analysis showed that lowering BP by 10 mmHg resulted in a 20% reduction in the risk of major cardiovascular events. However, despite various anti-hypertensive classes reducing specific clinical outcomes, i.e. diuretics appearing more effective for heart failure and CCBs more effective for stroke prevention with beta‐blockers and ACE‐inhibitors not ideal, overall all classes of anti-hypertensives had similar effects in reducing major cardiovascular disease [Citation178].

Currently, four groups of anti-hypertensive FDCs are listed in the WHO EML (21st Edition 2019). These include an ACE inhibitor plus a CCB, ACE inhibitor plus a thiazide or thiazide-like diuretic, ARB plus a CCB and an ARB plus a thiazide or thiazide-like diuretic [Citation111]. However, since lisinopril is preferred over other ACE inhibitors, telmisartan over other ARBs, amlodipine versus other once-daily CCBs, and hydrochlorothiazide (HCTZ) over other similar diuretics, the current 21st WHO EML lists lisinopril plus amlodipine, lisinopril plus hydrochlorothiazide, telmisartan plus amlodipine and telmisartan plus hydrochlorothiazide [Citation111]. This is in line with treatment guidance that recommends initiation with at least two anti-hypertensive medicines for those patients with markedly elevated blood pressure, and follows prior concerns about the limited availability of FDCs in the WHO EML [Citation179Citation181].

There have also been combinations of statins and anti-hypertensives to help reduce CV events including FDCs of amlodipine and atorvastatin. However, there have been mixed findings regarding their effectiveness including increased adherence as well as costs versus single tablets combined [Citation17,Citation139,Citation182Citation185]. Currently, no FDC containing a statin and an anti-hypertensive is listed in the WHO EML [Citation111].

Besides this, there have also been FDCs containing different lipid-lowering medicines including ezetimibe combined with either simvastatin, atorvastatin, or rosuvastatin [Citation186Citation190]. The belief is that by combining different lipid-lowering medicines with different mechanisms of action adherence can be enhanced along with improved effectiveness and outcomes [Citation191]. However, there have been concerns among health authorities regarding the effectiveness of ezetimibe in reducing CV events in reality, which has limited its use in practice [Citation192,Citation193]. Currently, no FDCs containing combinations of different lipid-lowering treatments with different mechanisms of action are listed in the WHO EML, potentially reflecting some of the controversies seen [Citation111].

There are also FDCs containing lipid-lowering medicines and oral antihyperglycemic agents (AHAs) to try and improve outcomes in patients with both dyslipidemia and T2DM through reducing the pill burden [Citation194]. However, currently, no FDCs containing a statin and AHA are listed in the WHO EML [Citation111].

Attention in recent years has turned to the development and availability of a ‘polypill’, which is an oral tablet containing low dose aspirin, a statin, and at least one anti-hypertensive medicine to prevent CV events [Citation195]. Such a pill is potentially seen as an affordable and cost-effective for the prevention of CVD especially in LMICs if the polypill was made available based on current public sector prices [Citation38,Citation40,Citation42,Citation143,Citation196Citation199]. Polypills have also been shown to enhance adherence, are well tolerated and reduce risk factors in both primary and secondary prevention [Citation87,Citation196,Citation200Citation202]. However, there are concerns that a single polypill may not be suitable for all patients and it could well be necessary to develop several different types of polypills to meet the needs of all patients to maximize effectiveness and efficiency [Citation203,Citation204]. In addition, the availability of FDCs has to be balanced against the increased risk of duplication of medicines among hypertensive patients being prescribed FDCs and concerns with prescribing a polypill initially without titration [Citation87,Citation205].

2.2. Type 2 diabetes mellitus (T2DM)

First-line treatment in patients with T2DM is typically metformin [Citation46,Citation206Citation209], with the subsequent initiation of additional oral antihyperglycemic agents (AHAs) if patients fail to achieve target HbA1c levels. FDCs have been developed to reduce the pill burden as well as potentially enhance adherence and outcomes in patients with T2DM [Citation46,Citation210Citation212], with reduced pill burden along with improved effectiveness and reduced side-effects welcomed by patients [Citation213]. Poor control of patients with T2DM is a concern especially among African countries [Citation162,Citation214Citation217].

FDCs include those with a sulfonyl urea (SU) and metformin, metformin and acarbose, DPP-4 (dipeptidyl peptidase-4) inhibitors and metformin, thiazolidinedione and metformin, alogliptin and pioglitazone, sodium-glucose transport protein 2 (SGLT2) inhibitors and metformin, and a SGLT2 inhibitor and dipeptidyl peptidase-4 (DPP-4) inhibitor [Citation210,Citation218Citation220]. FDCs containing metformin and a DPP-4 inhibitor, as well as metformin and SGLT2 inhibitors are seen in particular as providing metformin with complementary mechanisms of action to improve glycemic control whilst reducing the pill burden, and similarly with saxagliptin/dapagliflozin FDCs [Citation218,Citation221Citation224].

In their systematic review principally involving high-income countries, Vijayakumar et al. (2017) found improved adherence with FDCs leading to improved effectiveness [Citation225]. However, it was difficult to determine the actual level of clinical significance with no studies appearing to randomize patients to either the FDC or the separate components [Citation225]. Lokhandwala et al. (2016) suggested that improved adherence and compliance with FDCs may well translate into reduced health-care utilization and costs in the US [Citation226]. However, there are issues of affordability in LMICs, especially with the newer oral anti-diabetic medicines.

There are also concerns with both published and unpublished clinical trial data of five metformin containing FDCs in India which currently account for 80% of all metformin sales [Citation27]. Concerns include the limited number of patients in the clinical trials, which typically were not conducted in India, and whether improved health gain is seen for the FDC versus co-prescribing the individual components together. Evans et al. (2015) were also concerned with the typical length of follow-up in clinical trials with for instance one study involving only 40 patients followed up for just 2 weeks [Citation27]. In their critique of Evans et al. though, Kannan et al. (2015) stated that SU/metformin FDCs are particularly popular among general practitioners and patients in India as they contain lower doses of metformin to reduce or stop gastrointestinal side-effects as well as result in a rapid decrease in blood glucose concentrations. However, the relatively high doses of SUs used in the FDCs increases bodyweight worsening insulin resistance and reversing the beneficial CV effects when metformin is prescribed first line in higher doses [Citation59]. In view of this, Kannan et al. recommended using each medicine separately and titrating doses accordingly [Citation59]. This though can be a challenge in India with current high patient co-payments as the cost of the FDCs can often be cheaper than the combined costs of the separate tablets [Citation59].

The disadvantages of FDCs can include difficulties with determining the cause of poor effectiveness and/or the side effects of treatment, patients’ refusal to accept their disease if FDCs are prescribed as initial treatment instead of for instance metformin, and potentially higher costs [Citation227].

Currently, no FDC for patients with T2DM is currently listed in the WHO EML [Citation111], and there are issues with the affordability of most of these FDCs in LMICs.

2.3. Respiratory diseases

In patients with asthma, treatment strategies using combination inhalers of corticosteroids (ICS) and long-acting β agonists (LABAs) are seen as the most effective and safe approach to prevent exacerbations [Citation228Citation231]. For instance, Tohda et al. (2010) demonstrated that the FDC of fluticasone and salmeterol resulted in a higher proportion of totally controlled weeks per patient with asthma versus fluticasone [Citation232]. This builds on guidelines advocating the use of such combinations [Citation231,Citation233]. This was based on the evidence that LABAs can potentially increase the risk of mortality if used in patients with unstable asthma without the concomitant use of ICS therapy [Citation233,Citation234].

Published studies have also compared the effectiveness of ICS (fluticasone) and LABA (salmeterol) FDCs with other inhaled ICS containing regimens including LABAs in patients with chronic asthma. In the UK, Doull et al. (2007) ascertained that in adults, this FDC was cheaper than increasing the dose of fluticasone, and in children, the FDC was similarly effective compared with fluticasone plus salmeterol in separate inhalers; however, its use resulted in annual cost savings of between GB£47 and GB£77 per child based on UK costs [Citation235]. Studies in Canada have investigated the cost/QALY of the fluticasone/salmeterol FDC [Citation236], and in Japan, Tohda et al. (2010) demonstrated that the FDC of fluticasone and salmeterol resulted in lower mean direct management costs [Citation232]. Previously, Jonsson et al. (2004) in Sweden had demonstrated that a budesonide (ICS) and formoterol (LABA) FDC had improved effectiveness and lowered costs versus separate inhalers [Citation237]. These though are all studies from high-income countries.

Currently, only one ICS/LABA combination (budesonide and formoterol) is included in the WHO EML [Citation111]. This reflects the fact that the listed medicines within the WHO EML should be the example of the class for which there is the best evidence for effectiveness and safety. In some cases, this may be the first medicine that is licensed for marketing with no overall difference in terms of efficacy and safety data between the various FDCs available. In addition, the listed medicine or FDC is generally available at the lowest price based on international drug price information sources [Citation111].

LABA/ICS FDC inhalers have also been used in patients with chronic obstructive pulmonary disease (COPD) with studies assessing their effectiveness as well as their cost-effectiveness across countries including LMICs [Citation238Citation244]. However, recently there are concerns with an increased risk of serious pneumonia with LABA/ICS FDCs resulting in guidance that they should only principally be given to COPD patients with asthma-like symptoms [Citation23,Citation245,Citation246]. However, this combination is still being prescribed despite concerns [Citation247].

The costs of LABA/ICS FDC inhalers have started to come down among high-income countries with an increasing use of lower-cost FDCs combined with initiatives to reduce the steroid burden and improved monitoring of patients [Citation23,Citation231,Citation248]. This should further enhance their cost-effectiveness as well as their access among LMICs.

We have not included any evaluation of FDC inhalers containing an ICS plus a short-acting β agonist (SABA), with ICS typically recommended as first line treatment for patients with asthma before moving onto an ICS/ LABA FDC for maintenance therapy [Citation231]. As a result, limited use of ICS/ SABA combinations across countries and also discontinuations [Citation23].

LABA/long-acting antimuscarinic agent (LAMA) FDC inhalers also appear beneficial with enhancing bronchodilation in patients with COPD uncontrolled on single agents [Citation50,Citation249Citation254]. This builds on GOLD guidance which recommends that when a single bronchodilator fails to achieve the desired outcome a second bronchodilator from a different class may be added [Citation249]. There have also been studies assessing the costs and cost-effectiveness of LABA/LAMA FDCs typically in high-income countries [Citation249,Citation255Citation257]. However, currently, LAMAs are unaffordable in a number of LMICs including the public health-care system in South Africa. It is hoped that as more formulations are launched, costs will come down to more affordable levels to enhance access.

It is recognized that pharmacists and other professionals can assist in lowering the costs of medicines to treat patients with COPD through education and improved adherence [Citation258,Citation259]. These activities, coupled with lower costs of inhaler FDCs as different combinations are launched competing with others, should further enhance their use and improve patient care in a cost-efficient manner.

2.4. Pain

Combinations of medicines may often be needed for pain management, especially severe pain, with often a ‘multimechanistic’ approach needed [Citation53]. As a result, a combination of analgesics or an FDC containing analgesics with different mechanisms of action may be needed to increase the effectiveness and/or reduce the side effects versus increasing the doses of single agents alone [Citation53]. This is seen with the FDC of acetaminophen/ibuprofen in patients with moderate to severe postoperative dental pain where the combination provided greater and more rapid analgesia than comparable doses of either agent alone [Citation56]. Besides this, FDCs of tramadol and paracetamol may be opioid sparing without sacrificing effectiveness, which is seen as important given some of the recent concerns with tramadol especially its potential for abuse [Citation260Citation263]. In addition, a tramadol/paracetamol FDC can present a potential alternative to nonsteroidal anti–inflammatory drugs (NSAIDs) in the elderly reducing concerns with possible gastrointestinal side-effects [Citation264].

In their study, Cristancho et al. (2013) found that an FDC of acetaminophen and codeine (AC) had lower cost and was more effective in reducing pain within the first hours after administration versus an acetaminophen and hydrocodone (AH) FDC or an acetaminophen plus tramadol (AT) FDC [Citation265]. The costs/numbers needed to treat for each formulation were $1816 Colombian pesos/2.2 for AC, $4772 Colombian peso/2.3 for AH, and $5342/2.6 for AT. Using AC as the comparator, the ICER for AT from a payer’s perspective was $8790 Colombian pesos and $29,640 Colombian pesos for AH [Citation265].

FDCs of paracetamol and NSAIDs may also offer superior analgesia effects compared with either medicine alone [Citation55]. This is important especially in the elderly resulting in calls to develop an FDC of NSAIDs combined with a gastroprotective medicine such as a proton pump inhibitor or a high-dose histamine-2 receptor antagonist [Citation266].

There are concerns though regarding FDCs for management of pain. Many of these medicines are misused or dangerous in overdose and currently no FDC for pain management is included in the WHO EML [Citation111]. In addition, there are concerns that the general principles of pain management may be compromised by high use of FDCs including adhering to the general principles for the management of chronic pain [Citation267].

2.5. Malaria including prevention

Both dihydroartemisinin-piperaquine (DP) and sulphadoxine-pyrimethamine (SP) have been used for malaria prevention in pregnant mothers, with a recent review suggesting that intermittent preventive treatment may reduce maternal and placental malaria and that monthly DP appears more effective than SP in reducing placental malaria [Citation268].

