Skip to main content

Open Access 03.04.2024 | Review Article

Optimising oncology drug expenditure in Ireland

verfasst von: Ruth Kieran, Maeve Hennessy, Kate Coakley, Hazel O’Sullivan, Tim Cronin, Daire Lynch, Eibhlin Mulroe, Katie Cooke, Dearbhaile Collins, Seamus O’Reilly

Erschienen in: Irish Journal of Medical Science (1971 -)

Abstract

A combination of improvements in patient survival, increasing treatment duration, and the development of more expensive agents has led to a doubling of per-capita spending on cancer medicines in Ireland (2008–2018). Despite this, access to new drugs is poor in comparison to other EU countries. We examine methods to optimise oncology drug spending to facilitate access to newer anticancer agents. Key targets for spending optimisation (biosimilar use, clinical trials and expanded access programs, waste reduction, avoidance of futile treatment, and altered drug scheduling) were identified through an exploratory analysis. A structured literature search was performed, with a focus on articles relevant to the Irish Healthcare system, supplemented by reports from statutory bodies. At the present time, EMA-approved agents are available once approved by the NCPE. Optimising drug costs occurs through guideline-based practice and biosimilar integration, the latter provides €80 million in cost savings annually. Access to novel therapies can occur via over 50 clinical trials and 28 currently available expanded access programmes. Additional strategies include reversion to weight-based immunotherapy dosing, potentially saving €400,000 per year in our centre alone, vial sharing, and optimisation of treatment schedules. A variety of techniques are being employed by oncologists to optimise costs and increase access to innovation for patients. Use of biosimilars, drug wastage, and prescribing at end of life should be audited as key performance indicators, which may lead to reflective practice on treatment planning. Such measures could further optimise oncology drug expenditure nationally facilitating approval of new agents.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The past two decades have been characterised by a significant expansion in new treatment options. While only one new treatment was approved annually in the 1990s, by 2020, 10 new medicines were being approved per year, with 17 new approvals in 2021 [1]. Alongside improvements in diagnostics, surgery, and supportive care, these developments have greatly improved outcomes for many of our patients, for example in women diagnosed with ER + /Her2 + de novo metastatic breast cancer, approximately 75% are still alive after 5 years, compared to under 30% of those diagnosed in the pre-trastuzumab era [2].
As patients with advanced disease live longer, treatment durations are increasing, and in addition to the high costs of some of the more newly authorised drugs (Table 1), this has led to a commensurate increase in government spending, with per-capita spending on cancer medicines in Ireland doubling from €29 to €64/capita between 2008 and 2018 [3]. Annual drug spending exceeds €300 million [3], representing 27% of state spending on cancer, and 0.1% of total GDP. Despite this, Irish patients access to new oncology drugs remains relatively poor [1], at least partly due to financial constraints. Of the new drugs approved by the European Medicines Agency (EMA) between 2017 and 2020, only 51% were reimbursed by the HSE at the beginning of 2022, placing Ireland second last among the EU-15 countries [1].
Table 1
Current expenditure on highest-grossing solid-tumour drugs (by sales)
Drug
Reimbursed indications (Ireland)
Global sales (2021) [4]
Monthly cost per patientb
Pembrolizumaba
• Melanoma
• Advanced non-small cell lung cancer (NSCLC)
• Advanced urothelial carcinoma
• Advanced head and neck squamous cell carcinoma (HNSCC)
• Classical Hodgkin lymphoma
• Advanced cervical cancer
€17 billion
€8000
Nivolumaba
• Advanced melanoma
• Advanced NSCL
• Advanced renal cell carcinoma
• Advanced HNSCC
• Classical Hodgkin lymphoma
€7 billion
€7,500 [5]
Palbociclib
Hormone-sensitive Her2 negative advanced breast cancer
€5.1 billion
€2570 [6]
Pertuzumab
Her2 expressing breast cancer
€4 billion
€4000
Atezolizumab
Urothelial cancer
NSCLC
Small cell lung cancer
Triple-negative breast cancer
€3.4 billion
€6500
aWHO essential medicines list
bExtrapolated from available data; prices being paid may be subject to confidential negotiations and may in reality be lower than shown here
Alongside advocating for further resources, it is essential that oncologists try to maximise value for money using the available resources. In this article, we will examine mechanisms to provide high-quality care while reducing drug spending, including accessing treatment via expanded access programmes and clinical trials, use of biosimilar agents, optimisation of drug scheduling and dosing, reduction of drug wastage, and avoidance of treatment which is unlikely to be beneficial, particularly at the end of life.

