Abstract

Background

Canada has an established publicly funded health care system with a complex drug approval and funding process. After proof of efficacy (POE; key publication/presentation) and before becoming publicly accessible, each drug undergoes a Health Canada approval process, a health technology assessment (HTA), a pricing negotiation, and finally individual provincial funding agreements. We quantified potential life‐years lost during this process.

Methods

We analyzed drugs for advanced lung, breast, and colorectal cancer that underwent the HTA process between 2011 and 2016. Life‐years lost were calculated by multiplying documented improvement in progression‐free and overall survival, number of eligible patients, and time from POE to first public funding. For conservative calculation, we assumed all eligible patients in Canada had access at the time of first public funding, whereas in reality provinces fund at different time points.

Results

We analyzed 21 drugs. Of these, 15 have been funded publicly. The time from POE to first public funding ranged from 14.0 to 99.2 months (median 26.6 months). Total overall life‐years lost from POE to first public funding were 39,067 (lung 32,367; breast 6,691). Progression‐free life‐years lost from POE to first public funding were 48,037 (lung 9,139, breast 15,827, colorectal 23,071).

Conclusion

The number of potential life‐years lost during the drug regulatory and funding process in Canada is substantial, largely driven by delays to funding of colorectal cancer drugs. Recognizing that interprovincial differences exist and that eligible patients may not all receive a given drug, if even a fraction does so, the impact of delays remains substantive. Collaborative national initiatives are required to address this major barrier to treatment access.

Implications for Practice

Patients may spend lengthy periods of time awaiting access to new and effective cancer drugs. Patients with private drug insurance or personal funds or who reside in certain Canadian provinces may obtain some drugs sooner than others, potentially creating a two‐tiered access system. The cancer drug access and public funding system must be expedited to improve equity.

Introduction

It is estimated that one in two Canadians will receive a diagnosis of cancer in their lifetime and that one in four Canadians will die of cancer [1]. Frequently, cancer is diagnosed as advanced disease [1]. Substantial advances in our understanding of cancer biology and treatment have improved survival rates for patients with advanced malignancy [2]. However, stringent regulations may limit or delay access to new, effective cancer therapies [3]. Previous literature demonstrates that for 21 drugs effective in advanced malignancies, 1 life‐year is lost worldwide for every 12‐second delay from the time of drug discovery to approval by the U.S. Food and Drug Administration [4].

Canada has a publicly funded health care system with a unique and complex cancer drug regulatory and funding process. After a cancer drug is shown to be effective in a clinical trial, it must receive Health Canada approval to be marketed in Canada. The Patented Medicines Prices Review Board ensures that the prices of patented drugs in Canada are not excessive [5]. With Health Canada approval, the drug can be sold in Canada but is not publicly reimbursed by the provincial or territorial payers. Before the provinces and territories of Canada make a decision on whether or not they will publicly fund a cancer drug, the drug undergoes a health technology assessment (HTA) by the pan‐Canadian Oncology Drug Review (pCODR) [6]. The goal of the pCODR HTA is to make a nonbinding, evidence‐based funding recommendation to the provinces and territories of Canada (except Quebec, whose Institut National d'Excellence en Sante et en Services Sociaux undertakes a different process). Ultimately, pCODR makes a final funding recommendation, which can be a recommendation to fund the drug, to fund conditionally, or to not fund the drug. Subsequently, the pan‐Canadian Pharmaceutical Alliance negotiates pricing with the manufacturer. Finally, each province or territory makes an independent decision as to whether or not it will publicly reimburse the drug by considering the pCODR recommendation and also its own priorities, mandates, and budget.

Recognizing that the cancer drug approval and funding process in Canada takes time during which patients with advanced cancer may not have public access to new life‐prolonging drugs, we calculated the potential life‐years lost during this process in Canada.

Materials and Methods

We calculated potential life‐years lost during the cancer drug approval and funding process in Canada from the time each drug was shown to be effective in a positive clinical trial to the first provincial public reimbursement.

The local research ethics board was consulted and determined that its approval was not needed for this study because all data points collected and analyzed were publicly available.

Endpoints

The primary endpoints were the total number of overall survival (OS) and progression‐free survival (PFS) life‐years lost from proof of efficacy to first public reimbursement.

Secondary endpoints included (a) the total number of OS and PFS life‐years lost from proof of efficacy to completion of the HTA and (b) time intervals from proof of efficacy to HTA submission, HTA submission to HTA completion, HTA completion to first public reimbursement, and from first to last provincial funding.

Eligibility Criteria

Drugs that were eligible for inclusion were all drugs used in the treatment of advanced lung, breast, and colorectal cancer submitted to the pCODR HTA from January 2011 to December 2016. All drugs required a documented statistically significant improvement in OS and/or PFS in comparison to the control arm in a randomized trial. Although drugs submitted to the pCODR HTA after December 2016 were not included in the analysis, data points for all included drugs were updated and current as of July 2018.

Calculating Life‐Years Lost

To calculate the number of potential life‐years lost during the cancer drug regulatory and funding process, we took the time interval from proof of drug efficacy to first public funding multiplied by the survival advantage of the drug in years multiplied by the number of potentially eligible patients annually.

The date of proof of drug efficacy was defined as the date of publication or presentation (whichever came first) of the registration trial [7-26] used by pCODR in its analysis [27]. When pCODR used multiple trials in its analysis, the date of the earliest trial was considered the date of proof of drug efficacy.

When calculating the time interval for the HTA [27], if an interruption or resubmission took place, the whole time interval, including the break, was calculated.