There are also ongoing initiatives with DP combinations to improve their packaging to facilitate correct use to further improve their overall effectiveness and value, with published studies showing a single course treatment for uncomplicated falciparum malaria is well tolerated [Citation269,Citation270]. Assi et al. (2017) have also demonstrated that the artesunate–amodiaquine FDC is well tolerated to treat uncomplicated PCP malaria under real-life conditions [Citation271], with this combination now widely used. Banek et al. (2018) have shown that co-formulated artemether–lumefantrine (AL) and fixed dose amodiaquine–artesunate (AQAS) have high self-reported adherence rates among children [Citation272]. Overall, AL was less likely to be taken correctly at one of the study sites; however, it was better tolerated overall than AQAS which may enhance its overall utility in routine clinical care [Citation272]. Itoh et al. (2018) found the artemether-lumefantrine FDC effective in treating uncomplicated P. falciparum malaria among Brazilian patients in the Amazon jungle, strongly supporting the continued use of this FDC as a first-line therapy [Citation273]. Recently Ebenebe et al. (2018) compared the effectiveness of 3-day regimens of AL, artesunate-amodiaquine (AA), and DP among 910 under-five children in Nigeria. The authors found all three evaluated treatments were effective in the management of uncomplicated malaria in young children; however, DP appeared slightly more efficacious than either AL or AA [Citation274].

Studies are also ongoing to increase the dose of DP and extend the dosing schedule to four monthly doses to reduce the incidence of malaria especially during the high transmission season, and this may well continue to enhance the value of this FDC [Citation275].

Artemisinin-based combinations or chloroquine in combination with a short course of primaquine have also been shown to be highly effective in the treatment of vivax malaria in Brazil [Citation276]. An FDC of mefloquine combined with artesunate has also been studied in cases of falciparum malaria in the Brazilian Amazon basin and shown to have acceptable efficacy, safety, and tolerability [Citation277]. However, artesunate–mefloquine FDCs have been used infrequently in Africa due to a perceived poor tolerance to mefloquine although recent studies in children in Africa are now suggesting otherwise [Citation278].

There are concerns though with the number and availability of unapproved FDCs to treat malaria among LMICs with recent estimates suggesting almost half of the sales value and volume of antimalarials are generated by unapproved products [Citation279]. This calls for tighter regulatory process to avoid patient harm as well as appropriate training of pharmacists and their assistants when treatments are dispensed without a prescription [Citation62]. There are also concerns about the pharmacokinetic profile of some FDCs for malaria, which again requires further attention going forward.

Overall though, FDCs are routinely used to prevent and manage patients with malaria due to their effectiveness and tolerability, and this will remain. This is endorsed by the WHO, with six FDCs for malaria and its prevention currently included in the WHO EML [Citation111]. However, there is a paucity of information regarding the costs of treating patients with malaria as well as the cost-effectiveness of FDCs. Ezenduka et al. in 2017 estimated the costs of treating uncomplicated malaria at a public health-care facility in Nigeria [Citation280]. A recent study in Tanzania suggests that AL and DP as the first- and second-line treatment, respectively, for patients with malaria will save approximately US$64,423 per year whilst achieving a 3% reduction in the number of malaria cases compared with AL plus quinine [Citation281]. However, a policy that uses DP as the first-line anti-malarial medicine will consume an additional US$780,180 per year whilst achieving a further 5% reduction in the number of malaria cases versus AL followed by DP [Citation281].

Key areas of pharmacoeconomic research in the future will also center around funding monthly prophylaxis versus focused screening and treatment of identified cases [Citation282]. In addition, assessing the simultaneous use of artemether-lumefantrine, artesunate-amodiaquine, and dihydroartemisinin-piperaquine FDCs against strategies in which these treatments would be cycled or used sequentially [Citation283].

2.6. Tuberculosis (TB)

The treatment regimen for TB can be problematic with patients required to take four medicines during the two-month intensive phase followed by a continuation phase of 4 months with two medicines adversely affecting patient adherence in practice [Citation284Citation286]. These concerns have resulted in the development of FDCs to enhance adherence rates and reduce default rates. In a study from Brazil by Braga et al. (2015), FDCs containing rifampin (R), isoniazid (H), and pyrazinamide (Z) combined with ethambutol (E) reduced default rates and halted the potential increase in resistance rates compared with an FDC of RH plus pyrazinamide separately [Citation287]. However, a previous meta-analysis published in 2013 failed to show any difference in the acquisition of drug resistance, bacterial conversion rates after 2 months of treatment, or the incidence of adverse drug reactions between patients on FDCs versus administering the medicines separately [Citation288]. Gallardo et al. (2016) came to similar conclusions in their Cochrane review [Citation70]. Lima et al. (2017) also found that four-medicine FDCs did not improve culture conversion after 2 and 6 months of treatment versus the separate medicines; however, the FDCs provided greater patient comfort by reducing the pill burden as well as reducing gastrointestinal adverse effects [Citation289].

In Brazil, the FDC containing rifampicin, isoniazid, pyrazinamide, and ethambutol (HRZE) has been available since 2009 produced by local laboratories through a Partnership for Productive Development Agreement [Citation290]. In spite of the methodological limitations casting doubt on the findings [Citation291], an interrupted time series evaluating patient outcomes with this FDC found no difference in treatment abandonment rates although there was a trend toward decreased cure rates [Citation292].

Overall, FDCs can simplify treatment regimens, which may be important in some patients, along with instigating therapeutic drug monitoring (TDM) in selected patients to help detect non-adherence early as well as help manage potential drug:drug interactions [Citation293]. Having said this, TDM is likely to be unavailable or unaffordable in most LMICs. Strategies to further enhance adherence rates include opening more treatment centers as well as community outreach centers in rural areas where access is a concern [Citation294]. This builds on strategies to provide standardized anti-TB drug regimens free of charge to all patients administered under direct observation to improve patient outcomes combined with other measures such as patient support [Citation63,Citation295]. We are also seeing in South Africa standard operating procedures instigating enhanced adherence counseling in patients with continued positive smear tests [Citation296,Citation297], and this is continuing.

There can though be concerns with FDCs for patients with TB. These include concerns with poor bioavailability with some FDCs including those with rifampicin as well as concerns when some patients are switched between different rifampicin formulations without adequate monitoring. In addition, concerns with enzyme level elevations, increases in adverse drug reactions with some combinations, concerns with adverse drug reactions with certain FDCs for patients with both HIV and TB, as well as issues associated with TDM and dose adjustments especially with rifampicin [Citation18,Citation298]. Having said this, the WHO currently endorses five FDCs for the treatment of TB in its EML [Citation111].

2.7. Human immunodeficiency virus (HIV)

FDCs are also increasingly used in patients with HIV to reduce the pill burden, with studies showing a lower pill burden with FDCs appreciably enhances adherence rates. This is helped by greater patient preference for FDCs as well as improved outcomes including greater viral suppression and improved health-related quality of life (HRQoL) [Citation44,Citation299Citation309]. However, there is a large variability in the individual components of FDCs used to treat patients with HIV, which can confound the associations reported. As a result, the increase in adherence due to FDCs is not consistently transposed to improving patient outcomes with mixed results reported for viral suppression rates [Citation310] and health-related quality of life (HRQoL) [Citation311]. Key aspects associated with lower HRQoL included being single, smoking, and having co-morbid disease [Citation311].

The WHO endorses the use of FDCs containing tenofovir/lamivudine/dolutegravir (TLD) due to their improved tolerability and effectiveness, a reduced risk of resistance acquisition, lower discontinuation rates, and fewer drug interactions [Citation312,Citation313]. Meireles et al. (2019) in Brazil also found that a TLD combination of TDF/3TC (tenofovir/lamivudine) combined separately with dolutegravir (DTG) was more effective in suppressing viral load than a tenofovir/lamivudine/efavirenz (TLE) FDC, which was not driven by higher adherence rates [Citation314]. As a result, Phillips et al. (2018) modeled that DTG containing combinations is predicted to be both effective and cost-effective among sub-Saharan African countries [Citation315]. Zheng et al. (2018) also found that a generic DTG-based regimen is likely to be cost-effective in India; consequently, they believed this regimen should be recommended as initial therapy in patients newly diagnosed with HIV in India [Citation316]. Having said this, there are still concerns regarding DTG and its combinations among LMICs. These include the need for further studies to better determine the risk of adverse birth outcomes when DTG is initiated pre-conception as well as assessing its effectiveness when co-administered with treatments for patients with TB [Citation317]. However, there are ongoing studies assessing the optimal dosing regimen of DTG with rifampicin as well as its safety in pregnant women helping to address these concerns [Citation313,Citation318Citation320]. The interactions between efavirenz and bedaquiline are a problem for patients with HIV who have M/XDR-TB and who are taking bedaquiline containing FDCs. These patients need to be switched from a generic efavirenz-containing FDC regimen to twice-daily nevirapine with separate companion pills to address concerns [Citation321]. These patients also need to be closely monitored following switching to other antiretroviral (ART) FDCs as there can subsequently be low ART adherence in these patients [Citation321]. Patient monitoring should happen generally when patients are switched between ARTs.

Studies typically undertaken in high-income countries have shown that FDCs prescribed first-line are generally seen as more effective and less costly than other regimes [Citation302,Citation308,Citation322Citation324]. Cohen et al. (2013) in the US found lower pharmacy costs, fewer hospitalizations, and lower hospital costs in patients prescribed FDCs, which resulted in significantly lower overall total health-care costs [Citation32], with a study in Spain suggesting overall costs will increase with multiple tablet regimens versus FDCs due to a greater prevalence of adverse drug events [Citation72]. Colombo et al. (2014) in Italy also found lower costs for FDCs [Citation33]. However, Angeletti et al. (2014) in Italy found only a 1.5% reduction in average annual costs with FDCs versus multiple drug regimens [Citation325]. Contrasting this, Libre et al. (2018) demonstrated greater efficiency for multiple tablets combined in patients with HIV in France and Spain versus FDCs, which was mainly due to similar effectiveness but lower direct costs with multiple tablets [Citation49]. These differences may be sensitive to the availability of generic formulations [Citation49,Citation58].

Among LMICs, in Brazil, a cost analysis per responder was performed alongside a cohort study (J Costa unpublished data) and the authors found the mean annual cost per patient initiated on an FDC was lower than for those prescribed multiple tablet regimens [Citation301]. This was mainly due to lower costs of ART and lower switching rates. There was though no difference in effectiveness rates between groups after 12 months of treatment although overall a better cost-effectiveness ratio for the FDC [Citation326].

Overall, FDCs for patients with HIV are well accepted and endorsed in the WHO EML [Citation111]. Careful consideration is needed when the manufacturers of the different components of FDCs lower their unit costs potentially affecting overall prices and cost-effectiveness of FDCs versus multiple tablet regimens. Sweet et al. (2016) found that overall costs increased in the US with patented FDCs versus generic multiple drug regimens despite lower in-patient costs due to acquisition cost differences [Citation327].

There are also calls to develop a cotrimoxazole and isoniazid FDC together with pyridoxine to help prevent TB from developing in patients with HIV as a cost-effective option [Citation328].

2.8. Infectious diseases treated with antibiotics

We have not included combination antibiotics such as amoxicillin combined with an enzyme inhibitor in this paper as these medicines should be reserved under the recent WHO AWaRe list of antibiotics and there can be considerable concerns with their availability and use [Citation11,Citation12,Citation111Citation114]. Consequently, the prescribing and dispensing of these FDCs should not be encouraged but restricted.

2.9. Consolidated pharmacoeconomic findings in LMICs

contains details of published studies regarding the cost-effectiveness of FDCs among LMICs contained in Sections 2.1 to 2.8. Typically, there are considerably more published studies regarding FDCs for NCDs among higher-income countries versus LMICs.

Table 1. Published pharmacoeconomic studies of FDCs in LMICs.

In view of the comparative lack of pharmacoeconomic studies regarding FDCs in LMICs coupled with ongoing concerns, there is a need to consider both the positive points and concerns regarding FDCs among senior-level personnel in a range of LMICs to provide guidance regarding their future role and value as well as potential ways forward.

2.10. FDC availability across LMICs by disease area

contains details of FDCs available in the public sector, which means full or partial reimbursement from public sources, across a wide range of LMICs. This includes FDCs within the WHO EML as well as other FDCs within the country. Typically, where there are both private and public markets in a country, there is greater availability of FDCs in the private market, e.g. Brazil, South Africa, and Sudan, with typically no reference to the WHO EML. The exception can be for medicines for malaria, TB and HIV where in some countries these are dispensed free of charge in the public sector with the help of the Global Fund, e.g. Sudan.

Table 2. Availability of FDCs among the various LMICs (Public sector only and only oral medicines apart from inhalers).

The differences in the availability of different FDCs within and between the different countries reflect differences in the prevalence of infectious diseases between countries especially for malaria and TB. In addition, the priority for respiratory diseases versus CV disease, the potential wealth of the country especially regarding the number of FDCs for CV diseases, as well as the extent of regulatory control. For instance, we have not included India in in view of the appreciable number of FDCs still available in the country, which are often irrational, although this is starting to change [Citation1,Citation14,Citation27Citation29,Citation36]. Concerns with the prescribing of FDCs in India was emphasized in the study by Balat et al. (2014) where only 5.8%, 9.8%, and 10.9% FDCs prescribed by physicians in Ahmedabad city were included in the WHO EML (2010), National (2011), and Gujarat State (2011) EML, respectively, [Citation330]. Overall in India, there have been 98 different FDCs for CV diseases including hypertension, 26 FDCs for T2DM, 12 inhaler FDCs for patients with respiratory diseases including formoterol and budesonide, and 24 FDCs for the management of pain. There have also been 9 FDCs for malaria including 5 on the WHO EML, 5 FDCs for malaria including 2 on the WHO EML and 18 FDCs for HIV including one on the WHO EML (). As a result, there is an urgent need to sensitize physicians and undergraduates to potential concerns with the irrational prescribing of FDCs where pertinent () [Citation331]. This is beginning to happen in India with, as mentioned, a significant reduction in the number of FDCs available (328 in all) in recent years [Citation29].