Clinical trials

Clinical trials afford patients access to novel therapies and are essential in driving continued progress and advances in cancer care. Furthermore, they have clear economic benefits, as in industry-sponsored studies, the trial drugs are provided free of charge. The cost implications are significant: in a pre-pandemic review in Cork University Hospital, novel therapies were administered to patients to a value of €50,000–€90,000 each month [7]. A review of clinical trials currently open through Cancer Trials Ireland demonstrates 36 solid-tumour studies offering patients access to investigational agents (Table 2), with an additional 14 haematology trials open. Of the 287 patients accrued to these studies in 2022, 137 (48%) participated in trials investigating immunotherapies, including some of the most expensive marketed drugs. A previous analysis estimated that the return from state investment into cancer trials in Ireland is twice that which is invested, representing a significant return on investment to the state [8]. In 2016 the combined €3.63m funding from the Exchequer and other grants resulted in a saving of an estimated €6.5m in the cost of cancer drugs. Additionally, nearly €6m in tax revenues was generated with a contribution of €16.5m to Ireland’s gross domestic product (GDP) as well as support for over 230 largely highly skilled jobs [7]. One clinical trials unit included in this analysis reported that each €1 in grant funding received, yielded €3 in income from industry for trials, supporting the need for continued expansion and investment in cancer research units nationwide. In international studies, average drug cost avoidance per patient has been estimated at €10,000 to €16,000 [9, 10], with cost savings per centre up to €6 million a year [11], significantly exceeding the costs associated with running a clinical trial [12].
Table 2
Studies with investigational agent arm currently accruing adult patients via Cancer Trials Ireland (April, 2023, not including haematology trials)
 
Trial
Investigational arm
Comparator arm
Irish recruitment target
Breast cancer
1.
EPIK-B5
Alpelisib and Fulvestrant
Placebo and fulvestrant
12
2.
KEYNOTE-B49
Pembrolizumab and therapy of physician choice (TPC)
TPC
Not specified (NS)
3.
lidERA Breast Cancer/ GO42784/ TRIO 045
Giredestrant ± LHRH agonist
Endocrine TPC ± LHRH agonist
40
4.
SASCIA
Sacituzumab govitecan
TPC
40
5.
DESTINY-Breast12
Trastuzumab deruxtecan (compares patients with/without brain metastases)
13
6.
DESTINY-Breast05
Trastuzumab deruxtecan
Trastuzumab emtansine (T-DM1)
5
7.
ZEST/ GSK 213831
Niraparib
Placebo
6
8.
ALEXANDRA/IMpassion030
Chemotherapy with concomitant and maintenance atezolizumab
Chemotherapy
28
Lung cancer
9.
Krystal-7
Adagrasib and pembrolizumab
Platinum and pemetrexed and pembrolizumab
4
10.
MK7684A-008
Pembrolizumab/vibostolimab co-formulation and platinum/etoposide
Atezolizumab and platinum/etoposide
6 + 
11.
KRYSTAL-12
Adagrasib
Docetaxel
9
12.
CA224-104 (RELATIVITY)
Relatlimab and nivolumab and chemotherapy
Nivolumab and chemotherapy
6
13.
AcceleRET-Lung
Pralsetinib
TPC
3
14.
AbbVie M14-239
Telisotuzumab vedotin (assessing response rate)
9
Skin cancer
15.
MK-3475–630/KEYNOTE-630
Pembrolizumab
Placebo
4–8
16.
R2810-ONC-1788
Cemiplimab
Placebo
8
Genitourinary cancer
 
17.
MK6482-012
Pembrolizumab and belzutifan and lenvatinib
Pembrolizumab and lenvatinib
7
18.
MK3475-365
Pembrolizumab combination therapies
Multiple arms
4
19.
MK6482-011
Belzutifan and lenvatinib
Cabozantinib
8
20.
MK6482-022
Belzutifan and pembrolizumab
Placebo and pembrolizumab
15
21.
IMvigor011 B042843
Atezolizumab
Placebo
9
22.
DASL HiCaP
Darolutamide and androgen deprivation therapy and radiation (ADT/RT)
Placebo and ADT/RT
45
23.
MK3475-905/ KEYNOTE-905/ EV-303
Perioperative enfortumab vedotin and pembrolizumab
Cystectomy alone
8
Head and neck cancer
24.
CompARE
Cisplatin and radiation and durvalumab
Cisplatin and radiation
7
Gastrointestinal
25.
LEAP-015 (MK-7902–015)
lenvatinib plus pembrolizumab plus chemotherapy
Chemo alone
20
26.
DESTINY DS8201-A-U306
Trastuzumab deruxtecan
Ramucirumab and paclitaxel
8–9
27.
Astellas 8951-CL-5201
Zolbetuximab, chemotherapy
Chemotherapy
3
28.
Lithium
Lithium, chemotherapy (dose escalation study)
24
29.
ZWI-ZW25-301
Zanidatamab/chemotherapy ± tislelizumab
Trastuzumab/chemotherapy
6
Gynaecological cancers
30.
innovaTV 301 / ENGOT cx12
Tisotumab vedotin
TPC chemotherapy
20
31.
ENGOT-en15/ KEYNOTE-C93-00/ GOG-3064
Pembrolizumab
Platinum/paclitaxel
6
32.
ENGOT-ov65 / KEYNOTE-B96
Pembrolizumab plus paclitaxel ± bevacizumab
Placebo and paclitaxel ± bevacizumab
NS
33.
OVIHIPEC-2
Surgery with HIPEC
Surgery alone
NS
Basket trials (multiple/unknown tumour type)
 