We defined the time point of first public funding as the date on which the first province agreed to publicly reimburse the drug [27]. In this way, we considered the date of first provincial reimbursement as a surrogate for national public reimbursement, recognizing that all provinces may fund drugs at different timepoints or not at all.

When calculating time intervals to first public funding, we included only drugs with public funding in at least one province. For calculations of all other time intervals (i.e., to pCODR submission and pCODR HTA completion) we included all drugs in the data set regardless of funding status.

When determining the number of patients potentially eligible to receive each drug annually, we defined this as the number of patients dying annually of the particular cancer type in Canada [28]. Where subgroups of patients typically receive a given drug (e.g., epidermal growth factor receptor–positive non‐small cell lung cancer or Kirsten rat sarcoma [KRAS] wild‐type colorectal cancer), the size of the subgroup was taken into account. This was done by multiplying the total number of patients dying of each main cancer type in 2017 (lung, breast, colorectal) [29-31] by the estimated proportion of each relevant subgroup (extrapolated from the published literature) [14,32,33-40]. For example, 9,400 patients died of colorectal cancer in 2017 [29]. Because approximately 58% of patients with colorectal cancer are characterized as having KRAS wild‐type cancer [40,41], we determined that 9,400 × 0.58 results in 5,424 patients dying of KRAS wild‐type colorectal cancer per year.

Results

We assessed 21 drugs. At the time of database lock, 15 drugs had received public funding in at least one province. The median OS benefit of the drugs analyzed was undefined for ten drugs and ranged from 1.5 to 15.8 months (median 3.2 months) for 11 drugs with available randomized comparisons. The median PFS benefit for the drugs analyzed was undefined for three drugs and ranged from 0.7 to 10.0 months (median 3.9 months) for 18 drugs with available randomized comparisons. The number of patients potentially eligible to receive each drug annually ranged from 515 to 12,555 (median 5,000). Table 1 shows the list of drugs analyzed, their median survival advantages, and the number of patients potentially eligible to receive each drug annually in Canada.

Table 1

Drug efficacy and eligibility characteristics

Tumor siteDrug nameIndicationMedian OS gain in monthsaMedian PFS gain in monthsaNumber of eligible patients per year
LungAfatinib [7]First line, EGFR+4.21,703
LungCeritinib [42]ALK+3.8538
LungCrizotinib [43]Second line, ALK+3.9538
LungCrizotinib [44]First line, ALK+4.7538
LungNivolumab [10]Second line, nonsquamous2.812,555
LungNivolumab [11]Second line, squamous3.20.75,380
LungOsimertinib [12]Second line, EGFR T790M+5.71,158
LungPembrolizumab [15]First line, expressing PD‐L1 > 50%15.84.35,380
LungPembrolizumab [14]Second line, expressing PD‐L14.211,837
LungPemetrexed [13]Maintenance after first‐line platinum doublet2.91.312,555
BreastEribulin [17]Third line2.55,000
BreastEverolimus [16]HR+, Her2/neu−4.13,635
BreastLapatinib [21]HR+, Her2/neu+5.2515
BreastPalbociclib [18]HR+10.03,635
BreastPertuzumab [20]Her2/neu+, in combination with a taxane/trastuzumab15.76.31,000
BreastTrastuzumab Emtansine [19]Her2/neu+, following trastuzumab5.83.21,000
ColorectalAflibercept [26]Second+ line1.52.29,400
ColorectalBevacizumab [22]First line, with capecitabine, when oxaliplatin and irinotecan are unsuitable2.89,400
ColorectalCetuximab [23]First line, KRAS WT3.51.55,424
ColorectalPanitumumab [25]First line, KRAS WT1.65,424
ColorectalRegorafenib [45]Second+ line2.51.59,400
Tumor siteDrug nameIndicationMedian OS gain in monthsaMedian PFS gain in monthsaNumber of eligible patients per year
LungAfatinib [7]First line, EGFR+4.21,703
LungCeritinib [42]ALK+3.8538
LungCrizotinib [43]Second line, ALK+3.9538
LungCrizotinib [44]First line, ALK+4.7538
LungNivolumab [10]Second line, nonsquamous2.812,555
LungNivolumab [11]Second line, squamous3.20.75,380
LungOsimertinib [12]Second line, EGFR T790M+5.71,158
LungPembrolizumab [15]First line, expressing PD‐L1 > 50%15.84.35,380
LungPembrolizumab [14]Second line, expressing PD‐L14.211,837
LungPemetrexed [13]Maintenance after first‐line platinum doublet2.91.312,555
BreastEribulin [17]Third line2.55,000
BreastEverolimus [16]HR+, Her2/neu−4.13,635
BreastLapatinib [21]HR+, Her2/neu+5.2515
BreastPalbociclib [18]HR+10.03,635
BreastPertuzumab [20]Her2/neu+, in combination with a taxane/trastuzumab15.76.31,000
BreastTrastuzumab Emtansine [19]Her2/neu+, following trastuzumab5.83.21,000
ColorectalAflibercept [26]Second+ line1.52.29,400
ColorectalBevacizumab [22]First line, with capecitabine, when oxaliplatin and irinotecan are unsuitable2.89,400
ColorectalCetuximab [23]First line, KRAS WT3.51.55,424
ColorectalPanitumumab [25]First line, KRAS WT1.65,424
ColorectalRegorafenib [45]Second+ line2.51.59,400

aSurvival gain in comparison to the study control arm.

Abbreviations: —, not applicable; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; KRAS, Kirsten rat sarcoma; OS, overall survival; PFS, progression‐free survival; PD‐L1, programmed death‐ligand 1; WT, wild type.