2.11. Positive and negative issues with FDCs across disease areas

contains general positive clinical and economic considerations associated with FDCs that enhance their pharmacoeconomic profile, which is based on the perspectives and experiences of the senior level coauthors from multiple LMICs. contains details of additional benefits for the different infectious and noninfectious disease areas where FDCs are typically prescribed.

Table 3. Positive clinical and economic (general) considerations associated with FDCs.

Table 4. Positive clinical considerations with FDCs across disease areas.

and contain concerns that the senior level coauthors have regarding FDCs, which are both general and disease specific.

Table 5. General concerns regarding FDC.

Table 6. Clinical concerns regarding FDCs across disease areas.

contains suggestions for possible activities that can be undertaken within countries to enhance the prescribing of valued FDCs given the concerns that have existed regarding FDCs with appreciable variability in their availability among LMICs (). Potential activities include a greater role for patients and patient organizations to enhance the prescribing and adherence to valued FDCs to enable patients to attain and retain treatment goals especially those with chronic NCDs [Citation332]. In addition, helping to ensure FDCs are produced at low costs, which is especially important where there are issues of access and high co-payments, combined with incentives to enhance their development and availability [Citation333]. Finally, there is a great need for more published studies and health technology assessments demonstrating their value in LMICs given the current paucity of studies to date ().

documents potential activities that can be undertaken to reduce or negate the availability and prescribing of FDCs of limited value and/or where there are concerns with their irrationality.

BOX 1. Potential initiatives that can be undertaken by key stakeholder groups to enhance the availability and prescribing of valued FDCs.

BOX 2. Potential initiatives that can be undertaken by key stakeholder groups to reduce or negate the availability of FDCs where concerns.

2.12. Limitations

We are aware of a number of limitations to this paper. These include the fact that we did not undertake a systematic review for the reasons stated. As a result, there may be some biases in our findings. However, we tried to negate this through using senior-level academic and health authority personnel from across an appreciable number of LMICs to give guidance on potential publications regarding FDCs across multiple disease areas to include in this perspective paper as well as help with contextualization of the findings. This especially given the paucity of health economic studies of FDCs in LMICs versus high-income countries.

We are also aware that we did not include all LMICs. However, we did include LMICs from across continents to help address this. Overall, we believe our findings and suggestions are robust providing direction for the future.

3. Conclusions

FDCs are valued across a range of disease areas as seen by the number of FDCs listed in WHO and country EMLs (). This reflects their value with improving disease management, reducing adverse reactions and improving adherence rates. This is despite only a limited number of pharmacoeconomic analyses to date in high priority disease areas in LMICs versus high-income countries to fully appraise whether FDCs are pharmacoeconomically justified. Having said this, there are a number of concerns with FDCs including increasing the number of adverse reactions, reducing effectiveness in routine clinical practice, encouraging imprecise diagnoses and increasing costs which affect their overall value. Consequently, their availability and use need to be carefully managed in routine clinical care, with the use of FDCs enhanced by the availability of robust clinical and economic data. A number of activities are also needed to enhance their utility alongside more pharmacoeconomic analyses. These include greater education of physicians and patients of their value where pertinent alongside activities to further improve adherence rates especially in patients with chronic diseases. Concurrent with this, ongoing activities including stricter regulations to limit the availability and use of FDCs of limited value.

Overall, we are likely to see greater availability and use of valued FDCs across LMICs in the future to improve patient care as more evidence becomes available. This is especially important in patients with infectious diseases such as HIV and TB as well as NCDs including CV diseases and diabetes.

4. Expert opinion

We expect to see growing availability and use of FDCs in both infectious and non-infectious disease areas in the future building on their potential advantages. Advantages include improved response rates when combined especially where there are side-effect concerns at optimal doses of single agents. In addition, improved adherence rates through more simplified dosing regimens. Improved adherence rates are particularly important where there are complex treatment regimens and where patients are often on multiple medicines to help control their disease. These advantages are recognized by the endorsement of FDCs in priority disease areas by the WHO as well as by national and regional governments in their lists of medicines available within public health-care systems. FDCs can also be cost-effective; however, there is a paucity of such data within LMICs.

There are though recognized disadvantages with FDCs. These include the availability of irrational FDCs especially in countries such as India and Nepal, although this is changing. There are also concerns that the pharmacogenetics of patients will not be taken into consideration in their development, concerns with identifying which component is responsible for side-effects when these occur, challenges with dose adjustments and appreciable higher prices for the FDCs versus the separate components. Higher prices can persist when the components are available as lower-cost generics but the FDC prolongs the patent life. Concerns with dose adjustments can be helped by making multiple dosing forms available. We are likely to see an increasing number of studies conducted in LMICs demonstrating the clinical and economic advantages of FDCs to address such concerns. This will be helped by the growing capability of LMICs to conduct robust health technology assessments especially middle-income countries. The listing of FDCs within reimbursement and procurement lists will also be helped by realistic pricing versus the components separately. Pricing is particularly important where there are high patient co-payments. We are also likely to see improved regulatory standards to remove or negate marketing authorization of FDCs of limited value, or unapproved FDCs, building on previous initiatives in Europe via the EMA and in the US via the FDA. This is already happening in India leading to the removal of over 300 FDCs in recent years.

We are also likely to see educational initiatives to improve the knowledge of FDCs among physicians, pharmacists, and patients. This includes addressing concerns with over treatment if physicians and patients are not fully aware of the components of FDCs as well as helping with the transition from individual medicines to FDCs where necessary especially for patients with NCDs. Such activities are likely to be increasingly combined with initiatives to enhance adherence rates to prescribed medicines including educational activities and adherence clubs to further improve patient care especially in patients with asymptomatic chronic conditions. Consequently, as mentioned, we are likely to see growing utilization of valued FDCs across LMICs in the future especially with LMICs striving to achieve their SDGs. Alongside this, an increasing number of publications undertaken in LMICs demonstrating that FDCs are pharmacoeconomically sound within a number of disease areas although there will continue to be concerns with some of them.

Article Highlights

  • Fixed dose combinations (FDCs) are welcomed across countries illustrated by endorsement from the World Health Organisation

  • However there are concerns including their rationality, potential to increase adverse drug reactions, dosing schedules with peak effectiveness at different times, lack of titration and potentially higher prices

  • There is a paucity of data among low- and middle-income countries assessing their value and cost-effectiveness in routine clinical care affecting availability and funding

  • Perceived benefits regarding FDCs among senior-level personnel working in LMICs include simplifying the treatment schedule – especially important in complex disease areas, improved adherence rates and tolerability, reduced overall costs and reduced chances of stock-outs

  • Additional perceived concerns include the potential for overtreatment if physicians and patients are not fully aware of their constituents, potential to increase polypharmacy and missed doses have a greater impact on subsequent patient care

  • Initiatives to enhance the prescribing and dispensing of FDCs where valued include physician and patient education, developing quality indicators around their use, accelerating their registration and companies having realistic pricing expectations

  • Possible initiatives to reduce or negate the availability of FDCs where there are concerns include a requirement for companies seeking registration to provide robust health technology assessment data to support the application as well as improved physician education and greater interaction with national patient organisations