34.
CUPISCO
Targeted/immunotherapy guided by NGS
Platinum
NS
35.
LOXO 101
LOXO 101 (assess response rate)
7
36.
LYNK-002
Olaparib (assess response rate)
NS
Fewer than 5% of patients participate in clinical trials globally, and Irish data is consistent with this [13, 14]. Where trials are offered to patients, fewer than 5% of patients decline participation [13, 14], and the major barrier remains a lack of suitable open trials for tumour type/stage within a reasonable travel distance for the patient.

Expanded access programmes

Expanded access programmes (EAPs) allow patients to access new drugs outside of clinical trials [15], and may be utilised if a drug is approved by regulatory bodies but not yet reimbursed by the state [16]. A common model for these programmes is that the programme sponsor (typically the pharmaceutical company that hopes to market a new drug) provides the drug for free to selected patients after an application by their treating consultant; however, the programme may close to new patients at any time, sometimes before reimbursement is agreed. In Ireland, unlike most other European countries, there is no national protocol or standardised approach for the implementation of EAPs [17]. Consequently, there is a gap in the literature in terms of data and research examining the role that these programmes play in current cancer patient care in Ireland. Cronin et al. [18] assessed the role of expanded access medications in 3 oncology units. Over a 9-year period 193 patients had been enrolled in programmes. Importantly, the results also show that the number of EAPs being accessed increased annually, emphasising the increasing gap between drug approval and drug reimbursement in this jurisdiction. The Irish Society of Medical Oncology (ISMO) has established a registry of expanded access programmes to ensure equity of access of patients. A total of 33 drugs are currently provided for a total of 50 different cancer indications, reflecting the important role of EAPs in the landscape of cancer practice nationally (Table 3).
Table 3
Expanded access programmes currently open to patients in Ireland with solid tumour malignancies, April, 2023
 
Programme
Indication
Contact
GI cancer
Durvalumab
Locally advanced/metastatic bilary tract cancer, in combination with chemotherapy
AstraZeneca
Liposomal Irinotecan
Metastatic adenocarcinoma of the pancreas following progression on gemcitabine-based therapy
Servier Laboratories
Encorafenib
BRAF V600E mutant metastatic colorectal cancer, with cetuximab
Pierre Fabre
Regorafenib
Hepatocellular carcinoma, previously treated with sorafenib
Bayer
Atezolizumab
Advanced/unresectable hepatocellular carcinoma, with bevacizumab
Roche
Lung cancer
Amivantamab
Advanced NSCLC with activating EGFR exon 20 insertion mutations
Janssen
Atezolizumab
Adjuvant NSCLC, PDL1 ≥ 50%, post 4 cycles of platinum-based chemotherapy
Roche
Dacomitinib
Locally advanced/metastatic NSCLC with EGFR mutations
Pfizer
Gynaecological cancers
Dostarlimab
Post platinum, recurrent dMMR endometrial cancer
GSK
Lenvatinib
Relapsed/recurrent pMMR endometrial cancer, with pembrolizumab
Eisai
Breast cancer
Trastuzumab-deruxtecan
Unresectable/metastatic HER2-positive breast cancer, post 2 + prior anti-HER2-based regimens
AstraZeneca/ Daiichi-Sankyo
Abemaciclib
ER + , HER2- metastatic breast cancer
Eli Lilly
Alpelisib
PIK3CA-related overgrowth syndrome
Novartis
Genitourinary
Avelumab
Advanced/metastatic urothelial cancer, post-platinum
Merck-Pfizer
Darolutamide
Non-NCCP Protocol Approved Prostate Cancer
Bayer
Enfortumab vedotin
Locally advanced or metastatic urothelial cancer, previous platinum and previous PD1/PD-L1 inhibitor
Astellas
Thyroid/head and neck
Sorafenib
Progressive, locally advanced/metastatic differentiated thyroid carcinoma, refractory to radioactive iodine
Bayer
Selpercatinib
Patients under 18 years with RET-altered thyroid cancers
Eli Lilly
Melanoma
Tebentafusp
Uveal melanoma
Immunocore
Other/multiple indications
Larotrectinib
Locally advanced/unresectable tumours with NTRK gene fusion, or where surgical resection is likely to result in severe morbidity and have no satisfactory treatment options
Bayer
Erdafitinib
Metastatic carcinoma with confirmed FGFR alteration, having exhausted standard of care therapies
Janssen
Entrectinib
    1. NTRK fusion-positive solid tumour
    2. ROS-1 positive NSCLC
Roche
Adagrasib
KRAS G12C mutant tumours
Mirati Therapeutics
Pembrolizumab
    1. Metastatic/ Neoadjuvant/Adjuvant TNBC
    2. Relapsed/recurrent pMMR endometrial cancer, with Lenvatinib
MSD
Neratinib
Neratinib plus Trastuzumab plus Fulvestrant
Pierre Fabre
Details per ISMO list 2023—list is non-exhaustive, eligibility should be clarified with the sponsor on a per-patient basis