Table 1

Drug efficacy and eligibility characteristics

Tumor siteDrug nameIndicationMedian OS gain in monthsaMedian PFS gain in monthsaNumber of eligible patients per year
LungAfatinib [7]First line, EGFR+4.21,703
LungCeritinib [42]ALK+3.8538
LungCrizotinib [43]Second line, ALK+3.9538
LungCrizotinib [44]First line, ALK+4.7538
LungNivolumab [10]Second line, nonsquamous2.812,555
LungNivolumab [11]Second line, squamous3.20.75,380
LungOsimertinib [12]Second line, EGFR T790M+5.71,158
LungPembrolizumab [15]First line, expressing PD‐L1 > 50%15.84.35,380
LungPembrolizumab [14]Second line, expressing PD‐L14.211,837
LungPemetrexed [13]Maintenance after first‐line platinum doublet2.91.312,555
BreastEribulin [17]Third line2.55,000
BreastEverolimus [16]HR+, Her2/neu−4.13,635
BreastLapatinib [21]HR+, Her2/neu+5.2515
BreastPalbociclib [18]HR+10.03,635
BreastPertuzumab [20]Her2/neu+, in combination with a taxane/trastuzumab15.76.31,000
BreastTrastuzumab Emtansine [19]Her2/neu+, following trastuzumab5.83.21,000
ColorectalAflibercept [26]Second+ line1.52.29,400
ColorectalBevacizumab [22]First line, with capecitabine, when oxaliplatin and irinotecan are unsuitable2.89,400
ColorectalCetuximab [23]First line, KRAS WT3.51.55,424
ColorectalPanitumumab [25]First line, KRAS WT1.65,424
ColorectalRegorafenib [45]Second+ line2.51.59,400
Tumor siteDrug nameIndicationMedian OS gain in monthsaMedian PFS gain in monthsaNumber of eligible patients per year
LungAfatinib [7]First line, EGFR+4.21,703
LungCeritinib [42]ALK+3.8538
LungCrizotinib [43]Second line, ALK+3.9538
LungCrizotinib [44]First line, ALK+4.7538
LungNivolumab [10]Second line, nonsquamous2.812,555
LungNivolumab [11]Second line, squamous3.20.75,380
LungOsimertinib [12]Second line, EGFR T790M+5.71,158
LungPembrolizumab [15]First line, expressing PD‐L1 > 50%15.84.35,380
LungPembrolizumab [14]Second line, expressing PD‐L14.211,837
LungPemetrexed [13]Maintenance after first‐line platinum doublet2.91.312,555
BreastEribulin [17]Third line2.55,000
BreastEverolimus [16]HR+, Her2/neu−4.13,635
BreastLapatinib [21]HR+, Her2/neu+5.2515
BreastPalbociclib [18]HR+10.03,635
BreastPertuzumab [20]Her2/neu+, in combination with a taxane/trastuzumab15.76.31,000
BreastTrastuzumab Emtansine [19]Her2/neu+, following trastuzumab5.83.21,000
ColorectalAflibercept [26]Second+ line1.52.29,400
ColorectalBevacizumab [22]First line, with capecitabine, when oxaliplatin and irinotecan are unsuitable2.89,400
ColorectalCetuximab [23]First line, KRAS WT3.51.55,424
ColorectalPanitumumab [25]First line, KRAS WT1.65,424
ColorectalRegorafenib [45]Second+ line2.51.59,400

aSurvival gain in comparison to the study control arm.

Abbreviations: —, not applicable; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; KRAS, Kirsten rat sarcoma; OS, overall survival; PFS, progression‐free survival; PD‐L1, programmed death‐ligand 1; WT, wild type.

The majority of drugs analyzed (76%) received a positive pCODR recommendation. All drugs with a negative pCODR recommendation remain unfunded in all provinces to date.

The median time from proof of efficacy to first public funding was 26.6 months (range 14.0–99.2 months). Analyzing each component of the process, the median time from efficacy to the HTA submission was 12.2 months (range 0–68.8 months); median time for the HTA process itself was 8.7 months (range 5.6–33.5 months); median time from HTA completion to first public funding was 6.9 months (range 2.1–24.9 months). Figure 1 details the component time delays for each drug.

Delay from proof of efficacy to HTA process to first public funding.
Figure 1

Delay from proof of efficacy to HTA process to first public funding.

Abbreviations: ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; HTA, health technology assessment; KRAS, Kirsten rat sarcoma; PD‐L1, programmed death‐ligand 1; WT, wild type.

For all drugs with a PFS advantage (n = 18), total PFS life‐years lost from proof of efficacy to HTA completion were 50,481 (lung cancer 8,596, breast cancer 12,393, colorectal cancer 29,492). Delineating PFS life‐years lost for each component of the process, from proof of efficacy to start of HTA, 35,443 life‐years were lost, and an additional 15,038 life‐years were lost during the HTA process itself. Figure 2A shows a breakdown of progression‐free survival life‐years lost for efficacy to HTA completion for all drugs.

Potential life‐years lost from efficacy to completion of the health technology assessment (HTA) and first public funding. (A): Progression‐free survival (PFS) life‐years lost rom efficacy to HTA. (B): PFS life‐years lost from efficacy to funding. (C): Overall survival (OS) life‐years lost from efficacy to HTA. (D): OS life‐years lost from efficacy to funding.
Figure 2

Potential life‐years lost from efficacy to completion of the health technology assessment (HTA) and first public funding. (A): Progression‐free survival (PFS) life‐years lost rom efficacy to HTA. (B): PFS life‐years lost from efficacy to funding. (C): Overall survival (OS) life‐years lost from efficacy to HTA. (D): OS life‐years lost from efficacy to funding.