Declaration of Interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer Disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Gautam CS, Saha L. Fixed dose drug combinations (FDCs): rational or irrational: a view point. Br J Clin Pharmacol. 2008;65(5):795–796.
  • Sreedhar D, Subramanian G, Udupa N. Combination drugs: are they rational? Curr Sci. 2006;91:406.
  • EMA. Guideline on clinical development of fixed combination medicinal products. 2017 [cited 2019 Oct 10]. Available from https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-development-fixed-combination-medicinal-products-revision-2_en.pdf
  • Auwal F, Dahiru MN, Abdu-Aguye SN. Availability and rationality of fixed dose combinations available in Kaduna, Nigeria. Pharm Pract (Granada). 2019;17(2):1470.
  • Sawicki-Wrzask D, Thomsen M, Bjerrum OJ. An analysis of the fixed-dose combinations authorized by the European Union, 2009–2014: a focus on benefit-risk and clinical development conditions. Ther Innov Regul Sci. 2015;49(4):553–559.
  • Duconge J, Ruano G. Fixed-dose combination products and unintended drug interactions: urgent need for pharmacogenetic evaluation. Pharmacogenomics. 2015;16(15):1685–1688.
  • Bangalore S, Kamalakkannan G, Parkar S, et al. Fixed-dose combinations improve medication compliance: a meta-analysis. Am J Med. 2007;120(8):713–719.
  • Blaszczyk B, Miziak B, Czuczwar P, et al. A viewpoint on rational and irrational fixed-drug combinations. Expert Rev Clin Pharmacol. 2018;11(8):761–771.
  • Fortin A, Verbeeck RK, Jansen FH. Comparative oral bioavailability of non-fixed and fixed combinations of artesunate and amodiaquine in healthy Indian male volunteers. Eur J Clin Pharmacol. 2011;67(3):267–275.
  • Dubey R. Bioequivalence challenges in development of fixed-dose combination products: looking beyond reformulation. Expert Opin Drug Deliv. 2012;9(3):325–332.
  • McGettigan P, Roderick P, Kadam A, et al. Threats to global antimicrobial resistance control: centrally approved and unapproved antibiotic formulations sold in India. Br J Clin Pharmacol. 2019;85(1):59–70.
  • ReAct. Why are fixed dose combinations of antibiotics generally not a good idea? 2018 [cited 2019 Oct 15]. Available from: https://www.reactgroup.org/news-and-views/news-and-opinions/year-2018/why-are-fixed-dose-combinations-of-antibiotics-generally-not-a-good-idea/
  • Menditto E, Orlando V, De Rosa G, et al. Patient centric pharmaceutical drug product design-the impact on medication adherence. Pharmaceutics. 2020;12(1):E44.
  • Gupta YK, Ramachandran SS. Fixed dose drug combinations: issues and challenges in India. Indian J Pharmacol. 2016;48(4):347–349.
  • Bjerrum OJ, Gautam Y, Honore´ PH, et al. Drug-drug combinations revisited. Eur J Hosp Pharm. 2014;21:8–12.
  • Kalaba M, Godman B, Vuksanovic A, et al. Possible ways to enhance renin-angiotensin prescribing efficiency: republic of Serbia as a case history? J Comp Eff Res. 2012;1(6):539–549.
  • Clarke PM, Avery AM. Perspectives - Evaluating the costs and benefits of using combination therapies. MJA. 2014;200(9):1–3.
  • Iftikha S, Sarwar MR. Potential disadvantages associated with treatment of active tuberculosis using fixed-dose combination: a review of literature. J Basic Clin Pharm. 2017;8:S131–136.
  • Desai D, Wang J, Wen H, et al. Formulation design, challenges, and development considerations for fixed dose combination (FDC) of oral solid dosage forms. Pharm Dev Technol. 2013;18(6):1265–1276.
  • Poulter NR, Dolan E, Gupta AK, et al. Efficacy and safety of incremental dosing of a new single-pill formulation of perindopril and amlodipine in the management of hypertension. Am J Cardiovasc Drugs. 2019;19(3):313–323.
  • Abhyankar D, Shedage A, Gole M, et al. Pharmacokinetics of fixed-dose combination of tenofovir disoproxil fumarate, lamivudine, and efavirenz: results of a randomized, crossover, bioequivalence study. Int J STD AIDS. 2017;28(5):491–498.
  • Hanning SM, Lopez FL, Wong ICK, et al. Patient centric formulations for paediatrics and geriatrics: similarities and differences. Int J Pharm. 2016;512(2):355–359.
  • McCabe H, Godman B, Kurdi A, et al. Prescribing trends of inhaler treatments for asthma and chronic obstructive pulmonary disease within a resource-constrained environment in the Scottish national health service: findings and implications. Expert Rev Respir Med. 2019;13(7):679–689.
  • Podolsky SH, Greene JA. Combination drugs–hype, harm, and hope. N Engl J Med. 2011;365(6):488–491.
  • Wertheimer AI. The economics of polypharmacology: fixed dose combinations and drug cocktails. Curr Med Chem. 2013;20(13):1635–1638.
  • Poudel A, Mohamed Ibrahim MI, Mishra P, et al. Assessment of the availability and rationality of unregistered fixed dose drug combinations in Nepal: a multicenter cross-sectional study. Glob Health Res Policy. 2017;2:14.
  • Evans V, Roderick P, Pollock AM. Adequacy of clinical trial evidence of metformin fixed-dose combinations for the treatment of type 2 diabetes mellitus in India. BMJ Glob Health. 2018;3(2):e000263.
  • McGettigan P, Roderick P, Mahajan R, et al. Use of fixed dose combination (FDC) drugs in India: central regulatory approval and sales of FDCs containing non-steroidal anti-inflammatory drugs (NSAIDs), metformin, or psychotropic drugs. PLoS Med. 2015;12(5):e1001826; discussion e.
  • Miranda MRH, Dubey A, GS R, et al. Fixed-dose combinations banned in India: is it the right decision? An eye-opening review. Expert Opin Drug Saf. 2019;18(10):977–985.
  • Hussain S, Malik F, Mehmood W, et al. Assessment of bioavailability of rifampicin as a component of anti-tubercular fixed dose combination drugs marketed in Pakistan. J Bioequivalence Bioavailability. 2010;2:067071.
  • Khan B, Godman B, Babar A, et al. Assessment of active pharmaceutical ingredients in the registration procedures in Pakistan: implications for the future. GaBI J. 2016;5(4):156–163.
  • Cohen CJ, Meyers JL, Davis KL. Association between daily antiretroviral pill burden and treatment adherence, hospitalisation risk, and other healthcare utilisation and costs in a US medicaid population with HIV. BMJ Open. 2013;3:8.
  • Colombo GL, Castagna A, Di Matteo S, et al. Cost analysis of initial highly active antiretroviral therapy regimens for managing human immunodeficiency virus-infected patients according to clinical practice in a hospital setting. Ther Clin Risk Manag. 2014;10:9–15.
  • Dubrocq G, Rakhmanina N. The pharmacokinetics, pharmacodynamics, and clinical role of fixed dose combination of tenofovir disoproxil fumarate, lamivudine and reduced dose efavirenz (TLE-400) in treating HIV-1 infection. Expert Opin Drug Metab Toxicol. 2018;14(8):773–779.
  • Schellack N, Malan L. An overview of fixed-dose combinations of antihypertensive drugs in South Africa. South Afr Fam Pract. 2014;56(4):206–211.
  • Gupta R, Malhotra A, Malhotra P. Assessment of rational use of fixed dose combinations in hypertension in a tertiary care teaching hospital in north India. Int J Adv Med. 2018;5:1263–1267.
  • Gupta AK, Arshad S, Poulter NR. Compliance, safety, and effectiveness of fixed-dose combinations of antihypertensive agents: a meta-analysis. Hypertension. 2010;55(2):399–407.
  • Singh K, Crossan C, Laba TL, et al. Cost-effectiveness of a fixed dose combination (polypill) in secondary prevention of cardiovascular diseases in India: within-trial cost-effectiveness analysis of the UMPIRE trial. Int J Cardiol. 2018;262:71–78.
  • Vrijens B, Antoniou S, Burnier M, et al. Current situation of medication adherence in hypertension. Front Pharmacol. 2017;8:100.
  • Lin JK, Moran AE, Bibbins-Domingo K, et al. Cost-effectiveness of a fixed-dose combination pill for secondary prevention of cardiovascular disease in China, India, Mexico, Nigeria, and South Africa: a modelling study. Lancet Glob Health. 2019;7(10):e1346–e58.
  • Gonzalez-Gomez S, Melendez-Gomez MA, Lopez-Jaramillo P. Fixed-dose combination therapy to improve hypertension treatment and control in Latin America. Arch Cardiol Mex. 2018;88(2):129–135.
  • Becerra V, Gracia A, Desai K, et al. Cost-effectiveness and public health benefit of secondary cardiovascular disease prevention from improved adherence using a polypill in the UK. BMJ Open. 2015;5(5):e007111.
  • Deshmukh KBS, Qian J, Garza KB, et al. Health care costs associated with addition, titration, and switching antihypertensive medications after first-line treatment: results from a commercially insured sample. J Manag Care Spec Pharm. 2017;23(6):691–699.
  • Ramjan R, Calmy A, Vitoria M, et al. Systematic review and meta-analysis: patient and programme impact of fixed-dose combination antiretroviral therapy. Trop Med Int Health. 2014;19(5):501–513.
  • Hilleman DE. Adherence and health care costs with single-pill fixed-dose combinations in hypertension management. J Managed Care Pharm. 2014;20(1):93–100.
  • CADTH Common Drug. Reviews. Clinical review report: empagliflozin and metformin fixed-dose combination (Synjardy). Ottawa (ON): Canadian Agency for Drugs and Technologies in Health Copyright (c) 2017 Canadian Agency for Drugs and Technologies in Health. 2017.
  • Connor J, Rafter N, Rodgers A. Do fixed-dose combination pills or unit-of-use packaging improve adherence? A systematic review. Bull World Health Organ. 2004;82(12):935–939.
  • van Galen KA, Nellen JF, Nieuwkerk PT. The effect on treatment adherence of administering drugs as fixed-dose combinations versus as separate pills: systematic review and meta-analysis. AIDS Res Treat. 2014;2014:967073.
  • Llibre JM, de Lazzari E, Molina JM, et al. Cost-effectiveness of initial antiretroviral treatment administered as single vs. multiple tablet regimens with the same or different components. Enferm Infecc Microbiol Clin. 2018;36(1):16–20.
  • Cazzola M, Matera MG. Fixed-dose combination inhalers. Handb Exp Pharmacol. 2017;237:117–129.
  • Desmeules J, Rollason V, Piguet V, et al. Clinical pharmacology and rationale of analgesic combinations. Eur J Anaesthesiol Suppl. 2003;28:7–11.
  • Raffa RB. Pharmacology of oral combination analgesics: rational therapy for pain. J Clin Pharm Ther. 2001;26(4):257–264.
  • Raffa RB, Tallarida RJ, Taylor R Jr., et al. Fixed-dose combinations for emerging treatment of pain. Expert Opin Pharmacother. 2012;13(9):1261–1270.
  • O’Brien J, Pergolizzi J Jr, van de Laar M, et al. Fixed-dose combinations at the front line of multimodal pain management: perspective of the nurse-prescriber. Nurs Res Rev. 2013;3:9–22.
  • Ong CK, Seymour RA, Lirk P, et al. Combining paracetamol (acetaminophen) with nonsteroidal antiinflammatory drugs: a qualitative systematic review of analgesic efficacy for acute postoperative pain. Anesth Analg. 2010;110(4):1170–1179.
  • Daniels SE, Atkinson HC, Stanescu I, et al. Analgesic efficacy of an acetaminophen/ibuprofen fixed-dose combination in moderate to severe postoperative dental pain: a randomized, double-blind, parallel-group, placebo-controlled trial. Clin Ther. 2018;40(10):1765–76.e5.
  • Kawalec P, Holko P, Gawin M, et al. Effectiveness of fixed-dose combination therapy in hypertension: systematic review and meta-analysis. Arch Med Sci. 2018;14(5):1125–1136.
  • Walensky RP, Sax PE, Nakamura YM, et al. Economic savings versus health losses: the cost-effectiveness of generic antiretroviral therapy in the United States. Ann Intern Med. 2013;158(2):84–92.
  • Kannan S, Mahadevan S, Ramakrishnan A. Fixed dose combinations for type 2 diabetes. Lancet Diabetes Endocrinol. 2015;3(6):408.
  • WHO. Fixed‐dose combinations for HIV/AIDS, tuberculosis, and malaria. 2003 [cited 2019 Oct 10]. Available from: https://apps.who.int/medicinedocs/pdf/s6172e/s6172e.pdf.
  • Purdy M, Robinson M, Wei K, et al. The economic case for combating malaria. Am J Trop Med Hyg. 2013;89(5):819–823.
  • Mwita SJM, Marwa K, Hamasaki K, et al. Medicines dispensers’ knowledge on the implementation of an artemisinin-based combination therapy policy for the treatment of uncomplicated malaria in Tanzania. J Pharm Health Serv Res. 2017;8:227–233.
  • Kibuule D, Rennie TW, Ruswa N, et al. Effectiveness of community-based DOTS strategy on tuberculosis treatment success rates in Namibia. Int J Tuberc Lung Dis. 2019;23(4):441–449.
  • Wang H, Wolock TM, Carter A, et al. Estimates of global, regional, and national incidence, prevalence, and mortality of HIV, 1980–2015: the global burden of disease study 2015. Lancet HIV. 2016;3(8):e361–87.
  • Rankgoane-Pono G, Tshikuka JG, Magafu MGMD, et al. Incidence of diabetes mellitus-related comorbidities among patients attending two major HIV clinics in Botswana: a 12-year retrospective cohort study. BMC Res Notes. 2018;11(1):90.
  • Haacker M, Barnighausen T, Atun R. HIV and the growing health burden from noncommunicable diseases in Botswana: modelling study. J Glob Health. 2019;9(1):010428.
  • Njuguna B, Kiplagat J, Bloomfield GS, et al. Prevalence, risk factors, and pathophysiology of dysglycemia among people living with HIV in Sub-Saharan Africa. J Diabetes Res. 2018;2018:6916497.
  • Mutemwa M, Peer N, de Villiers A, et al. Prevalence, detection, treatment, and control of hypertension in human immunodeficiency virus (HIV)-infected patients attending HIV clinics in the Western Cape Province, South Africa. Medicine (Baltimore). 2018;97(35):e12121.
  • Berkowitz N, Okorie A, Goliath R, et al. The prevalence and determinants of active tuberculosis among diabetes patients in Cape Town, South Africa, a high HIV/TB burden setting. Diabetes Res Clin Pract. 2018;138:16–25.