Biosimilars

The term biosimilar describes a biologic drug which has been developed such that there are no ‘clinically meaningful differences’ between it and the reference medicine which has already been licenced [19]. Although the EU considers EMA licensed biosimilars interchangeable [20], unlike generic drugs, the Health (Pricing and Supply of Medical Goods) Act 2013 in Ireland specifically excludes biosimilars from being added to the ‘list of interchangeable medicinal products’, so that these medicines cannot undergo generic substitution. Because of this, clinicians must intentionally decide to switch to these, if appropriate.
To provide an idea of the scope of the cost savings provided by biosimilars, a 2019 study modelled the impact of choosing biosimilar trastuzumab (a monoclonal antibody targeting HER2) instead of its reference product (Herceptin) in 28 European countries over 5 years [21], determining that €1.13–€2.27 billion could be saved based on epidemiological data [21]. The same model estimated that 55,082–116,196 additional patients could gain access to trastuzumab over the 5-year period [21]. Real-world data has revealed savings of GBP £100 million for trastuzumab and adalimumab biosimilars the UK National Health Service (NHS) in 2018 alone [22].
Across all biosimilars, annual savings in Ireland exceed €80 million [23]. Biosimilars currently account for 52–54% of the market share of oncology biologicals in Ireland [23, 24], compared to over 90% in Austria, the Netherlands, and Norway, suggesting scope for further expansion [24].
A 2021 literature review investigated the impact of non-medical switching policies which replace reference biologic products with biosimilars [25], and found that cost savings were achieved in most cases [25], but the concept of biosimilar substitution does not currently have sufficient support from prescribers [26]. Some barriers to more extensive uptake globally identified include patient and prescriber perception of biosimilars, lack of awareness, and issues regarding interchangeability and substitution of biosimilars [27]. Recently updated American Society of Clinical Oncology (ASCO) guidelines support interchangeable use of biologics, and note that to date, no immunogenicity concerns for any approved oncology biosimilars have been identified by the FDA [28]. As experience with using biosimilars has grown substantially, hesitation from oncologists may reduce over time [22]. It has been recommended that there should be scheduled evaluations of patient experience included in biosimilar implementation plans [29], which may help strengthen the case for increased use.

Drug dosing and scheduling

Historically, drug dosing has been determined by the maximum tolerated dose established in phase 1 trials; however, moving to a minimum efficacious dose may be more appropriate. A number of drugs are commonly used at a lower dose than that in their original trials [30], such as the PARP inhibitor niraparib, where the use of a 200 mg starting dose in patients weighing < 77kg is associated with better tolerance and can save €2700 compared to a 300 mg dose [6], without impact on survival outcomes [31].
While immunotherapies have improved outcomes for many patients, controversy exists around the 200/400 mg (pembrolizumab) or 240/480 mg (nivolumab) ‘flat’ doses that are typically prescribed, irrespective of body surface area. While early studies with pembrolizumab investigated a 2 mg/kg dose [3234], when pharmacokinetic data suggested a 200 mg flat dose provided similar exposure distributions [35], research shifted to using this as a standard. As the median patient weight in this study was 77.2 kg, most patients received more product via flat-dose dispensing. In one Cork University Hospital study [36], flat dosing resulted in increased costs of €22,693 and €4018 per patient (pembrolizumab and nivolumab, respectively) in comparison to weight based, with an annual total cost of almost €400,000 in our hospital alone. Current product licences [37, 38], National Cancer Control Programme (NCCP) protocols [39], and most recent studies use flat-based dosing. Consequently, weight-based dosing is an off-label use; however, a global reversion to use of weight-based pembrolizumab dosing could save $0.8 billion per year [40].
Chemotherapy de-escalation in metastatic colorectal cancer is well established, where patients may receive 8–24 weeks of ‘induction’ chemotherapy with a fluoropyrimidine and oxaliplatin/irinotecan before moving to single-agent maintenance. While oxaliplatin and irinotecan are not expensive drugs by the standards of immunotherapies, no survival advantage has been seen by continuing them in meta-analysis [41]. Similarly, ‘treatment holidays’ can improve quality of life and tolerance of treatment in those with stable/responding advanced disease, particularly in colorectal cancer. While the COIN study (observation and reintroduction of therapy as-needed compared to maintenance therapy) did not meet non-inferiority endpoints [42], a later meta-analysis found no evidence of impaired survival [43], with similarly positive findings in the FOCUS4-N trial [44], while using cetuximab on an observation/reintroduction schedule saves over £35,000 per patient annually [45, 46] (almost €49,000, adjusting for inflation). In addition to savings on drug products, drug de-escalation and treatment holidays reduce demands for slots on oncology daywards, a resource which is severely limited in many hospitals [47], and which cannot easily be increased without major investment. Unlike other countries [48], Ireland does not mandate stopping immunotherapy after any set period [39]. The optimal duration of immunotherapy in metastatic disease is being investigated in trials such as DANTE [49], Safe Stop, and STOP-GAP [50], and if strong evidence is found for treatment interruptions after 2 years, this may be a significant cost reduction.
Some agents have interactions with food, which could potentially be leveraged to reduce drug costs. Abiraterone (a hormonal agent used in advanced prostate cancer) is typically taken as a 1000 mg dose on an empty stomach. A 250 mg dose taken with a high-fat meal may yield similar control of PSA [51] to the conventional 1000 mg dose, though long-term survival outcomes have not been reported. This strategy is being used in some low-income countries [52], and the US National Comprehensive Cancer Network (NCCN) guidelines include 250 mg with food as an option for patients where financial toxicity limits access to care [53]. This is not being practised in Irish centres, and a 250 mg dose is not available in the Irish market. Coadministration of grapefruit with the kinase inhibitor lapatinib could also reduce the dose required [5456], potentially saving over €2000 per patient per month [6]. Similar avenues are being explored with other agents [57, 58], though no research on clinical outcomes has been conducted; thus, these strategies are not established enough to use in our patients.