Abbreviations: ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; KRAS, Kirsten rat sarcoma; PD‐L1, programmed death‐ligand 1; WT, wild type.

Considering only drugs with a PFS advantage that were funded in at least one province (n = 12), total PFS life‐years lost from efficacy to first public funding were 48,037 (lung cancer 9,139, breast cancer 15,827, colorectal cancer 23,071). Figure 2B shows a breakdown of progression‐free life‐years lost from efficacy to public funding for each drug.

For all drugs with an OS advantage (n = 11), total OS life‐years lost from proof of efficacy to HTA completion were 45,218 (lung cancer 22,494, breast cancer 5,379, colorectal cancer 17,345). Delineating OS life‐years lost for each component of the process, from proof of efficacy to start of HTA, 24,756 life‐years were lost, and an additional 20,461 life‐years were lost for the HTA process itself. Figure 2C shows a breakdown of overall survival life‐years lost from efficacy to HTA completion for all drugs.

Considering only drugs with an OS advantage that were funded in at least one province (n = 8), total overall survival life‐years lost were 39,067 (lung cancer 32,376, breast cancer 6,691). Figure 2D shows a breakdown of overall survival life‐years lost from efficacy to first public funding for each drug.

For drugs that were funded in more than one province (n = 15), time intervals from first to last provincial funding ranged from 2.8 to 23.8 months (median 6.2 months). Table 2 shows time intervals from first to last provincial funding for all drugs funded in at least two provinces.

Table 2

Time intervals from first to last provincial funding

Drug (indication)Months from first to last provincial funding
Lung cancer
Afatinib (first line, EGFR+)9.4
Crizotinib (first line, ALK+)4.4
Crizotinib (second line, ALK+)6.2
Nivolumab (second line, nonsquamous)5.1
Nivolumab (second line, squamous)5.1
Pembrolizumab (first line, PD‐L1+)5.7
Pembrolizumab (second line, PD‐L1+)5.7
Pemetrexed (maintenance)20.7
Breast cancer
Eribulin (third+ line)23.8
Everolimus (HR+)10.8
Palbociclib (HR+)2.8
Pertuzumab (Her2/neu+)17.8
Trastuzumab emtansine (second line, Her2/neu+)7.7
Colorectal cancer
Bevacizumab (first line, with capecitabine)4.3
Panitumumab (first line, KRAS WT)15.0
Drug (indication)Months from first to last provincial funding
Lung cancer
Afatinib (first line, EGFR+)9.4
Crizotinib (first line, ALK+)4.4
Crizotinib (second line, ALK+)6.2
Nivolumab (second line, nonsquamous)5.1
Nivolumab (second line, squamous)5.1
Pembrolizumab (first line, PD‐L1+)5.7
Pembrolizumab (second line, PD‐L1+)5.7
Pemetrexed (maintenance)20.7
Breast cancer
Eribulin (third+ line)23.8
Everolimus (HR+)10.8
Palbociclib (HR+)2.8
Pertuzumab (Her2/neu+)17.8
Trastuzumab emtansine (second line, Her2/neu+)7.7
Colorectal cancer
Bevacizumab (first line, with capecitabine)4.3
Panitumumab (first line, KRAS WT)15.0

Abbreviations: ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; KRAS, Kirsten rat sarcoma; D‐L1, programmed death‐ligand 1; WT, wild type.

Table 2

Time intervals from first to last provincial funding

Drug (indication)Months from first to last provincial funding
Lung cancer
Afatinib (first line, EGFR+)9.4
Crizotinib (first line, ALK+)4.4
Crizotinib (second line, ALK+)6.2
Nivolumab (second line, nonsquamous)5.1
Nivolumab (second line, squamous)5.1
Pembrolizumab (first line, PD‐L1+)5.7
Pembrolizumab (second line, PD‐L1+)5.7
Pemetrexed (maintenance)20.7
Breast cancer
Eribulin (third+ line)23.8
Everolimus (HR+)10.8
Palbociclib (HR+)2.8
Pertuzumab (Her2/neu+)17.8
Trastuzumab emtansine (second line, Her2/neu+)7.7
Colorectal cancer
Bevacizumab (first line, with capecitabine)4.3
Panitumumab (first line, KRAS WT)15.0
Drug (indication)Months from first to last provincial funding
Lung cancer
Afatinib (first line, EGFR+)9.4
Crizotinib (first line, ALK+)4.4
Crizotinib (second line, ALK+)6.2
Nivolumab (second line, nonsquamous)5.1
Nivolumab (second line, squamous)5.1
Pembrolizumab (first line, PD‐L1+)5.7
Pembrolizumab (second line, PD‐L1+)5.7
Pemetrexed (maintenance)20.7
Breast cancer
Eribulin (third+ line)23.8
Everolimus (HR+)10.8
Palbociclib (HR+)2.8
Pertuzumab (Her2/neu+)17.8
Trastuzumab emtansine (second line, Her2/neu+)7.7
Colorectal cancer
Bevacizumab (first line, with capecitabine)4.3
Panitumumab (first line, KRAS WT)15.0

Abbreviations: ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; Her2/neu, human epidermal growth factor receptor 2; HR, hormone receptor; KRAS, Kirsten rat sarcoma; D‐L1, programmed death‐ligand 1; WT, wild type.

Discussion

An improved understanding of cancer biology and the advent of molecularly targeted therapies and other novel drug classes are helping patients with advanced malignancy live longer without the toxicities of conventional chemotherapy [2]; however, the prices for cancer drugs are increasing [46] because of the enormous costs of drug development and market factors [5,47-49]. It is clear that careful consideration is required when selecting drugs for public reimbursement in the context of a publicly funded health care system.