  • Gallardo CR, Rigau Comas D, Valderrama Rodriguez A, et al. Fixed-dose combinations of drugs versus single-drug formulations for treating pulmonary tuberculosis. Cochrane Database Syst Rev. 2016;(5):Cd009913.
  • Caplan MR, Daar ES, Corado KC. Next generation fixed dose combination pharmacotherapies for treating HIV. Expert Opin Pharmacother. 2018;19(6):589–596.
  • Homar F, Lozano V, Martínez-Gómez J, et al. Cost analysis of HIV treatment and drug-related adverse events when fixed-dose combinations of antiretrovirals (FDCs) were stopped, versus continuation with FDCs. Health Econ Rev. 2012;2(1):16.
  • WHO Global tuberculosis report (Full) 2019 [cited 2019 Oct 12]. Available from: https://apps.who.int/iris/bitstream/handle/10665/329368/9789241565714-eng.pdf?ua=1
  • Mahipala P, Dorji G, Tisocki K, et al. A critical review of addressing cardiovascular and other non-communicable diseases through a primary health care approach in the South-East Asia Region. Cardiovasc Diagn Ther. 2019;9(2):150–157.
  • International Diabetes Federation. IDF Atlas Ninth Edition. 2019. Available from: https://diabetesatlas.org/upload/resources/material/20200106_152211_IDFATLAS9e-final-web.pdf
  • Dwyer-Lindgren L, Cork MA, Sligar A, et al. Mapping HIV prevalence in sub-Saharan Africa between 2000 and 2017. Nature. 2019;570(7760):189–193.
  • Murray CJ, Ortblad KF, Guinovart C, et al. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990–2013: a systematic analysis for the global burden of disease study 2013. Lancet. 2014;384(9947):1005–1070.
  • Hamid S, Groot W, Pavlova M. Trends in cardiovascular diseases and associated risks in sub-Saharan Africa: a review of the evidence for Ghana, Nigeria, South Africa, Sudan and Tanzania. Aging Male. 2019;22(3):169–176.
  • Salvi S, Kumar GA, Dhaliwal RS. The burden of chronic respiratory diseases and their heterogeneity across the states of India: the global burden of disease study 1990–2016. Lancet Glob Health. 2018;6(12):e1363–e74.
  • WHO. Global health observatory (GHO) data. Mortality and global health estimates. 2016 [cited 2019 Oct 11]. Available from: http://who.int/gho/mortality_burden_disease/en/
  • Cameron A, Ewen M, Ross-Degnan D, et al. Medicine prices, availability, and affordability in 36 developing and middle-income countries: a secondary analysis. Lancet. 2009;373(9659):240–249.
  • Aregbeshola BS, Khan SM. Out-of-pocket payments, catastrophic health expenditure and poverty among households in Nigeria 2010. Int J Health Policy Manag. 2018;7(9):798–806.
  • Russo G, Bloom G, McCoy D. Universal health coverage, economic slowdown and system resilience: Africa’s policy dilemma. BMJ Glob Health. 2017;2(3):e000400–e.
  • Hogan DR, Stevens GA, Hosseinpoor AR, et al. Monitoring universal health coverage within the sustainable development goals: development and baseline data for an index of essential health services. Lancet Glob Health. 2018;6(2):e152–e68.
  • Ranabhat CL, Kim CB, Park MB, et al. Multiple disparities in adult mortality in relation to social and health care perspective: results from different data sources. Global Health. 2017;13(1):57.
  • Morton S, Pencheon D, Squires N. Sustainable development goals (SDGs), and their implementation: a national global framework for health, development and equity needs a systems approach at every level. Br Med Bull. 2017;124(1):81–90.
  • Brimble M, Tay D, Seabrook R, et al. Cardiovascular polypill - current and evolving landscape for primary and secondary prevention. 2016 [cited 2019 Oct 10]. Available from https://wellcome.ac.uk/sites/default/files/cardiovascular-polypill-feb17.pdf
  • Haque M, McKimm J, Godman B, et al. Initiatives to reduce postoperative surgical site infections of the head and neck cancer surgery with a special emphasis on developing countries. Expert Rev Anticancer Ther. 2019;19(1):81–92.
  • Godman B, Wettermark B, van Woerkom M, et al. Multiple policies to enhance prescribing efficiency for established medicines in Europe with a particular focus on demand-side measures: findings and future implications. Front Pharmacol. 2014;5:106.
  • Godman B, Malmstrom RE, Diogene E, et al. Are new models needed to optimize the utilization of new medicines to sustain healthcare systems? Expert Rev Clin Pharmacol. 2015;8(1):77–94.
  • Godman B, Bucsics A, Vella Bonanno P, et al. Barriers for access to new medicines: searching for the balance between rising costs and limited budgets. Front Public Health. 2018;6:328.
  • Godman B, Malmstrom RE, Diogene E, et al. Dabigatran - a continuing exemplar case history demonstrating the need for comprehensive models to optimize the utilization of new drugs. Front Pharmacol. 2014;5:109.
  • Ermisch M, Bucsics A, Vella Bonanno P, et al. Payers’ views of the changes arising through the possible adoption of adaptive pathways. Front Pharmacol. 2016;7:305.
  • Campbell SM, Godman B, Diogene E, et al. Quality indicators as a tool in improving the introduction of new medicines. Basic Clin Pharmacol Toxicol. 2015;116(2):146–157.
  • Godman B, Shrank W, Andersen M, et al. Policies to enhance prescribing efficiency in europe: findings and future implications. Front Pharmacol. 2010;1:141.
  • Moorkens E, Vulto AG, Huys I, et al. Policies for biosimilar uptake in Europe: an overview. PloS One. 2017;12(12):e0190147.
  • Bochenek T, Abilova V, Alkan A, et al. Systemic measures and legislative and organizational frameworks aimed at preventing or mitigating drug shortages in 28 European and Western Asian countries. Front Pharmacol. 2017;8:942.
  • Godman B, Grobler C, Van-De-Lisle M, et al. Pharmacotherapeutic interventions for bipolar disorder type II: addressing multiple symptoms and approaches with a particular emphasis on strategies in lower and middle-income countries. Expert Opin Pharmacother. 2019;20(18):2237–2255.
  • Godman B, Haque M, McKimm J, et al. Ongoing strategies to improve the management of upper respiratory tract infections and reduce inappropriate antibiotic use particularly among lower and middle-income countries: findings and implications for the future. Curr Med Res Opin. 2020;36(2):301–327.
  • Godman B, Basu D, Pillay Y, et al. Review of ongoing activities and challenges to improve the care of patients with Type 2 diabetes across Africa and the implications for the future. In Press Front Pharmacol. 2020. DOI: 10.3389/fphar.2020.00108
  • World Bank. World bank country and lending groups - country classifications. 2018 [cited 2019 Oct 12]. Available from: https://datahelpdesk.worldbank.org/knowledgebase/articles/906519-world-bank-country-and-lending-groups.
  • OECD/EU. Health at a glance: Europe 2018: state of health in the EU cycle. Paris: OECD Publishing; 2018 [cited 2019 Oct 12]. Available from: https://www.oecd-ilibrary.org/docserver/health_glance_eur-2018-en.pdf?expires=1548072191&id=id&accname=guest&checksum=C37CC57793822050370C2BC1A2CEA2CF
  • Phelan M, Cook C. A treatment revolution for those who can afford it? Hepatitis C treatment: new medications, profits and patients. BMC Infect Dis. 2014;14(Suppl 6):S5.
  • Henry B. Drug pricing & challenges to hepatitis C treatment access. J Health Biomed Law. 2018;14:265–283.
  • Iyengar S, Tay-Teo K, Vogler S, et al. Prices, costs, and affordability of new medicines for hepatitis c in 30 countries: an economic analysis. PLoS Med. 2016;13(5):e1002032–e.
  • Kamal-Yanni M. Hepatitis C drug affordability. Lancet Glob Health. 2015;3(2):e73–4.
  • Aggarwal R, Chen Q, Goel A, et al. Cost-effectiveness of hepatitis C treatment using generic direct-acting antivirals available in India. PloS One. 2017;12(5):e0176503.
  • Saez C. WHO - more hepatitis C patients being treated in developing countries; price still an issue. 2016 [cited 2019 Oct 11]. Available from: https://www.ip-watch.org/2016/10/27/hepatitis-c-patients-treated-developing-countries-price-still-issue/
  • Andrieux-Meyer I, Cohn J, de Araujo ES, et al. Disparity in market prices for hepatitis C virus direct-acting drugs. Lancet Glob Health. 2015;3(11):e676–7.
  • Pande S. Steroid containing fixed drug combinations banned by government of India: a big step towards dermatologic drug safety. Indian J Drugs Dermatol. 2016;2:1–2.
  • World Health Organization. Model list of essential medicines. 21st list 2019 [cited 2019 Oct 11]. Available from: https://apps.who.int/iris/bitstream/handle/10665/325771/WHO-MVP-EMP-IAU-2019.06-eng.pdf?ua=1
  • Budd E, Cramp E, Sharland M, et al. Adaptation of the WHO essential medicines list for national antibiotic stewardship policy in England: being AWaRe. J Antimicrob Chemother. 2019;74(11):3384–3389.
  • Hsia Y, Sharland M, Jackson C, et al. Consumption of oral antibiotic formulations for young children according to the WHO access, watch, reserve (AWaRe) antibiotic groups: an analysis of sales data from 70 middle-income and high-income countries. Lancet Infect Dis. 2019;19(1):67–75.
  • Sharland M, Pulcini C, Harbarth S, et al. Classifying antibiotics in the WHO essential medicines list for optimal use-be AWaRe. Lancet Infect Dis. 2018;18(1):18–20.
  • Thera MA, Sehdev PS, Coulibaly D, et al. Impact of trimethoprim-sulfamethoxazole prophylaxis on falciparum malaria infection and disease. J Infect Dis. 2005;192(10):1823–1829.
  • Hobbs CV, Anderson C, Neal J, et al. Trimethoprim-sulfamethoxazole prophylaxis during live malaria sporozoite immunization induces long-lived, homologous, and heterologous protective immunity against sporozoite challenge. J Infect Dis. 2017;215(1):122–130.
  • Manyando C, Njunju EM, D’Alessandro U, et al. Safety and efficacy of co-trimoxazole for treatment and prevention of Plasmodium falciparum malaria: a systematic review. PloS One. 2013;8(2):e56916.
  • Wirtz VJ, Hogerzeil HV, Gray AL, et al. Essential medicines for universal health coverage. Lancet. 2017;389(10067):403–476.
  • World Health Organisation. The selection and use of essential medicines: report of the WHO Expert Committee. 2015 [cited 2019 Oct 10]. Available from: http://apps.who.int/medicinedocs/documents/s22190en/s22190en.pdf
  • Mensah GA, Roth GA, Sampson UK, et al. Mortality from cardiovascular diseases in sub-Saharan Africa, 1990–2013: a systematic analysis of data from the global burden of disease study 2013. Cardiovasc J Afr. 2015;26(2 Suppl 1):S6–10.
  • Roth GA, Johnson C, Abajobir A, et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol. 2017;70(1):1–25.
  • Zuhlke L Why heart disease is on the rise in South Africa. 2016 [cited 2019 Oct 11]. Available from: http://theconversation.com/why-heart-disease-is-on-the-rise-in-south-africa-66167
  • Vally M, Irhuma MOE. Primary prevention of coronary artery disease. S Afr Family Pract. 2018;60(2):32–37.
  • Keetile M, Navaneetham K, Letamo G. Patterns and determinants of hypertension in Botswana. Z Gesundh Wiss. 2015;23(5):311–318.
  • Keates AK, Mocumbi AO, Ntsekhe M, et al. Cardiovascular disease in Africa: epidemiological profile and challenges. Nat Rev Cardiol. 2017;14(5):273–293.
  • Ministry of Health Kenya. Kenya national guidelines for cardiovascular diseases management - division of non-communicable diseases ministry of health. 2018 [cited 2019 Oct 12]. Available from: http://www.health.go.ke/wp-content/uploads/2018/06/Cardiovascular-guidelines-2018_A4_Final.pdf
  • Maduagu ATL, Oguntona CRB, Oguntona EB, et al. Prevalence of coronary heart diseases risk factors in adults population living in nigeria’s largest urban city. J Nutr Disord Ther. 2015;5:153.
  • Ofori-Asenso R, Garcia D. Cardiovascular diseases in Ghana within the context of globalization. Cardiovasc Diagn Ther. 2016;6(1):67–77.
  • Xie L, Frech-Tamas F, Marrett E, et al. A medication adherence and persistence comparison of hypertensive patients treated with single-, double- and triple-pill combination therapy. Curr Med Res Opin. 2014;30(12):2415–2422.
  • Naderi SH, Bestwick JP, Wald DS. Adherence to drugs that prevent cardiovascular disease: meta-analysis on 376,162 patients. Am J Med. 2012;125(9):882–7.e1.
  • Barrios V, Escobar C, Echarri R. Fixed combinations in the management of hypertension: perspectives on lercanidipine-enalapril. Vasc Health Risk Manag. 2008;4(4):847–853.
  • Vlachopoulos C, Grammatikou V, Kallistratos M, et al. Effectiveness of perindopril/amlodipine fixed dose combination in everyday clinical practice: results from the EMERALD study. Curr Med Res Opin. 2016;32(9):1605–1610.
  • Mancia G, Asmar R, Amodeo C, et al. Comparison of single-pill strategies first line in hypertension: perindopril/amlodipine versus valsartan/amlodipine. J Hypertens. 2015;33(2):401–411.
  • Benjamin IJ, Kreutz R, Olsen MH, et al. Fixed-dose combination antihypertensive medications. Lancet. 2019;394(10199):637–638.
  • Putignano D, Orlando V, Monetti VM, et al. Fixed versus free combinations of antihypertensive drugs: analyses of real-world data of persistence with therapy in Italy. Patient Prefer Adherence. 2019;13:1961–1969.
  • Huo Y, Gu Y, Ma G, et al. China STudy of valsartan/amlodipine fixed-dose combination-bAsed long-Term blood pressUre management in HypertenSive patients: a one-year registry (China STATUS III). Curr Med Res Opin. 2019;35(8):1441–1449.
  • Ghiadoni L. Management of high blood pressure in type 2 diabetes: perindopril/indapamide fixed-dose combination and the ADVANCE trial [corrected]. Expert Opin Pharmacother. 2010;11(10):1647–1657.
  • Du L-P, Cheng Z-W, Zhang Y-X, et al. The impact of fixed-dose combination versus free-equivalent combination therapies on adherence for hypertension: a meta-analysis. J Clin Hypertens (Greenwich, CT). 2018;20(5):902–907.