Avoidance of futile treatments

Preventing the use of futile treatments in palliative patients is another method of reducing the financial impact of cancer care. The receipt of systemic anticancer therapy (SACT) at the end of life is associated with poor quality of death, death in hospital, and higher health care costs [59, 60]. Consequently, 30-day mortality rate after SACT is used as a key performance indicator (KPI), both in Ireland [61] and internationally [62]. The National Cancer Strategy (2017–2026) recommends that < 25% of patients should receive SACT in the last month of life [63]. Early referrals to palliative care are associated with less aggressive end of life care [64], improved overall survival, and quality of life [65].
A retrospective study of 582 patients with advanced malignancies that underwent treatment at two Dublin hospitals between 2015 and 2017 found that 22% and 6% received SACT in the last 4 and 2 weeks of life, respectively [66]. A Cork and Mercy University Hospital study assessing immune checkpoint inhibitors (ICI) demonstrated 20% of metastatic patients prescribed ICI received this in the last 30 days of life [67]. In both studies the institutions were compliant with the Cancer Strategy’s KPI and the majority of patients (92% [67] and 88% [66]) were referred to palliative care. The timing of palliative care referrals is important, with O’Sullivan et al. demonstrating that late palliative care referrals (≤ 3 months before death) were associated with hospitalisations in the last month of life [67]. Adopting early palliative care referrals allows patients and their caregivers time to manage treatment expectations, plan end-of-life care, and ultimately reduce healthcare costs [64, 68, 69].
In addition to overtreatment in the advanced setting, efforts must be made to de-escalate unnecessary treatments in patients undergoing radical intent therapy. In breast cancer, trials have sought to reduce the number of agents given adjuvantly in HER2-positive breast cancer. Biomarkers can be used to guide adjuvant SACT selection: in early ER-positive breast cancer, the use of a somatic 21 gene recurrence score led to a 62.5% reduction in chemotherapy in Irish patients, saving the health system €1,900,000 over 8 years. In other solid tumours, circulating tumour DNA (ctDNA) is being investigated as a de-escalation tool. In the dynamic study, stage II colorectal cancer patients that were ctDNA negative postoperatively had adjuvant chemotherapy omitted, with no impact on the primary endpoint of 2-year recurrence-free survival [70]. This technology does not yet have the same sensitivity for adjuvant treatment selection in other cancers, such as NSCLC, where atezolizumab still provides benefit in ctDNA-negative patients [71].

Reducing drug wastage

Apart from strategies to look at reducing spending, we must try to ensure that the money we do spend is used economically. Drug wastage accounts for 4–18% of all cancer drug spending in the USA [7274], and while some of this is unavoidable, drug waste mitigation strategies have been shown to reduce drug spending by up to 17% [74]. Given the €300 million annual spending, even a small reduction in wastage is likely to be very impactful.