Notwithstanding the importance of insightful decision making and a process to do so, the number of potential life‐years lost during the cancer drug regulatory and funding process in Canada is substantial. We calculated a total of 39,076 overall survival life‐years lost and 48,037 progression‐free life‐years lost from the time of proof of drug efficacy to first public reimbursement for 21 drugs used in advanced lung, breast, and colorectal cancer. Although we attempted to make the most conservative calculations possible, we recognize that these calculations represent estimates. However, even if the true value is only a fraction of our calculations, the loss remains substantial.

Considering the time from proof of efficacy to first public reimbursement, we found that the longest time interval occurred while awaiting drug submission to pCODR by the drug manufacturer. The median time delay while awaiting submission to pCODR was approximately 1 year. Determining reasons for delayed submission to pCODR was beyond the scope of this analysis, but we can hypothesize that Canada is a smaller market than other countries and therefore may not be a priority for drug marketing by pharmaceutical companies; in fact, it is known that drug applications are submitted to Health Canada a median of about 1 year later than they are submitted to the U.S. Food and Drug Administration and the European Medicines Agency [50].

The pCODR process was implemented in 2011 by the Ministries of Health (except Quebec), replacing the interim Joint Oncology Drug Review [5], with the goal of informing provincial reimbursement decisions based on clinical efficacy and cost‐effectiveness [51]. Although previous research suggests that pCODR improved the median time from Health Canada approval to public funding from 522 to 393 days [52], the median time required for the HTA was 141 days (range 112–282) and only 50% met the target of 140 days [53]. In our present analysis, the median time for the pCODR HTA was longer, at close to 9 months. This discrepency likely results from different methodologies used to calculate time intervals for the pCODR HTA and the selection of included drugs.

Although we considered time to first provincial reimbursement as a surrogate for public access nationwide in order to make the most conservative possible estimates of potential life‐years lost, in reality, provinces and territories all fund drugs at different time points (if at all). This lends itself to the potential for great variability among provinces and territories, and we see in the present study that the time delay from first to last provincial funding may be as long as nearly 2 years. Substantial variability in provincial funding decisions is supported by previous research [5,53]. In a survey evaluating provincial discordance in drug funding decisions, over 90% of policymakers stated that the most common reason to not fund a drug recommended by pCODR was budget constraints [54]. The Canadian Federal Government is currently investigating the possibility of a national pharmacare system, which may have the potential to eradicate these interprovincial discrepancies [55].

In some ways, our calculations are underestimates of the true number of life‐years lost. We did not take into account additional life‐years lost after the first provincial funding while awaiting all subsequent provinces to fund. We were also unable to calculate overall survival life‐years lost for drugs without a documented OS advantage in a clinical trial because of crossover. To compensate for this, we calculated progression‐free life‐years lost. Although PFS is not a reliable surrogate for OS [56], progression‐free years may be meaningful from a patient perspective because longer PFS may improve health‐related quality of life [57-59], although this may be considered controversial. Progression‐free life‐years lost are not as meaningful as overall survival life‐years lost, as they do not describe actual years of life lost, but we believe they nonetheless help us to describe the cost of the drug funding process in patient‐centered terms.

Although our calculations are underestimates in some ways, in other ways our calculations are overestimates of the true number of life‐years lost. While awaiting approval by Health Canada for marketing in the country, Health Canada may allow limited release of a drug under its Special Access Program. Some patients may access these drugs through other clinical trials. However, most public drug plans do not cover the cost of such drugs prior to approval [5], so we did not take into account the availability of special access programs in our calculations. Similarly, we did not consider available means of funding aside from public reimbursement such as compassionate use programs, private insurers, or use of personal funds. Although some patients may access drugs sooner than others through such avenues, this may create a two‐tiered access system and does not represent equitable public availability of drugs. In addition to investigating two‐tiered access within Canada, future study should also assess variations in drug access among peer nations. This would require a thorough understanding of the important trials considered during the regulatory and approval process in the United States, Europe, and Japan, to name a few.

We recognize that calculating the time from proof of efficacy to public funding ignores the fact that regulatory approval by Health Canada is an important step in the process and may occur long after the publication or presentation of the registration trial. Using the date of registration trial publication or presentation may have therefore inflated our calculations of life‐years lost. Although the date of first market access guarantees revenue from sale of a given drug, this date is not as meaningful from a patient perspective, as patients yearn to access drugs the moment clinical trials prove their efficacy.

Although we calculated the life‐years lost for all potentially eligible patients, we recognize that not all eligible patients may receive a given drug because of factors such as performance status and patient choice. There is patient attrition with each line of systemic therapy [60], and some patients may not receive a referral to a medical oncologist at all [61]. As such, the number of patients who would actually receive a given drug is likely a smaller group than the number potentially eligible to receive it.

Finally, some of the drugs we investigated could be considered “me too” drugs, so although there was a delay in approval, patients may have had access to an alternative and comparable option. We did not consider “life‐years saved” by the availability of similar alternative (publicly funded) agents.

Conclusion

Although a cancer drug regulatory and funding process is necessary in the context of any publicly funded health care system, we highlight the need to refine our processes by developing national initiatives to minimize this barrier to treatment access. This study highlights the many moving parts in the process. Initiatives are needed, including more rapid applications into the process, a nimble HTA that consistently meets its own time targets and can adapt to the evolving clinical trial environment, and a post‐HTA process that allows transparency in cost negotiation and equality between provinces. Many initiatives are currently being explored, and we hope that they will help expedite the cancer drug funding process and improve equity across the provinces and territories of Canada.