  • Ihm SH, Shin J, Park CG, et al. Efficacy of a fixed dose combination of irbesartan and atorvastatin (Rovelito((R))) in Korean adults with hypertension and hypercholesterolemia. Drug Des Devel Ther. 2019;13:633–645.
  • Williams B, Mancia G, Spiering W, et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur Heart J. 2018;39(33):3021–3104.
  • Wan X, Ma P, Zhang X. A promising choice in hypertension treatment: fixed-dose combinations. Asian J Pharm. 2014;9(1):1–7.
  • Weir MR, Hsueh WA, Nesbitt SD, et al. A titrate-to-goal study of switching patients uncontrolled on antihypertensive monotherapy to fixed-dose combinations of amlodipine and olmesartan medoxomil ± hydrochlorothiazide. J Clin Hypertens. 2011;13(6):404–412.
  • Vedanthan R, Bernabe-Ortiz A, Herasme OI, et al. Innovative approaches to hypertension control in low- and middle-income countries. Cardiol Clin. 2017;35(1):99–115.
  • Nashilongo MM, Singu B, Kalemeera F, et al. Assessing adherence to antihypertensive therapy in primary health care in namibia: findings and implications. Cardiovasc Drugs Ther. 2017;31(5–6):565–578.
  • Nielsen JO, Shrestha AD, Neupane D, et al. Non-adherence to anti-hypertensive medication in low- and middle-income countries: a systematic review and meta-analysis of 92443 subjects. J Hum Hypertens. 2017;31(1):14–21.
  • Awad A, Osman N, Altayib S. Medication adherence among cardiac patients in Khartoum State, Sudan: a cross-sectional study. Cardiovasc J Afr. 2017;28(6):350–355.
  • Godman B, Acurcio F, Guerra Junior AA, et al. Initiatives among authorities to improve the quality and efficiency of prescribing and the implications. J Pharm Care Health Syst. 2014;1(3):1–15.
  • Rampamba EM, Meyer JC, Godman B, et al. Evaluation of antihypertensive adherence and its determinants at primary healthcare facilities in rural South Africa. J Comp Eff Res. 2018;7(7):661–672.
  • Rampamba EM, Meyer JC, Helberg E, et al. Knowledge of hypertension and its management among hypertensive patients on chronic medicines at primary health care public sector facilities in South Africa; findings and implications. Expert Rev Cardiovasc Ther. 2017;15(8):639–647.
  • Fox MP, Pascoe S, Huber AN, et al. Adherence clubs and decentralized medication delivery to support patient retention and sustained viral suppression in care: results from a cluster randomized evaluation of differentiated ART delivery models in South Africa. PLoS Med. 2019;16:7.
  • Kalungia CA, Mwale M, Sondashi IS, et al. Availability of essential antihypertensive and antidiabetic medicines in public health facilities in Lusaka district, Zambia. Med J Zambia. 2017;44(3):140–148.
  • Mitkova ZE, Tachkov K, Petrova G, et al. Factors influencing generics and fixed dose combinations recommendation by pharmacists for cardiology patients. AJPP. 2015;9(43):1020–1025.
  • Petrova G, Doneva M, Mitkova Z, et al. Generics and fixed-dose combinations in cardiology: satisfaction analysis of pharmacists and cardiologists. Biotechnol Biotechnol Equip. 2016;30(1):204–211.
  • Hennekens C. Fixed-dose combination therapy with statins: strengths, limitations and clinical and regulatory considerations. Am J Cardiovasc Drugs. 2008;8(3):155–160.
  • Sica D. Rationale for fixed-dose combinations in the treatment for hypertension. Drugs. 2002;62(3):443–462.
  • Redon J, Pichler G. Comparative study of the efficacy of olmesartan/amlodipine vs. perindopril/amlodipine in peripheral blood pressure after missed dose in type 2 diabetes. J Hypertens. 2016;34(2):359–367.
  • Taddei S. Fixed-dose combination therapy in hypertension: pros. High Blood Press Cardiovasc Prev. 2012;19(2):55–57.
  • Volpe M, Tocci G, de la Sierra A, et al. Personalised single-pill combination therapy in hypertensive patients: an update of a practical treatment platform. High Blood Press Cardiovasc Prev. 2017;24(4):463–472.
  • Gorostidi M. de la Sierra A. Combination therapies for hypertension - why we need to look beyond RAS blockers. Expert Rev Clin Pharmacol. 2018;11(9):841–853.
  • Mbui JM, Oluka MN, Guantai EM, et al. Prescription patterns and adequacy of blood pressure control among adult hypertensive patients in Kenya; findings and implications. Expert Rev Clin Pharmacol. 2017;10(11):1263–1271.
  • James PA, Oparil S, Carter BL, et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the eighth joint national committee (JNC 8). Jama. 2014;311(5):507–520.
  • Mwita JC, Francis JM, Omech B, et al. Glycaemic, blood pressure and low-density lipoprotein-cholesterol control among patients with diabetes mellitus in a specialised clinic in Botswana: a cross-sectional study. BMJ Open. 2019;9(7):e026807.
  • Simons LA, Chung E, Ortiz M. Long-term persistence with single-pill, fixed-dose combination therapy versus two pills of amlodipine and perindopril for hypertension: Australian experience. Curr Med Res Opin. 2017;33(10):1783–1787.
  • Gadzhanova S, Roughead EE, Bartlett LE. Long-term persistence to mono and combination therapies with angiotensin converting enzymes and angiotensin II receptor blockers in Australia. Eur J Clin Pharmacol. 2016;72(6):765–771.
  • Weir MR. The rationale for combination versus single-entity therapy in hypertension. Am J Hypertens. 1998;11(10):163s–9s.
  • Berry KM, Parker WA, Mchiza ZJ, et al. Quantifying unmet need for hypertension care in South Africa through a care cascade: evidence from the SANHANES, 2011‐2012. BMJ Glob Health. 2017;2(3):e000348.
  • Cappuccio FP, Miller MA. Cardiovascular disease and hypertension in sub-Saharan Africa: burden, risk and interventions. Intern Emerg Med. 2016;11(3):299–305.
  • Sherrill B, Halpern M, Khan S, et al. Single-pill vs free-equivalent combination therapies for hypertension: a meta-analysis of health care costs and adherence. J Clin Hypertens. 2011;13(12):898–909.
  • Stankus V, Hemmelgarn B, Campbell NR, et al. Reducing costs and improving hypertension management. Can J Clin Pharmacol. 2009;16(1):e151–5.
  • Akazawa M, Fukuoka K. Economic impact of switching to fixed-dose combination therapy for Japanese hypertensive patients: a retrospective cost analysis. BMC Health Serv Res. 2013;13:124.
  • Dickson M, Plauschinat CA. Compliance with antihypertensive therapy in the elderly: a comparison of fixed-dose combination amlodipine/benazepril versus component-based free-combination therapy. Am J Cardiovasc Drugs. 2008;8(1):45–50.
  • Tung YC, Lin YS, Wu LS, et al. Clinical outcomes and healthcare costs in hypertensive patients treated with a fixed-dose combination of amlodipine/valsartan. J Clin Hypertens. 2015;17(1):51–58.
  • Costa FV. Improving adherence to treatment and reducing economic costs of hypertension: the role of olmesartan-based treatment. High Blood Press Cardiovasc Prev. 2017;24(3):265–274.
  • Ferrario CM, Panjabi S, Buzinec P, et al. Clinical and economic outcomes associated with amlodipine/renin-angiotensin system blocker combinations. Ther Adv Cardiovasc Dis. 2013;7(1):27–39.
  • Yusuf S, Teo KK, Pogue J, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358(15):1547–1559.
  • Dalal K, Ganguly B, Gor A. Assessment of rationality of fixed dose combinations approved in CDSCO List. J Clin Diagn Res. 2016;10(4):Fc05–8.
  • Lloyd-Sherlock P, Beard J, Minicuci N, et al. Hypertension among older adults in low- and middle-income countries: prevalence, awareness and control. Int J Epidemiol. 2014;43(1):116–128.
  • Ettehad D, Emdin CA, Kiran A, et al. Blood pressure lowering for prevention of cardiovascular disease and death: a systematic review and meta‐analysis. Lancet. 2016;387(10022):957‐967.
  • Kishore SP, Salam A, Rodgers A, et al. Fixed-dose combinations for hypertension. Lancet. 2018;392(10150):819–820.
  • Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American college of cardiology/American heart association task force on clinical practice guidelines. J Am Soci Hypertens. 2018;12(8):579.e1-.e73.
  • Mancia G, Fagard R, Narkiewicz K, et al. 2013 ESH/ESC guidelines for the management of arterial hypertension: the task force for the management of arterial hypertension of the European society of hypertension (ESH) and of the European society of cardiology (ESC). J Hypertens. 2013;31(7):1281–1357.
  • Bashir S, Sherwani MU, Shabbir I, et al. Efficacy of fix dose combination (atorvastatin and amlodipine) in treatment of uncontrolled hypertension and dyslipidemia. J Ayub Medical College Abbottabad. 2011;23(3):97–100.
  • Schaffer AL, Buckley NA, Pearson SA. Who benefits from fixed-dose combinations? Two-year statin adherence trajectories in initiators of combined amlodipine/atorvastatin therapy. Pharmacoepidemiol Drug Saf. 2017;26(12):1465–1473.
  • Sharrock T The cost-effectiveness of fixed-dose combinations for preventive cardiovascular pharmacotherapy. 2018 [cited 2019 Oct 10]. Available from: https://ourarchive.otago.ac.nz/bitstream/handle/10523/8471/SharrockTal2018MPH.pdf?sequence=3&isAllowed=y
  • Bartlett LE, Pratt N, Roughead EE. Does a fixed-dose combination of amlodipine and atorvastatin improve persistence with therapy in the Australian population? Curr Med Res Opin. 2018;34(2):305–311.
  • Ma YB, Chan P, Zhang Y, et al. Evaluating the efficacy and safety of atorvastatin + ezetimibe in a fixed-dose combination for the treatment of hypercholesterolemia. Expert Opin Pharmacother. 2019;20(8):917–928.
  • Zhu Y, Hu H, Yang J, et al. The efficacy and safety of statin in combination with ezetimibe compared with double-dose statin in patients with high cardiovascular risk: a meta-analysis. Bosn J Basic Med Sci. 2019.
  • Schlackow I, Kent S, Herrington W, et al. Cost-effectiveness of lipid lowering with statins and ezetimibe in chronic kidney disease. Kidney Int. 2019;96(1):170–179.
  • Mazza A, Torin G, D’Amicis C, et al. Cost-effectiveness of rosuvastatin/ezetimibe therapy in high-risk hypertensive patients with uncontrolled hypercholesterolemia by a previous simvastatin/ezetimibe treatment. J Hypertens. 2019;37(e–Supplement 1):e228.
  • Bartlett LE, Pratt N, Roughead EE. Does tablet formulation alone improve adherence and persistence: a comparison of ezetimibe fixed dose combination versus ezetimibe separate pill combination? Br J Clin Pharmacol. 2017;83(1):202–210.
  • Pappa E, Rizos CV, Filippatos TD, et al. Emerging fixed-dose combination treatments for hyperlipidemia. J Cardiovasc Pharmacol Ther. 2019;24(4):315–322.
  • Godman B, Schwabe U, Selke G, et al. Update of recent reforms in Germany to enhance the quality and efficiency of prescribing of proton pump inhibitors and lipid-lowering drugs. PharmacoEconomics. 2009;27(5):435–438.
  • Leporowski A, Godman B, Kurdi A, et al. Ongoing activities to optimize the quality and efficiency of lipid-lowering agents in the Scottish national health service: influence and implications. Expert Rev Pharmacoecon Outcomes Res. 2018;18(6):655–666.
  • Bae JC, Min KW, Kim YH, et al. Efficacy and safety of fixed-dose combination therapy with gemigliptin (50 mg) and rosuvastatin compared with monotherapy in patients with type 2 diabetes and dyslipidaemia (BALANCE): a multicentre, randomized, double-blind, controlled, phase 3 trial. Diabetes Obes Metab. 2019;21(1):103–111.
  • Mogielnicki M, Swieczkowski D, Bachorski W, et al. The food and drug administration (FDA) and the European medicines agency (EMA) perspective on cardiovascular polypill: a multidimensional concept. Cardiol J. 2016;23(5):515–517.
  • Lopez-Jaramillo P, Gonzalez-Gomez S, Zarate-Bernal D, et al. Polypill: an affordable strategy for cardiovascular disease prevention in low-medium-income countries. Ther Adv Cardiovasc Dis. 2018;12(6):169–174.
  • Barrios V, Kaskens L, Castellano JM, et al. Usefulness of a cardiovascular polypill in the treatment of secondary prevention patients in spain: a cost-effectiveness study. Rev Esp Cardiologia. 2017;70(1):42–49.
  • Gaziano TA, Pandya A, Sy S, et al. Modeling the cost effectiveness and budgetary impact of Polypills for secondary prevention of cardiovascular disease in the United States. Am Heart J. 2019;214:77–87.
  • Nansseu JR, Tankeu AT, Kamtchum-Tatuene J, et al. Fixed-dose combination therapy to reduce the growing burden of cardiovascular disease in low- and middle-income countries: feasibility and challenges. J Clin Hypertens. 2018;20(1):168–173.
  • Huffman MD, Xavier D, Perel P. Uses of polypills for cardiovascular disease and evidence to date. Lancet. 2017;389(10073):1055–1065.
  • Webster R, Bullen C, Patel A, et al. Impact of switching to polypill based therapy by baseline potency of medication: post-hoc analysis of the SPACE Collaboration dataset. Int J Cardiol. 2017;249:443–447.
  • Roshandel G, Khoshnia M, Poustchi H, et al. Effectiveness of polypill for primary and secondary prevention of cardiovascular diseases (PolyIran): a pragmatic, cluster-randomised trial. Lancet. 2019;394(10199):672–683.
  • Franczyk B, Gluba-Brzozka A, Jurkiewicz L, et al. Embracing the polypill as a cardiovascular therapeutic: is this the best strategy? Expert Opin Pharmacother. 2018;19(17):1857–1865.
  • Kolte D, Aronow WS, Banach M. Polypills for the prevention of cardiovascular diseases. Expert Opin Investig Drugs. 2016;25(11):1255–1264.
  • Moriarty F, Bennett K, Fahey T. Fixed-dose combination antihypertensives and risk of medication errors. Heart. 2019;105(3):204–209.
  • Chatterjee S, Khunti K, Davies MJ. Type 2 diabetes. Lancet. 2017;389(10085):2239–2251.
  • American Diabetes Association. (7) Approaches to glycemic treatment. Diabetes Care. 2015;38(Suppl):S41–8.
  • Driver C, Bamitale KDS, Kazi A, et al. Cardioprotective effects of metformin. J Cardiovasc Pharmacol. 2018;72(2):121–127.
