Diabetes (type 2) - canagliflozin: appraisal consultation document

The Department of Health has asked the National Institute for Health and Care Excellence (NICE) to produce guidance on using canagliflozin in the NHS in England. The Appraisal Committee has considered the evidence submitted by the manufacturer and the views of non-manufacturer consultees and commentators, and clinical specialists and patient experts.

This document has been prepared for consultation with the consultees. It summarises the evidence and views that have been considered, and sets out the draft recommendations made by the Committee. NICE invites comments from the consultees and commentators for this appraisal (see section 9) and the public. This document should be read along with the evidence base (the evaluation report).

The Appraisal Committee is interested in receiving comments on the following:

  • Has all of the relevant evidence been taken into account?
  • Are the summaries of clinical and cost effectiveness reasonable interpretations of the evidence?
  • Are the provisional recommendations sound and a suitable basis for guidance to the NHS?
  • Are there any aspects of the recommendations that need particular consideration to ensure we avoid unlawful discrimination against any group of people on the grounds of race, gender, disability, religion or belief, sexual orientation, age, gender reassignment, pregnancy and maternity?

Note that this document is not NICE's final guidance on this technology. The recommendations in section 1 may change after consultation.

After consultation:

  • The Appraisal Committee will meet again to consider the evidence, this appraisal consultation document and comments from the consultees.
  • At that meeting, the Committee will also consider comments made by people who are not consultees.
  • After considering these comments, the Committee will prepare the final appraisal determination (FAD).
  • Subject to any appeal by consultees, the FAD may be used as the basis for NICE’s guidance on using canagliflozin in the NHS in England.

For further details, see the Guides to the technology appraisal process.

The key dates for this appraisal are:

Closing date for comments: 17 March 2014

Second Appraisal Committee meeting: 25 March 2014

Details of membership of the Appraisal Committee are given in section 8, and a list of the sources of evidence used in the preparation of this document is given in section 9.


Note that this document is not NICE's final guidance on this technology. The recommendations in section 1 may change after consultation.

1 Appraisal Committee's preliminary recommendations

1.1 Canagliflozin in a dual therapy regimen in combination with metformin is recommended as an option for treating type 2 diabetes, only if:

  • the person is at significant risk of hypoglycaemia or its consequences or
  • a sulfonylurea is contraindicated or not tolerated.

1.2 Canagliflozin in a triple therapy regimen in combination with metformin and a sulfonylurea is recommended as an option for treating type 2 diabetes.

1.3 Canagliflozin in combination with insulin with or without other antidiabetic drugs is recommended as an option for treating type 2 diabetes.

1.4 People currently receiving treatment initiated within the NHS with canagliflozin that is not recommended for them by NICE in this guidance should be able to continue treatment until they and their NHS clinician consider it appropriate to stop.

2 The technology

2.1 Canagliflozin (Invokana, Janssen-Cilag) is an orally administered selective sodium–glucose cotransporter-2 (SGLT-2) inhibitor. It lowers blood glucose in people with type 2 diabetes by blocking the reabsorption of glucose in the kidneys and promoting excretion of excess glucose in the urine.

2.2 Canagliflozin has a European marketing authorisation for treating type 2 diabetes in adults aged 18 years and older to improve glycaemic control as:

  • monotherapy 'when diet and exercise alone do not provide adequate glycaemic control in patients for whom the use of metformin is considered inappropriate due to intolerance or contraindications'.
  • add-on therapy 'with other glucose-lowering medicinal products including insulin, when these, together with diet and exercise, do not provide adequate glycaemic control'.

The recommended starting dosage of canagliflozin is 100 mg once daily. In patients tolerating canagliflozin 100 mg once daily who have an estimated glomerular filtration rate (eGFR) of at least 60 ml/min/1.73 m2 or creatinine clearance (CrCl) of at least 60 ml/min and need tighter glycaemic control, the dosage can be increased to 300 mg once daily. For patients with renal impairment, the summary of product characteristics notes that canagliflozin should not be initiated in patients with an eGFR of less than 60 ml/min/1.73m2 or CrCl of less than 60 ml/min. In patients tolerating canagliflozin whose eGFR falls persistently below 60 ml/min/1.73 m2 or whose CrCl persistently falls below 60 ml/min, the dose of canagliflozin should be adjusted to or maintained at 100 mg once daily. Canagliflozin should be discontinued when eGFR is persistently below 45 ml/min/1.73 m2 or CrCl is persistently below 45 ml/min.

2.3 The summary of product characteristics states the following adverse reactions for canagliflozin are the most commonly reported: hypoglycaemia in combination with insulin or a sulphonylurea, vulvovaginal candidiasis, urinary tract infection, and polyuria or pollakiuria (that is, urinary frequency). For full details of adverse reactions and contraindications, see the summary of product characteristics.