Reducing wastage within the hospital

Many drugs are dispensed as single-patient vials; however, these often contain more product than a typical patient needs, with up to 33% of product wasted in some cases [73]. Guidelines in many countries advise that once opened, vials must be disposed of within 6 hours [75], but even within these constraints, saving the ‘waste’ product from two or more vials to later formulate a dose for an extra patient (‘vial sharing’) can result in significant savings. With the addition of steps such as dose banding (already standard NCCP policy), or grouping patients being treated with the same drugs on the same days, up to 45% of drug waste expenditure can be avoided [76]. Closed-system drug transfer devices used in compounding may maintain drug sterility beyond the 6-hour window [77], allowing significantly more vial sharing [78], but were not yet universal in Irish hospitals at the time of last audit [79].
We have already discussed above how the adoption of flat dosing may not be beneficial—while this practice does reduce ‘wastage’ in theory, in reality, it moves us from merely discarding leftover drug to infusing leftover drug into patients.

Reducing wastage of oral treatments

For patients who receive oral anti-cancer therapy, drug wastage is experienced by 25–41% of patients [8083], and in those who discontinue treatment unexpectedly, 46–85% [83, 84] have ‘leftover’ doses. The cost of these drugs can be significant, with unopened packages worth a median of €2600 per patient in a Dutch study [83].
Traditionally, such medications are discarded; however, several drug repositories are now established, primarily in the USA, such as the Wexner Medical Center Oral Oncology Drug Repository Program [85], and have been endorsed by ASCO guidelines [86]. These allow patients to donate leftover medications, with the intent of these being used for un- or underinsured patients, and one European scheme found a 68% waste reduction, with net savings of $1591 in selected cohorts [87]. In non-oncological studies, most patients indicate willingness to accept re-dispensed medications [88, 89], while in a small oncology study, theoretical willingness to participate in drug reuse was over 80% [90]. To our knowledge, no adverse outcomes have been reported from existing repository programmes.
In patients at higher risks of toxicity, consideration could be given to how medications are dispensed, both by modifying the dosage formulations administered and by split-fill dispensing, where patients may collect only part of an oral cycle at a time. This keeps residual medicines within the pharmacy supply chain and allows drugs to be reallocated to other patients. Wastage via these mechanisms can be reduced significantly [91] in up to 34% of patients [81], saving $1000 [80, 81] per patient. This may be particularly important with more expensive oral agents, such as palbociclib, in which at least 18% of women require a dose reduction [92], with an estimated cost of approximately $5000 per patient [9294]. Anticipating the potential for dose reductions when deciding how the prescription should be formulated may be another cost saver in some cases (see Table 4). While these methods are slightly more inconvenient for the patient, their economic advantages are worth considering, especially in the initial weeks of treatment. Unlike other strategies, these methods can easily be implemented by an individual clinician without significant administrative hurdles.
Table 4
Reducing waste by modified dispensing
Many drugs are marketed in multiple drug strengths, such as sunitinib, typically prescribed at a 50 mg daily dose. This is available in Ireland in 50-mg (€4832/28 capsules), 25-mg (€2421/28 capsules), and 12.5-mg (€1214/28 capsules) formulations [6]
Typical prescribing:
John starts sunitinib for the first time and receives a 28-day supply of a single 50 mg capsule daily. He has significant toxicity, and after 12 days his dose is reduced to 37.5mg. He has 16 days of leftover drug, worth €2761, which is discarded
Split-fill prescribing:
John is asked to collect only a 14-day supply of 50mg capsules at first. When he becomes unwell on day 12, he has 2 capsules left, worth €345
Dose-reduction anticipation:
John receives 4 × 12.5 mg capsules every day
When his dose is reduced to 37.5 mg, he takes 3 × 12.5 mg capsules, and has no wasted drug

Discussion

We have outlined potential strategies for optimising drug spending without impacting patient outcomes (Table 5). While some of these will require an initial administrative or infrastructural investment, such as altering pharmacy compounding procedures or opening local access to new clinical trials, many others can be undertaken by an individual clinician almost immediately, such as investigating options for early access drugs, moving to split fill dispensing, or de-escalating individual treatments. Though the individual impact of these simpler interventions is smaller, the cumulative effect if all staff are mindful of costs is likely to be very significant.
Table 5
Key strategies for optimising drug expenditure
 