Acknowledgments

The authors thank Louiselle Godbout and Chris Booth.

Author Contributions

Conception/design: Joanna Gotfrit, David J. Stewart, Paul Wheatley‐Price

Collection and/or assembly of data: Joanna Gotfrit, John J.W. Shin

Data analysis and interpretation: Joanna Gotfrit, Ranjeeta Mallick

Manuscript writing: Joanna Gotfrit, John J.W. Shin, Ranjeeta Mallick, David J. Stewart, Paul Wheatley‐Price

Final approval of manuscript: Joanna Gotfrit, John J.W. Shin, Ranjeeta Mallick, David J. Stewart, Paul Wheatley‐Price

Disclosures

David J. Stewart: Roche Canada, Boehringer‐Ingelheim Canada, Merck Canada, AstraZeneca Canada, Bristol‐Myers Squibb Canada (H, C/A), Celgene (RF); Paul Wheatley‐Price: Takeda, Roche, AstraZeneca, Merck, Abbvie, Bristol‐Myers Squibb (SAB). The other authors indicated no financial relationships.

(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board

References

1

Canadian Cancer Statistics Advisory Committee
.
Canadian Cancer Statistics: A 2018 Special Report on Cancer Incidence by Stage
.
Toronto, ON, Canada
:
Canadian Cancer Society
;
2018
. Available at cancer.ca/Canadian-Cancer-Statistics-2018-EN. Accessed September 17, 2018.

2

Miller
 
KD
,
Siegel
 
RL
,
Lin
 
CC
et al.
Cancer treatment and survivorship statistics, 2016
.
CA Cancer J Clin
 
2016
;
66
:
271
289
.

3

Stewart
 
DJ
,
Whitney
 
SN
,
Kurzrock
 
R
.
Equipoise lost: Ethics, costs, and the regulation of cancer clinical research
.
J Clin Oncol
 
2010
;
28
:
2925
2935
.

4

Stewart
 
DJ
,
Stewart
 
AA
,
Wheatley‐Price
 
P
et al.
The importance of greater speed in drug development for advanced malignancies
.
Cancer Med
 
2018
;
7
:
1824
1836
.

5

Issues of Access to Cancer Drugs in Canada. A Report for the Canadian Cancer Action Network
.
Toronto, ON, Canada
:
Canadian Cancer Action Network
;
2008
.

6

CADTH pan‐Canadian Oncology Drug Review. Canadian Association of Drugs and Technologies in Health (CADTH) Web site. Available at https://cadth.ca/pcodr. Updated 2016. Accessed 2016.

7

Sequist
 
LV
,
Yang
 
JC
,
Yamamoto
 
N
et al.
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
 
2013
;
31
:
3327
3334
.

8

Crinò
 
L
,
Kim
 
D
,
Riely
 
GJ
et al.
Initial phase II results with crizotinib in advanced ALK‐positive non‐small cell lung cancer (NSCLC): PROFILE 1005
.
J Clin Oncol
 
2011
;
29
(
suppl 15
):
7514A
.

9

Shaw
 
AT
,
Kim
 
DW
,
Mehra
 
R
et al.
Ceritinib in ALK‐rearranged non‐small‐cell lung cancer
.
N Engl J Med
 
2014
;
370
:
1189
1197
.

10

Borghaei
 
H
,
Paz‐Ares
 
L
,
Horn
 
L
et al.
Nivolumab versus docetaxel in advanced nonsquamous non‐small‐cell lung cancer
.
N Engl J Med
 
2015
;
373
:
1627
1639
.

11

Brahmer
 
J
,
Reckamp
 
KL
,
Baas
 
P
et al.
Nivolumab versus docetaxel in advanced squamous‐cell non‐small‐cell lung cancer
.
N Engl J Med
 
2015
;
373
:
123
135
.

12

Mok
 
TS
,
Wu
 
YL
,
Ahn
 
MJ
et al.
Osimertinib or platinum‐pemetrexed in EGFR T790M‐positive lung cancer
.
N Engl J Med
 
2017
;
376
:
629
640
.

13

Paz‐Ares
 
L
,
de Marinis
 
F
,
Dediu
 
M
et al.
Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non‐squamous non‐small‐cell lung cancer (PARAMOUNT): A double‐blind, phase 3, randomised controlled trial
.
Lancet Oncol
 
2012
;
13
:
247
255
.

14

Herbst
 
RS
,
Baas
 
P
,
Kim
 
DW
et al.
Pembrolizumab versus docetaxel for previously treated, PD‐L1‐positive, advanced non‐small‐cell lung cancer (KEYNOTE‐010): A randomised controlled trial
.
Lancet
 
2016
;
387
:
1540
1550
.

15

Reck
 
M
,
Rodriguez‐Abreu
 
D
,
Robinson
 
AG
et al.
Pembrolizumab versus chemotherapy for PD‐L1‐positive non‐small‐cell lung cancer
.
N Engl J Med
 
2016
;
375
:
1823
1833
.

16

Baselga
 
J
,
Campone
 
M
,
Piccart
 
M
et al.
Everolimus in postmenopausal hormone‐receptor‐positive advanced breast cancer
.
N Engl J Med
 
2012
;
366
:
520
529
.

17

Cortes
 
J
,
O'Shaughnessy
 
J
,
Loesch
 
D
et al.
Eribulin monotherapy versus treatment of physician's choice in patients with metastatic breast cancer (EMBRACE): A phase 3 open‐label randomised study
.
Lancet
 
2011
;
377
:
914
923
.