  • Montvida O, Shaw J, Atherton JJ, et al. Long-term trends in antidiabetes drug usage in the U.S.: real-world evidence in patients newly diagnosed with type 2 diabetes. Diabetes Care. 2018;41(1):69–78.
  • Jain RK. Empagliflozin/linagliptin single-pill combination therapy for patients with type 2 diabetes mellitus. Expert Opin Pharmacother. 2017;18(6):545–549.
  • Blonde L, San Juan ZT. Fixed-dose combinations for treatment of type 2 diabetes mellitus. Adv Ther. 2012;29(1):1–13.
  • Bajaj HS, Ye C, Jain E, et al. Glycemic Improvement with a fixed-dose combination of DPP-4 inhibitor + metformin in patients with type 2 diabetes (GIFT study). Diabetes Obes Metab. 2018;20(1):195–199.
  • Bluher M, Kurz I, Dannenmaier S, et al. Pill burden in patients with type 2 diabetes in germany: subanalysis from the prospective, noninterventional PROVIL study. Clin Diabetes. 2015;33(2):55–61.
  • Musenge EM, Manankov A, Mudenda B, et al. Glycaemic control in diabetic patients in Zambia. Pan Afr Med J. 2014;19:354.
  • Rwegerera GM, Masaka A, Pina-Rivera Y, et al. Determinants of glycemic control among diabetes mellitus patients in a tertiary clinic in Gaborone, Botswana: findings and implications. Hosp Pract. 2019;47(1):34–41.
  • Stephani V, Opoku D, Beran D. Self-management of diabetes in Sub-Saharan Africa: a systematic review. BMC Public Health. 2018;18(1):1148.
  • Badi S, Abdalla A, Altayeb L, et al. Adherence to antidiabetic medications among sudanese individuals with type 2 diabetes mellitus: a cross-sectional survey. J Patient Exp. 2019;1–6.
  • Blonde L, Dipp S, Cadena D. Combination glucose-lowering therapy plans in T2DM: case-based considerations. Adv Ther. 2018;35(7):939–965.
  • Wang J-S, Huang C-N, Hung Y-J, et al. Acarbose plus metformin fixed-dose combination outperforms acarbose monotherapy for type 2 diabetes. Diabetes Res Clin Pract. 2013;102(1):16–24.
  • González-Ortiz M, Guerrero-Romero JF, Violante-Ortiz R, et al. Efficacy of glimepiride/metformin combination versus glibenclamide/metformin in patients with uncontrolled type 2 diabetes mellitus. J Diabetes Complications. 2009;23(6):376–379.
  • Garnock-Jones KP. Saxagliptin/Dapagliflozin: a review in type 2 diabetes mellitus. Drugs. 2017;77(3):319–330.
  • Davidson JA, Sloan L. Fixed-dose combination of canagliflozin and metformin for the treatment of type 2 diabetes: an overview. Adv Ther. 2017;34(1):41–59.
  • Hu J, Zou P, Zhang S, et al. Empagliflozin/metformin fixed-dose combination: a review in patients with type 2 diabetes. Expert Opin Pharmacother. 2016;17(18):2471–2477.
  • Harris SB. The power of two: an update on fixed-dose combinations for type 2 diabetes. Expert Rev Clin Pharmacol. 2016;9(11):1453–1462.
  • Vijayakumar TM, Jayram J, Meghana Cheekireddy V, et al. Safety, efficacy, and bioavailability of fixed-dose combinations in type 2 diabetes mellitus: a systematic updated review. Curr Ther Res Clin Exp. 2017;84:4–9.
  • Lokhandwala T, Smith N, Sternhufvud C, et al. A retrospective study of persistence, adherence, and health economic outcomes of fixed-dose combination vs. loose-dose combination of oral anti-diabetes drugs. J Med Econ. 2016;19(3):203–212.
  • Cersosimo E, Johnson EL, Chovanes C, et al. Initiating therapy in patients newly diagnosed with type 2 diabetes: combination therapy vs a stepwise approach. Diabetes Obes Metab. 2018;20(3):497–507.
  • Loymans RJ, Gemperli A, Cohen J, et al. Comparative effectiveness of long term drug treatment strategies to prevent asthma exacerbations: network meta-analysis. BMJ. 2014;348:g3009.
  • Papi A, Brightling C, Pedersen SE, et al. Asthma. Lancet. 2018;391(10122):783–800.
  • Papi, A, Canonica, GW, Maestrelli, P, & Paggiaro, P. et al. Rescue use of beclomethasone and albuterol in a single inhaler for mild asthma. N Engl J Med. 2007;356(20):2040‐52.
  • Reddel, HK, FitzGerald, JM, Bateman, ED, Bacharier, LB, Becker, A, & Brusselle, G. (2019). 2019: a fundamental change in asthma management: treatment of asthma with short-acting bronchodilators alone is no longer recommended for adults and adolescents. The European Respiratory Journal, 53(6), 1901046.
  • Tohda Y, Nishima S, Arakawa I, et al. [Cost-effectiveness of salmeterol/fluticasone combination therapy vs. fluticasone propionate in Japanese asthmatic patients]. Yakugaku Zasshi. 2010;130(4):593–603.
  • Beasley R, Fingleton J, Weatherall M. Restriction of LABA use to combination ICS/LABA inhaler therapy in asthma. Thorax. 2013;68(2):119–120.
  • Nelson HS, Weiss ST, Bleecker ER, et al. The salmeterol multicenter asthma research trial: a comparison of usual pharmacotherapy for asthma or usual pharmacotherapy plus salmeterol. Chest. 2006;129(1):15–26.
  • Doull I, Price D, Thomas M, et al. Cost-effectiveness of salmeterol xinafoate/fluticasone propionate combination inhaler in chronic asthma. Curr Med Res Opin. 2007;23(5):1147–1159.
  • Ismaila AS, Risebrough N, Li C, et al. COST-effectiveness of salmeterol/fluticasone propionate combination (Advair((R))) in uncontrolled asthma in Canada. Respir Med. 2014;108(9):1292–1302.
  • Jonsson B, Berggren F, Svensson K, et al. An economic evaluation of combination treatment with budesonide and formoterol in patients with mild-to-moderate persistent asthma. Respir Med. 2004;98(11):1146–1154.
  • Dalal AA, St Charles M, Petersen HV, et al. Cost-effectiveness of combination fluticasone propionate-salmeterol 250/50 microg versus salmeterol in severe COPD patients. Int J Chron Obstruct Pulmon Dis. 2010;5:179–187.
  • Tee A, Chow WL, Burke C, et al. Cost-effectiveness of indacaterol/glycopyrronium in comparison with salmeterol/fluticasone combination for patients with moderate-to-severe chronic obstructive pulmonary disease: a LANTERN population analysis from Singapore. Singapore Med J. 2018;59(7):383–389.
  • Bjermer L, van Boven JFM, Costa-Scharplatz M, et al. Indacaterol/glycopyrronium is cost-effective compared to salmeterol/fluticasone in COPD: FLAME-based modelling in a Swedish population. Respir Res. 2017;18(1):206.
  • Reza Maleki-Yazdi M, Molimard M, Keininger DL, et al. Cost effectiveness of the long-acting beta2-adrenergic agonist (LABA)/long-acting muscarinic antagonist dual bronchodilator indacaterol/glycopyrronium versus the LABA/inhaled corticosteroid combination salmeterol/fluticasone in patients with chronic obstructive pulmonary disease: analyses conducted for Canada, France, Italy, and Portugal. Appl Health Econ Health Policy. 2016;14(5):579–594.
  • Altaf M, Zubedi AM, Nazneen F, et al. Cost-effectiveness analysis of three different combinations of inhalers for severe and very severe chronic obstructive pulmonary disease patients at a tertiary care teaching hospital of South India. Perspect Clin Res. 2015;6(3):150–158.
  • Nannini L, Cates CJ, Lasserson TJ, et al. Combined corticosteroid and long-acting beta-agonist in one inhaler versus placebo for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2007(4):Cd003794.
  • Calverley PM, Anderson JA, Celli B, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356(8):775–789.
  • National Institute For Health And Care Excellence. Guideline Chronic obstructive pulmonary disease in over 16s: diagnosis and management. 2018 [cited 2019 Oct 20]. Available from: https://www.nice.org.uk/guidance/ng115/resources/chronic-obstructive-pulmonary-disease-in-over-16s-diagnosis-and-management-pdf-66141600098245.
  • GOLD. Teaching slide set. 2018 [cited 2019 Oct 12]. Available from: https://goldcopd.org/gold-teaching-slide-set/
  • Stancheva B, Pencheva V, Petrova D, et al. Inhaled corticosteroids in stable COPD – international recommendations and reality in Bulgaria. Eur Respir J. 2017;50:PA677.
  • Price D, Small I, Haughney J, et al. Comparative cost-effectiveness of therapy change from fluticasone/salmeterol to beclometasone dipropionate/formoterol (Fostair 100/6®). Primary Care Respir J. 2013;22. Abstract 29 [cited 2019 Oct 12]. Available at URL. : https://www.nature.com/articles/pcrj2013105#Sec29
  • van Boven JF, Kocks JW, Postma MJ. Cost-effectiveness and budget impact of the fixed-dose dual bronchodilator combination tiotropium-olodaterol for patients with COPD in the Netherlands. Int J Chron Obstruct Pulmon Dis. 2016;11:2191–2201.
  • Dale PR, Cernecka H, Schmidt M, et al. The pharmacological rationale for combining muscarinic receptor antagonists and beta-adrenoceptor agonists in the treatment of airway and bladder disease. Curr Opin Pharmacol. 2014;16:31–42.
  • Ichinose M, Minakata Y, Motegi T, et al. Study design of VESUTO((R)): efficacy of tiotropium/olodaterol on lung hyperinflation, exercise capacity, and physical activity in Japanese patients with chronic obstructive pulmonary disease. Adv Ther. 2017;34(7):1622–1635.
  • ZuWallack R, Allen L, Hernandez G, et al. Efficacy and safety of combining olodaterol Respimat((R)) and tiotropium HandiHaler((R)) in patients with COPD: results of two randomized, double-blind, active-controlled studies. Int J Chron Obstruct Pulmon Dis. 2014;9:1133–1144.
  • Miravitlles M, Urrutia G, Mathioudakis AG, et al. Efficacy and safety of tiotropium and olodaterol in COPD: a systematic review and meta-analysis. Respir Res. 2017;18(1):196.
  • Buhl R, Maltais F, Abrahams R, et al. Tiotropium and olodaterol fixed-dose combination versus mono-components in COPD (GOLD 2-4). Eur Respir J. 2015;45(4):969–979.
  • Chan MC, Tan EC, Yang MC. Cost-effectiveness analysis of a fixed-dose combination of indacaterol and glycopyrronium as maintenance treatment for COPD. Int J Chron Obstruct Pulmon Dis. 2018;13:1079–1088.
  • Hoogendoorn M, Corro Ramos I, Baldwin M, et al. Long-term cost-effectiveness of the fixed-dose combination of tiotropium plus olodaterol based on the DYNAGITO trial results. Int J Chron Obstruct Pulmon Dis. 2019;14:447–456.
  • Capel M, Mareque M, Alvarez CJ, et al. Cost-effectiveness of fixed-dose combinations therapies for chronic obstructive pulmonary disease treatment. Clin Drug Investig. 2018;38(7):611–620.
  • Abdulsalim S, Unnikrishnan MK, Manu MK, et al. Structured pharmacist-led intervention programme to improve medication adherence in COPD patients: a randomized controlled study. Res Social Administrative Pharm. 2018;14(10):909–914.
  • Abdulsalim S, Unnikrishnan MK, Manu MK, et al. Impact of a clinical pharmacist intervention on medicine costs in patients with chronic obstructive pulmonary disease in India. Pharmacoecon Open. 2019.
  • O’Brien J, Pergolizzi JV Jr, van de Laar M, et al. Fixed-dose combinations at the front line of multimodal pain management: perspective of the nurse-prescriber. Nurs Res Rev. 2013;3:9–22.
  • WHO. Expert committee on drug dependence, tramadol: pre-review report agenda item 5.3. 2017. Available from: http://www.who.int/medicines/access/controlled-substances/PreReview_Tramadol.pdf?ua=1
  • WHO. WHO expert committee on drug dependence, forty first report. 2019 [cited 2019 Oct 15]. Available from: https://apps.who.int/iris/bitstream/handle/10665/325073/9789241210270-eng.pdf?ua=1
  • Fynn A, Helberg E, Godman B, et al. Drug utilization review of tramadol hydrochloride in a regional hospital in South Africa; findings and implications. Hosp Pract (1995). 2020. DOI:10.1080/21548331.2020.1724454
  • Merchante IM, Pergolizzi JV Jr., van de Laar M, et al. Tramadol/paracetamol fixed-dose combination for chronic pain management in family practice: a clinical review. ISRN Family Med. 2013;2013(638469):1–15.
  • Cristancho RA, Vecino AI, Misas JD. Cost/effectiveness evaluation of three fixed combinations of acetaminophen and opioids in the management of acute pain in Colombia. Rev Colomb Anestesiol. 2015;43:87–94 [cited 2019 Oct 10]. Available at URL http://www.scielo.org.co/pdf/rca/v43n1/v43n1a11.pdf
  • Moore RA, Derry S, Simon LS, et al. Nonsteroidal anti-inflammatory drugs, gastroprotection, and benefit-risk. Pain Pract. 2014;14(4):378–395.
  • Department of Health Republic of South Africa. South African adult hospital level essential medicines list chapter 26: pain - nemlc recommendations from the meeting of 26 September 2019 [cited 2019 Oct 20]. Available from: https://docs.mymembership.co.za/docmanager/3c53e82b-24f2-49e1-b997-5a35803be10a/00143867.pdf
  • Olaleye A, Okusanya BO, Oduwole O, et al. A systematic review and meta-analysis of dihydroartemisinin-piperaquine versus sulphadoxine-pyrimethamine for malaria prevention in pregnancy. Int J Obstetrics Gynaecology. 2019;146(1):43–55.
  • Medicines for Malaria Venture - Developing antimalarials to save lives. Eurartesim® (dihydroartemisinin-piperaquine). 2019 [cited 2019 Oct 15]. Available from: https://www.mmv.org/access/products-projects/eurartesim-dihydroartemisinin-piperaquine
  • Baiden R, Oduro A, Halidou T, et al. Prospective observational study to evaluate the clinical safety of the fixed-dose artemisinin-based combination Eurartesim® (dihydroartemisinin/piperaquine), in public health facilities in Burkina Faso, Mozambique, Ghana, and Tanzania. Malar J. 2015;14:160.
  • Assi SB, Aba YT, Yavo JC, et al. Safety of a fixed-dose combination of artesunate and amodiaquine for the treatment of uncomplicated plasmodium falciparum malaria in real-life conditions of use in Cote d’Ivoire. Malar J. 2017;16(1):8.
  • Banek K, Webb EL, Smith SJ, et al. Adherence to treatment with artemether-lumefantrine or amodiaquine-artesunate for uncomplicated malaria in children in Sierra Leone: a randomized trial. Malar J. 2018;17(1):222.