Summary
Potential impact
Tasks for further implementation
Government
Clinicians
Avoiding unnecessary treatment
Includes personalising adjuvant therapies and considering treatment holidays/therapy de-escalation, or offering best supportive care as a valid option where appropriate.
May reduce the burden of time-consuming and potentially toxic treatments for patients, the use of resources such as day ward treatment slots, and drug spending.
Personalised adjuvant therapy in breast cancer saved €1,900,000 over an 8 year period
Greater use of treatment holidays from agents such as cetuximab could save up to €49,000 per patient
Continue work within national cancer strategy framework to avoid chemotherapy at the end of life
• Ongoing investigator-originated studies are needed to establish safety of therapy de-escalation
• Consider discussing treatment breaks with patients where clinically appropriate
Reducing drug waste
Strategies such as vial sharing, dose banding and closed system drug transfer devices can reduce wastage of intravenous drug products.
Split fill dispensing and drug re-dispensing could reduce wastage for oral products.
Drug waste mitigation strategies can reduce drug spending by up to 17% internationally. Difficult to quantify in Irish context due to lack of national data, but likely to be significant given the €300 million annual drugs spending.
• Consider including drug wastage as a KPI in next national cancer strategy/data collection at national level regarding this
• National level review of the viability of drug re-dispensing
• Consider trial of split fill dispensing in individual units
• Awareness of which medications contribute to wastage most (high-cost/short shelf life)
Utilising biosimilars
Where biosimilar agents are available, transition to routine use of these can significantly reduce costs, both directly and by reducing the cost of the reference product.  
Across all biosimilars, annual savings exceed €80 million. 
Continue price negotiations where biosimilars are available
Consider switch to use of biosimilar products where more cost effective
Clinical trials
In addition to gaining access to new drugs, significant financial burden moves from the HSE to the study sponsor, who may fund staff costs, radiology or laboratory costs, and drug product.
Increasing the number of open trials, and the numbers of patients enrolled, may result in greater savings.
Cost savings of over €10,000 per patient enrolled, €16.5 million overall contribution to GDP (2016 data)
• Increasing state funding for Cancer Trials Ireland
• Ongoing investment in infrastructure needed to support trials (radiology, dayward) to make sites attractive to sponsors
• Ongoing awareness of open trials, willingness to refer patients to other centres for enrolment where appropriate
• Continue to liaise with potential sponsors and complete study feasibility assessments
Expanded access programmes
Where drugs are EMEA approved but not yet reimbursed in Ireland, some companies may offer an Expanded Access Programme. These programmes are not publicly advertised, and ISMO has begun maintaining a database of open programmes. 
Difficult to quantify due to lack of data on utilisation
• NCCP-coordinated register of availability and utilisation of EAPs
• Awareness of open programmes
• Consider approaching pharmaceutical companies to enquire about options in individual cases.
At the same time, some of the cost optimisations discussed here require local or national policy changes, and significant investment of time and effort from oncology physicians, pharmacists, and other staff. While reducing spending in one area will increase funding available in other areas, and hence indirectly increase access to new drugs or treatments, oncology funding is not ringfenced, and it is possible that ‘savings’ from the oncology sector may be reallocated to an entirely different area of government spending. Allowing cost savings to be reinvested locally would require significant policy change. One precedent for this is the ‘Best Value Biologic’ initiative, used in rheumatology for drugs such as adalimumab, in which the HSE Medicines Management Programme issued information for prescribers on the selected preferred biosimilars [95], and provided a €500 gain share to the hospital for each patient switched to a preferred biosimilar [95], saving €22.7 million in a year [95]. After national rollout of NCIS, a central NCCP-led review of biosimilar usage patterns could be conducted to estimate potential cost savings of further biosimilar use and the impact of a similar incentive strategy in oncology. If successful, this could be expanded to other areas of cost optimisation; however, it would require a HSE and hospital commitment that incentive funding was provided in addition to routine funding increases, not instead of.
The current (2017–2026) Cancer Strategy [63] aimed to increase participation in therapeutic clinical trials to 6% by 2020. Updated figures were not available as of 2021 [61], and the NCCP aims to improve intelligence gathering via the National Cancer Information System after its national implementation. Monitoring of this KPI is essential, and we must continue investing in our hospitals, investigators, and trials units, to make Irish study sites maximally attractive to sponsors, as well as develop additional non-industry sponsored trials. In addition to trials investigating new agents, increased funding for investigator-initiated trials could focus on dose and scheduling optimisation. The ASCO Value in Cancer Care consortium is continuing to explore avenues such as leveraging drug interactions to achieve cost savings [96], and this is an evolving research area that may yield more options in the future.
Although an informal database of the expanded access programmes currently open has been created, this is maintained by ISMO volunteers, and no information on activity or patient outcomes is available, except to the programme sponsor. A national database for all expanded access programmes would be desirable, with mandatory registration by prescribers. This would provide real-world data on activity and toxicity, often in rare tumour subtypes, facilitate regulatory bodies in approval assessments, and would also ensure equity of access for patients. Ideally, this would be linked with the National Cancer Information System (NCIS), a single national computerised system that will record and store information of cancer diagnosis and treatments for all patients treated within the public system. First planned in 2013, this system is slowly being expanded, but does not yet capture data from all sites, limiting our ability to capture data on other KPIs, such as those related to chemotherapy in the last month of life [61]. In the interim, individual cancer units should audit this themselves, alongside auditing rationalisation of other medications.
‘Drug wastage’ is not being captured at a national level in Ireland, and this could be considered for inclusion in the next National Cancer Strategy. While some wastage is inevitable to allow efficient patient flow through the dayward (e.g. many units begin compounding low-cost drugs before all blood results are finalised for patients who appear well, to minimise waiting times), at a hospital level, residual drug contents in vials, and dose reductions/treatment cancellations after drugs have been compounded should be monitored, and investment could be considered in closed-system transfer devices where not already in place. Research on modelling potential savings with split-fill prescribing, and into the acceptability of oncology drug repositories to Irish patients and clinicians is ongoing in Cork, and could be expanded nationally. The establishment of drug repositories required local legislative changes in many US states [97], and this is also likely to be necessary prior to the introduction of any schemes in Ireland.
Ireland is not unique in the challenges it faces, and Cortes et al. [98] have examined the global difficulties in accessing cancer medicines, including challenges that we are fortunate not to face, such as the marketing of substandard generics with limited regulatory oversight. They propose some of strategies that could be considered at an Irish or EU level, such as risk-sharing approaches or value-based pricing, or removing sales tax from all medicines. Oral medications are already VAT exempt; however, injectable products are not, which may increase financial toxicity and limit access for patients not covered by the drug payment scheme.
Ultimately while the methods discussed here may help optimise spending, as clinicians we must continue to advocate for appropriate allocation of budgetary resources, and remain mindful of what we prescribe, who we prescribe to, and when to stop prescribing.