18

Finn
 
RS
,
Crown
 
JP
,
Lang
 
I
et al.
The cyclin‐dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first‐line treatment of oestrogen receptor‐positive, HER2‐negative, advanced breast cancer (PALOMA‐1/TRIO‐18): A randomised phase 2 study
.
Lancet Oncol
 
2015
;
16
:
25
35
.

19

Verma
 
S
,
Miles
 
D
,
Gianni
 
L
et al.
Trastuzumab emtansine for HER2‐positive advanced breast cancer
.
N Engl J Med
 
2012
;
367
:
1783
1791
.

20

Baselga
 
J
,
Cortes
 
J
,
Kim
 
SB
et al.
Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer
.
N Engl J Med
 
2012
;
366
:
109
119
.

21

Johnston
 
S
,
Pippen
 
J
 Jr
,
Pivot
 
X
et al.
Lapatinib combined with letrozole versus letrozole and placebo as first‐line therapy for postmenopausal hormone receptor‐positive metastatic breast cancer
.
J Clin Oncol
 
2009
;
27
:
5538
5546
.

22

Tebbutt
 
NC
,
Wilson
 
K
,
Gebski
 
VJ
et al.
Capecitabine, bevacizumab, and mitomycin in first‐line treatment of metastatic colorectal cancer: Results of the Australasian Gastrointestinal Trials Group Randomized Phase III MAX Study
.
J Clin Oncol
 
2010
;
28
:
3191
3198
.

23

Van Cutsem
 
E
,
Kohne
 
CH
,
Hitre
 
E
et al.
Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer
.
N Engl J Med
 
2009
;
360
:
1408
1417
.

24

Grothey
 
A
,
Van Cutsem
 
E
,
Sobrero
 
A
et al.
Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo‐controlled, phase 3 trial
.
Lancet
 
2013
;
381
:
303
312
.

25

Douillard
 
JY
,
Siena
 
S
,
Cassidy
 
J
et al.
Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first‐line treatment in patients with previously untreated metastatic colorectal cancer: The PRIME study
.
J Clin Oncol
 
2010
;
28
:
4697
4705
.

26

Van Cutsem
 
E
,
Tabernero
 
J
,
Lakomy
 
R
et al.
Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin‐based regimen
.
J Clin Oncol
 
2012
;
30
:
3499
3506
.

27

Find a review (pCODR). Canadian Agency for Drugs and Technologies in Health (CADTH) Web site. Available at https://www.cadth.ca/pcodr/find-a-review. Updated 2018. Accessed 2018.

28

Canadian Cancer Society's Advisory Committee on Cancer Statistics
.
Canadian Cancer Statistics 2016
.
Toronto, ON, Canada
:
Canadian Cancer Society
;
2016
.

29

Colorectal cancer statistics: Incidence and mortality 2017. Canadian Cancer Society Web site. Available at http://www.cancer.ca/en/cancer‐information/cancer‐type/colorectal/statistics/?region=on. Accessed 2018.

30

Breast cancer statistics: Incidence and mortality 2017. Canadian Cancer Society Web site. Available at http://www.cancer.ca/en/cancer‐information/cancer‐type/breast/statistics/?region=on. Accessed 2018.

31

Canadian Cancer Society. Lung cancer statistics: Incidence and mortality 2017. Canadian Cancer Society Web site. Available at http://www.cancer.ca/en/cancer‐information/cancer‐type/lung/statistics/?region=on. Accessed 2018.

32

What is non‐small cell lung cancer? American Cancer Society Web site. Available at https://www.cancer.org/cancer/non‐small‐cell‐lung‐cancer/about/what‐is‐non‐small‐cell‐lung‐cancer.html. Accessed 2018.

33

Pao
 
W
,
Miller
 
VA
.
Epidermal growth factor receptor mutations, small‐molecule kinase inhibitors, and non‐small‐cell lung cancer: Current knowledge and future directions
.
J Clin Oncol
 
2005
;
23
:
2556
2568
.

34

Koivunen
 
JP
,
Mermel
 
C
,
Zejnullahu
 
K
et al.
EML4‐ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer
.
Clin Cancer Res
 
2008
;
14
:
4275
4283
.

35

Reck
 
M
,
Rodriguez‐Abreu
 
D
,
Robinson
 
AG
et al.
Appendix of pembrolizumab versus chemotherapy for PD‐L1‐positive non‐small‐cell lung cancer
.
N Engl J Med
 
2016
;
375
:
1823
1833
.

36

Arcila
 
ME
,
Oxnard
 
GR
,
Nafa
 
K
et al.
Rebiopsy of lung cancer patients with acquired resistance to EGFR inhibitors and enhanced detection of the T790M mutation using a locked nucleic acid‐based assay
.
Clin Cancer Res
 
2011
;
17
:
1169
1180
.

37

Anderson
 
WF
,
Katki
 
HA
,
Rosenberg
 
PS
.
Incidence of breast cancer in the United States: Current and future trends
.
J Natl Cancer Inst
 
2011
;
103
:
1397
1402
.

38

Wolff
 
AC
,
Hammond
 
ME
,
Schwartz
 
JN
et al.
American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer
.
J Clin Oncol
 
2007
;
25
:
118
145
.

39

Howlader
 
N
,
Altekruse
 
SF
,
Li
 
CI
et al.
US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status
.
J Natl Cancer Inst
 
2014
;
106
:
dju055
.

40

Karapetis
 
CS
,
Khambata‐Ford
 
S
,
Jonker
 
DJ
et al.
K‐ras mutations and benefit from cetuximab in advanced colorectal cancer
.
N Engl J Med
 
2008
;
359
:
1757
1765
.