  • Itoh M, Negreiros Do Valle S, Farias S, et al. Efficacy of artemether-lumefantrine for uncomplicated plasmodium falciparum malaria in Cruzeiro do Sul, Brazil, 2016. Am J Trop Med Hyg. 2018;98(1):88–94.
  • Ebenebe JC, Ntadom G, Ambe J, et al. Efficacy of artemisinin-based combination treatments of uncomplicated falciparum malaria in under-five-year-old nigerian children ten years following adoption as first-line antimalarials. Am J Trop Med Hyg. 2018;99(3):649–664.
  • Chotsiri P, Zongo I, Milligan P, et al. Optimal dosing of dihydroartemisinin-piperaquine for seasonal malaria chemoprevention in young children. Nat Commun. 2019;10(1):480.
  • Daher A, Pereira D, Lacerda MVG, et al. Efficacy and safety of artemisinin-based combination therapy and chloroquine with concomitant primaquine to treat plasmodium vivax malaria in Brazil: an open label randomized clinical trial. Malar J. 2018;17(1):45.
  • Ferreira MVD, Vieira JLF, Almeida ED, et al. Pharmacokinetics of mefloquine administered with artesunate in patients with uncomplicated falciparum malaria from the Brazilian Amazon basin. Malar J. 2018;17(1):268.
  • Sirima SB, Ogutu B, Lusingu JPA, et al. Comparison of artesunate-mefloquine and artemether-lumefantrine fixed-dose combinations for treatment of uncomplicated plasmodium falciparum malaria in children younger than 5 years in sub-Saharan Africa: a randomised, multicentre, phase 4 trial. Lancet Infect Dis. 2016;16(10):1123–1133.
  • Ligade VS, Thakar TM, Dengale SJ. Fixed dose combinations of anti-tubercular, antimalarial and antiretroviral medicines on the Indian market: critical analysis of ubiquity, sales and regulatory status. Trop Med Int Health. 2019;24(2):238–246.
  • Ezenduka CC, Falleiros DR, Godman BB. Evaluating the treatment costs for uncomplicated malaria at a public healthcare facility in Nigeria and the implications. Pharmacoecon Open. 2017;1(3):185–194.
  • Mori AT, Norheim OF, Robberstad B. Budget impact analysis of using dihydroartemisinin-piperaquine to treat uncomplicated malaria in children in Tanzania. PharmacoEconomics. 2016;34(3):303–314.
  • Manning J, Lon C, Spring M, et al. Cluster-randomized trial of monthly malaria prophylaxis versus focused screening and treatment: a study protocol to define malaria elimination strategies in Cambodia. Trials. 2018;19(1):558.
  • Nguyen TD, Olliaro P, Dondorp AM, et al. Optimum population-level use of artemisinin combination therapies: a modelling study. Lancet Glob Health. 2015;3(12):e758–66.
  • MSF. Treating drug-sensitive Tb in India: implementation of daily therapy with fixed dose combinations. Policy brief 2015 [cited 2019 Oct 10]. Available from: https://www.msfaccess.org/sites/default/files/MSF_assets/TB/Docs/TB_Briefing_FDC_Daily_regimen_India_eng_2015.pdf
  • WHO. Guidelines for treatment of drug-susceptible tuberculosis and patient care. 2017 [cited 2019 Oct 11]. Available from: https://apps.who.int/iris/bitstream/handle/10665/255052/9789241550000-eng.pdf.
  • Ali AO, Prins MH. Patient non adherence to tuberculosis treatment in Sudan: socio demographic factors influencing non adherence to tuberculosis therapy in Khartoum State. Pan Afr Med J. 2016;25:80.
  • Braga JU, Trajman A. Effectiveness of RHZE-FDC (fixed-dose combination) compared to RH-FDC + Z for tuberculosis treatment in Brazil: a cohort study. BMC Infect Dis. 2015;15(1):81.
  • Albanna AS, Smith BM, Cowan D, et al. Fixed-dose combination antituberculosis therapy: a systematic review and meta-analysis. Eur Respir J. 2013;42(3):721–732.
  • Lima GC, Silva EV, Magalhães P, et al. Efficacy and safety of a four-drug fixed-dose combination regimen versus separate drugs for treatment of pulmonary tuberculosis: a systematic review and meta-analysis. Braz J Microbiol. 2017;48(2):198–207.
  • Zuim R, Menezes A, Trajman A. A experiência brasileira com a implementação do 4:1 dose fixa combinada para o tratamento da tuberculose. J Epidemiologia E Serviços De Saúde. 2014;23:537–540.
  • Maciel EL, Braga JU, Bertolde AI, et al. Reflections upon the article “Evaluation of the impact that the changes in tuberculosis treatment implemented in Brazil in 2009 have had on disease control in the country”Authors’ replyEvaluation of the impact that the changes in tuberculosis treatment implemented in Brazil in 2009 have had on disease control in the countryHow do you know which health care effectiveness research you can trust a guide to study design for the PerplexedSegmented regression analysis of interrupted time series studies in medication use researchConducting interrupted time series analysis for single-and multiple-group comparisonsInterrupted time series regression for the evaluation of public health interventions a tutorialHow to obtain the confidence interval from a P value. J Bras Pneumol. 2018;44(3):249–252.
  • Rabahi MF, Silva Junior J, Conde MB. Evaluation of the impact that the changes in tuberculosis treatment implemented in Brazil in 2009 have had on disease control in the country. J Bras Pneumol. 2017;43(6):437–444.
  • Zuur MA, Akkerman OW, Forsman LD, et al. Fixed-dose combination and therapeutic drug monitoring in tuberculosis: friend or foe? Eur Respir J. 2016;48:1230–1233.
  • Ali MH, Alrasheedy AA, Kibuule D, et al. Assessment of multidrug-resistant tuberculosis (MDR-TB) treatment outcomes in Sudan; findings and implications. Expert Rev Anti Infect Ther. 2019;17(11):927–937.
  • Kibuule D, Verbeeck RK, Nunurai R, et al. Predictors of tuberculosis treatment success under the DOTS program in Namibia. Expert Rev Respir Med. 2018;12(11):979–987.
  • Department of Health South Africa. Adherence guidelines for HIV, TB and NCDs - standard operating procedures. 2016. Available from: https://www.nacosa.org.za/wp-content/uploads/2018/05/SOP-Adherence-counselling-A5-booklet-19-03-2017.pdf
  • National Department of Health South Africa. Standard operating procedures for minimum package of interventions to support linkage to care, adherence and retention in care, adherence guidelines for HIV, TB and NCDs. Pretoria, South Africa, 2016 [cited 2019 Oct 12]. Available from: http://www.differentiatedcare.org/Portals/0/adam/Content/_YiT3_-qmECUkmkpQvZAIA/File/SOP%20A5%20booklet%2020-05-2016.pdf
  • Court R, Chirehwa MT, Wiesner L, et al. Quality assurance of rifampicin-containing fixed-drug combinations in South Africa: dosing implications. Int J Tuberc Lung Dis. 2018;22(5):537–543.
  • Sax PE, Meyers JL, Mugavero M, et al. Adherence to antiretroviral treatment and correlation with risk of hospitalization among commercially insured HIV patients in the United States. PloS One. 2012;7(2):e31591.
  • Davies NECG. Fixed-dose combination for adults accessing antiretroviral therapy. South Afr J HIV Med. 2013;14(1). Available from https://sajhivmed.org.za/index.php/hivmed/article/view/104/168
  • Costa J, Ceccato M, Silveira MR, et al. Effectiveness of antiretroviral therapy in the single-tablet regimen era. Revista de saude publica. 2018;52:87.
  • Clay PG, Yuet WC, Moecklinghoff CH, et al. A meta-analysis comparing 48-week treatment outcomes of single and multi-tablet antiretroviral regimens for the treatment of people living with HIV. AIDS Res Ther. 2018;15(1):17.
  • Cotte L, Ferry T, Pugliese P, et al. Effectiveness and tolerance of single tablet versus once daily multiple tablet regimens as first-line antiretroviral therapy - Results from a large french multicenter cohort study. PloS One. 2017;12(2):e0170661–e.
  • Sutton SS, Magagnoli J, Hardin JW. Odds of viral suppression by single-tablet regimens, multiple-tablet regimens, and adherence level in HIV/AIDS patients receiving antiretroviral therapy. Pharmacotherapy. 2017;37(2):204–213.
  • Chen Y, Chen K, Kalichman SC. Barriers to HIV medication adherence as a function of regimen simplification. Ann Behav Med. 2017;51(1):67–78.
  • Tarrier L, Kegg S. Who gets single tablet regimens (STR), and why? J Int AIDS Soc. 2014;17(4 Suppl 3):19777.
  • Aldir I, Horta A, Serrado M. Single-tablet regimens in HIV: does it really make a difference? Curr Med Res Opin. 2014;30(1):89–97.
  • Clay PG, Nag S, Graham CM, et al. Meta-analysis of studies comparing single and multi-tablet fixed dose combination HIV treatment regimens. Medicine (Baltimore). 2015;94(42):e1677.
  • Hirasen K, Evans D, Maskew M, et al. The right combination - treatment outcomes among HIV-positive patients initiating first-line fixed-dose antiretroviral therapy in a public sector HIV clinic in Johannesburg, South Africa. Clin Epidemiol. 2018;10:17–29.
  • Altice F, Evuarherhe O, Shina S, et al. Adherence to HIV treatment regimens: systematic literature review and meta-analysis. Patient Prefer Adherence. 2019;13:475–490.
  • Costa JO, Pearson SA, Acurcio FA, et al. Health-related quality of life among HIV-infected patients initiating treatment in Brazil in the single-tablet regimen era. AIDS Care. 2019;31(5):572–581.
  • Usaid Global Health Supply Chain Program Procurement and Supply Management. The dolutegravir opportunity - managing supply chain risk for the introduction of a new antiretroviral (ARV) medicine. [cited 2019 Oct 12]. Available from: https://www.ghsupplychain.org/sites/default/files/2019-07/20_HIV-AIDS%20TLD%201%20pager.pdf
  • WHO. Dolutegravir (DTG) and the fixed dose combination (FDC) of tenofovir/lamivudine/dolutegravir (TLD). Briefing note - April 30 2018 [cited 2019 Oct 11]. Available from: https://www.who.int/hiv/pub/arv/DTG-TLD-arv_briefing_2018.pdf?ua=1
  • Meireles MV, Pascom ARP, Duarte EC, et al. Comparative effectiveness of first-line antiretroviral therapy: results from a large real-world cohort after the implementation of dolutegravir. AIDS. 2019;33(10):1663–1668.
  • Phillips AN, Cambiano V, Nakagawa F, et al. Cost-effectiveness of public-health policy options in the presence of pretreatment NNRTI drug resistance in sub-Saharan Africa: a modelling study. Lancet HIV. 2018;5(3):e146–e54.
  • Zheng A, Kumarasamy N, Huang M, et al. The cost-effectiveness and budgetary impact of a dolutegravir-based regimen as first-line treatment of HIV infection in India. J Int AIDS Soc. 2018;21(3):e25085.
  • Dorward J, Lessells R, Drain PK, et al. Dolutegravir for first-line antiretroviral therapy in low-income and middle-income countries: uncertainties and opportunities for implementation and research. Lancet HIV. 2018;5(7):e400–e4.
  • Dooley K, Kaplan R, Mwelase N et al. Safety and efficacy of dolutegravir-based art in TB/HIV coinfected adults at week 24. 25th croi 4–7 March 2018 [cited 2019 Oct 15]. Oral abstract 33. Available from: http://www.croiconference.org/sessions/safety-and-efficacy-dolutegravir-based-art-tbhiv-coinfected-adults-week-24.
  • Hill A, Clayden P, Thorne C, et al. Safety and pharmacokinetics of dolutegravir in HIV-positive pregnant women: a systematic review. J Virus Erad. 2018;4(2):66–71.
  • Zash R, Jacobson D, Mayondi G et al. Dolutegravir/tenofovir/emtricitabine (DTG/TDF/FTC) started in pregnancy is as safe as efavirenz/tenofovir/emtricitabine (EFV/TDF/FTC) in nationwide birth outcomes surveillance in Botswana. IAS 2017 - conference on HIV pathogenesis treatment and prevention. . [cited 2019 Oct 15]. Available from: http://www.natap.org/2017/IAS/IAS_142.htm
  • O’Donnell MR, Padayatchi N, Daftary A, et al. Antiretroviral switching and bedaquiline treatment of drug-resistant tuberculosis HIV co-infection. Lancet HIV. 2019;6(3):e201–e4.
  • Pialoux G, Marcelin AG, Cawston H, et al. Cost-effectiveness of dolutegravir/abacavir/lamivudine in HIV-1 treatment-Naive (TN) patients in France. Expert Rev Pharmacoecon Outcomes Res. 2018;18(1):83–91.
  • Girouard MP, Sax PE, Parker RA, et al. The cost-effectiveness and budget impact of 2-drug dolutegravir-lamivudine regimens for the treatment of HIV infection in the United States. Clinl Infect Dis. 2016;62(6):784–791.
  • Beck EJ, Mandalia S, Sangha R, et al. Lower healthcare costs associated with the use of a single-pill ARV regimen in the UK, 2004–2008. PloS One. 2012;7(10):e47376–e.
  • Angeletti C, Pezzotti P, Antinori A, et al. Antiretroviral treatment based cost saving interventions may offset expenses for new patients and earlier treatment start. HIV Med. 2014;15:165–174.
  • de Oliveira Costa J Pharmacoepidemiological and pharmacoeconomic analysis of antiretroviral treatment in single tablet regimen from the perspective of the Brazilian national health system. 2019 [cited 2019 Oct 20]. Available from:: https://repositorio.ufmg.br/handle/1843/30142
  • Sweet DE, Altice FL, Cohen CJ, et al. Cost-effectiveness of single- versus generic multiple-tablet regimens for treatment of HIV-1 infection in the United States. PloS One. 2016;11(1):e0147821–e.
  • Harries AD, Lawn SD, Suthar AB, et al. Benefits of combined preventive therapy with co-trimoxazole and isoniazid in adults living with HIV: time to consider a fixed-dose, single tablet coformulation. Lancet Infect Dis. 2015;15(12):1492–1496.
  • Gaziano TA, Opie LH, Weinstein MC. Cardiovascular disease prevention with a multidrug regimen in the developing world: a cost-effectiveness analysis. Lancet. 2006;368(9536):679–686.
  • Balat JD, Gandhi AM, Patel PP, et al. A study of use of fixed dose combinations in Ahmedabad, India. Indian J Pharmacol. 2014;46(5):503–509.
  • Gautam CS, Aditya S. Irrational drug combination: need to Sensitize undergraduates. Ind J Pharmacol. 2006;38:167–170.
  • Medication with reason. [cited 2019 Oct 20]. Available from: https://www.liekysrozumom.sk/
  • Dionisio D, Gass R, McDermott P, et al. What strategies to boost production of affordable fixed-dose anti-retroviral drug combinations for children in the developing world? Curr HIV Res. 2007;5(2):155–187.