Declarations

Not required (literature review only); no other ethical issues raised.

Conflict of interest

MH is a Breakthrough Cancer Fellowship Recipient. TC is a Breakthrough Cancer Summer Student Grant recipient. Educational sponsorship received from Jansen, MSD, Novartis.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
7.
Zurück zum Zitat Kelly D, Brady C, Sui J et al (2017) Cancer care costs and clinical trials. Ir Med J 110(4):557PubMed Kelly D, Brady C, Sui J et al (2017) Cancer care costs and clinical trials. Ir Med J 110(4):557PubMed
14.
Zurück zum Zitat Kelly C, Smith M, Flynn S et al (2016) Accrual to cancer clinical trial. Ir Med J 109:436PubMed Kelly C, Smith M, Flynn S et al (2016) Accrual to cancer clinical trial. Ir Med J 109:436PubMed
18.
Zurück zum Zitat Cronin T, Ronayne C, O’Donovan N et al (2023) The Impact of Expanded Access Programs for Systemic Anticancer Therapy in an Irish Cancer Centre. Ir J Med Sci Cronin T, Ronayne C, O’Donovan N et al (2023) The Impact of Expanded Access Programs for Systemic Anticancer Therapy in an Irish Cancer Centre. Ir J Med Sci
66.
73.
Zurück zum Zitat Bach PB, Conti RM, Muller RJ et al (2016) Overspending driven by oversized single dose vials of cancer drugs. BMJ 352CrossRef Bach PB, Conti RM, Muller RJ et al (2016) Overspending driven by oversized single dose vials of cancer drugs. BMJ 352CrossRef
75.
Zurück zum Zitat United States Pharmacopoeia (2022) General Chapter 797 Pharmaceutical Compounding – Sterile Preparations. USP-NF 2023:1 United States Pharmacopoeia (2022) General Chapter 797 Pharmaceutical Compounding – Sterile Preparations. USP-NF 2023:1
77.
Zurück zum Zitat Ho KV, Edwards MS, Solimando DA Jr, Johnson AD (2016) Determination of extended sterility for single-use vials using the phaseal closed-system transfer device. J Hematol Oncol Pharm 6(2):46–50 Ho KV, Edwards MS, Solimando DA Jr, Johnson AD (2016) Determination of extended sterility for single-use vials using the phaseal closed-system transfer device. J Hematol Oncol Pharm 6(2):46–50
91.
Zurück zum Zitat Young S, Zigmond M, Lee S (2015) Evaluating the effects of a 14-day oral chemotherapy dispensing protocol on adherence, toxicity, and cost. J Hematol Oncol Pharm 5:75–80 Young S, Zigmond M, Lee S (2015) Evaluating the effects of a 14-day oral chemotherapy dispensing protocol on adherence, toxicity, and cost. J Hematol Oncol Pharm 5:75–80
Metadaten
Titel
Optimising oncology drug expenditure in Ireland
verfasst von
Ruth Kieran
Maeve Hennessy
Kate Coakley
Hazel O’Sullivan
Tim Cronin
Daire Lynch
Eibhlin Mulroe
Katie Cooke
Dearbhaile Collins
Seamus O’Reilly
Publikationsdatum
03.04.2024
Verlag
Springer International Publishing
Erschienen in
Irish Journal of Medical Science (1971 -)
Print ISSN: 0021-1265
Elektronische ISSN: 1863-4362
DOI
https://doi.org/10.1007/s11845-024-03672-y

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.