41

Tan
 
C
,
Du
 
X
.
KRAS mutation testing in metastatic colorectal cancer
.
World J Gastroenterol
 
2012
;
18
:
5171
5180
.

42

Shaw
 
AT
,
Kim
 
TM
,
Crino
 
L
et al.
Ceritinib versus chemotherapy in patients with ALK‐rearranged non‐small‐cell lung cancer previously given chemotherapy and crizotinib (ASCEND‐5): A randomised, controlled, open‐label, phase 3 trial
.
Lancet Oncol
 
2017
;
18
:
874
886
.

43

Shaw
 
AT
,
Kim
 
DW
,
Nakagawa
 
K
et al.
Crizotinib versus chemotherapy in advanced ALK‐positive lung cancer
.
N Engl J Med
 
2013
;
368
:
2385
2394
.

44

Solomon
 
BJ
,
Mok
 
T
,
Kim
 
DW
et al.;
PROFILE 1014 Investigators
.
First‐line crizotinib versus chemotherapy in ALK‐positive lung cancer [correction appears in N Engl J Med 2015;373:1582]
.
N Engl J Med
 
2014
;
371
:
2167
2177
.

45

Li
 
J
,
Qin
 
S
,
Xu
 
R
et al.
Regorafenib plus best supportive care versus placebo plus best supportive care in Asian patients with previously treated metastatic colorectal cancer (CONCUR): A randomised, double‐blind, placebo‐controlled, phase 3 trial
.
Lancet Oncol
 
2015
;
16
:
619
629
.

46

Rimer
 
BK
. The imperative of addressing cancer drug costs and value. National Cancer Institute Web site. Available at https://www.cancer.gov/news‐events/cancer‐currents‐blog/2018/presidents‐cancer‐panel‐drug‐prices. Published March 15, 2018. Accessed April 14, 2018.

47

Bach
 
PB
.
Limits on Medicare's ability to control rising spending on cancer drugs
.
N Engl J Med
 
2009
;
360
:
626
633
.

48

Howard
 
DH
,
Bach
 
PB
,
Berndt
 
ER
et al.
Pricing in the market for anticancer drugs
.
J Econ Perspect
 
2015
;
29
:
139
162
.

49

Cressman
 
S
,
Browman
 
GP
,
Hoch
 
JS
et al.
A time‐trend economic analysis of cancer drug trials
.
The Oncologist
 
2015
;
20
:
729
736
.

50

Rawson
 
NSB
.
Canadian, European and United States new drug approval times now relatively similar
.
Regul Toxicol Pharmacol
 
2018
;
96
:
121
126
.

51

Hoch
 
JS
,
Sabharwal
 
M
.
Informing Canada's cancer drug funding decisions with scientific evidence and patient perspectives: The Pan‐Canadian Oncology Drug Review
.
Curr Oncol
 
2013
;
20
:
121
124
.

52

Srikanthan
 
A
,
Mai
 
H
,
Penner
 
N
et al.
Impact of the pan‐Canadian Oncology Drug Review on provincial concordance with respect to cancer drug funding decisions and time to funding
.
Curr Oncol
 
2017
;
24
:
295
301
.

53

Rawson
 
NSB
.
Has pCODR Improved Access to Oncology Drugs? Timeliness and Provincial Acceptance of pan‐Canadian Oncology Drug Review Recommendations
.
Vancouver, BC, Canada
:
Fraser Institute
; July
2014
. Available at https://www.fraserinstitute.org/research/has‐pcodr‐improved‐access‐oncology‐drugs. Accessed March 3, 2018.

54

Srikanthan
 
A
,
Penner
 
N
,
Chan
 
KKW
et al.
Understanding the reasons for provincial discordance in cancer drug funding‐a survey of policymakers
.
Curr Oncol
 
2018
;
25
:
257
261
.

55

Government of Canada launches Advisory Council on the Implementation of National Pharmacare [press release]
.
Ottawa, ON, Canada
:
Health Canada
, June 20,
2018
.

56

Michiels
 
S
,
Saad
 
ED
,
Buyse
 
M
.
Progression‐free survival as a surrogate for overall survival in clinical trials of targeted therapy in advanced solid tumors
.
Drugs
 
2017
;
77
:
713
719
.

57

Axelrod
 
RC
.
The significance of progression‐free survival as a clinical end point in oncology
.
Drug Benefit Trends
 
2010
;
22
:
39
45
.

58

Hurvitz
 
SA
,
Lalla
 
D
,
Crosby
 
RD
et al.
Use of the metastatic breast cancer progression (MBC‐P) questionnaire to assess the value of progression‐free survival for women with metastatic breast cancer
.
Breast Cancer Res Treat
 
2013
;
142
:
603
609
.

59

Griebsch
 
I
,
Palmer
 
M
,
Fayers
 
PM
et al.
Is progression‐free survival associated with a better health‐related quality of life in patients with lung cancer? Evidence from two randomised trials with afatinib
.
BMJ Open
 
2014
;
4
:e005762.

60

Yang
 
F
,
Chen
 
L
,
Cheung
 
W
.
Attrition in the treatment of metastatic non‐small cell lung cancer
.
Int J Radiat Oncol Biol Phys
 
2017
;
98
:
237
.

61

Sacher
 
AG
,
Le
 
LW
,
Lau
 
A
et al.
Real‐world chemotherapy treatment patterns in metastatic non‐small cell lung cancer: Are patients undertreated?
 
Cancer
 
2015
;
121
:
2562
2569
.

Author notes

Disclosures of potential conflicts of interest may be found at the end of this article.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)