2.4 According to the manufacturer's submission, the drug’s price is £39.20 for canagliflozin 100 mg (30 tablets) and £49.99 for canagliflozin 300 mg (30 tablets). The expected annual cost of canagliflozin is £477.26 for the 100 mg daily dosage and £608.83 for the 300 mg daily dosage. Costs may vary in different settings because of negotiated procurement discounts.

3 The manufacturer's submission

The Appraisal Committee (section 8) considered evidence submitted by the manufacturer of canagliflozin and a review of this submission by the Evidence Review Group (ERG; section 9).

Clinical effectiveness

3.1 The manufacturer's literature search identified 85 citations for use in the systematic literature review. It identified 11 clinical trials that evaluated canagliflozin in combination therapy for treating type 2 diabetes. The manufacturer indicated that 5 of these trials would not provide useful information for approaching the decision problem. No non-randomised clinical trials were included in the manufacturer’s submission.

3.2 Of the 6 randomised controlled trials in the manufacturer’s submission, 2 evaluated canagliflozin as part of dual therapy in combination with metformin (DIA3006 and DIA3009), 3 as part of triple therapy in combination with metformin and either a sulfonylurea or pioglitazone (DIA3012, DIA3002, DIA3015) and 1 with insulin with or without other antidiabetic treatments (DIA3008 insulin sub-study). All studies except DIA3015 evaluated 2 doses of canagliflozin (100 mg and 300 mg). DIA3015 used the higher dose only.

  • DIA3009 compared canagliflozin 100 mg (n=483) and canagliflozin 300 mg (n=485) with glimepiride (n=484) as part of dual therapy in combination with metformin for 104 weeks (52-week core double blind and 52-week extension double blind).
  • DIA3006 compared canagliflozin 100 mg (n=368) and canagliflozin 300 mg (n=367) with sitagliptin 100 mg (n=366) and with placebo/sitagliptin (n=183) as part of dual therapy in combination with metformin for 52 weeks (26-week core double blind and 26-week extension double blind).
  • DIA3015 compared canagliflozin 300 mg (n=378) with sitagliptin 100 mg (n=378) as part of triple therapy in combination with metformin and a sulfonylurea for 52 weeks.
  • DIA3002 compared canagliflozin 100 mg (n=157) and canagliflozin 300 mg (n=156) with placebo (n=156) as part of triple therapy in combination with metformin and a sulfonylurea for 52 weeks (26-week core double blind and 26-week extension double blind).
  • DIA3012 compared canagliflozin 100 mg (n=115) and canagliflozin 300 mg (n=114) with placebo (n=115; crossover to sitagliptin at 26 weeks) as part of triple therapy in combination with metformin and pioglitazone for 52 weeks (26-week core double blind and 26-week extension double blind).
  • The DIA3008 insulin sub-study compared canagliflozin 100 mg (n=566) and canagliflozin 300 mg (n=587) with placebo (n=565) as an add-on treatment to insulin with or without other antidiabetic drugs for 18 weeks. It was part of the ongoing CANVAS safety study in 4330 patients with high risk or history of cardiovascular disease, which will report in 2017.

3.3 Patients were eligible for these trials if they had type 2 diabetes and inadequate glycaemic control on existing treatment (1 or 2 background therapies for all studies except the DIA3008 insulin sub-study, for which the maximum number of add-on treatments was not stated). Inadequate glycaemic control was defined as an HbA1c level of 7.0–10.5%, except for DIA3009 in which the range was 7.0–9.5%. All patients enrolled into CANVAS had a history of, or were at high risk of, cardiovascular disease; those enrolled in the DIA3008 insulin sub-study were on insulin alone or in combination with standard of care. The trials enrolled 12–28% patients in Europe, of whom 27 were in the UK.

3.4 The primary outcome for all studies was change in HbA1c from baseline to the end of the double-blind treatment period. Secondary outcomes included change in body weight, change in systolic blood pressure, and incidence of hypoglycaemia. Results for all trials reported in the manufacturer’s submission were for the modified intention-to-treat populations (defined as randomised patients who received at least 1 dose of study drug using a last observation carried forward approach).

Manufacturer's results for dual therapy

3.5 In DIA3009, mean change in HbA1c [minus glimepiride] at week 52 was −0.01% (95% confidence interval [CI] −0.109 to 0.085) for canagliflozin 100 mg plus metformin compared with glimepiride and metformin. At week 52, canagliflozin 300 mg and metformin produced a statistically superior mean reduction in HbA1c compared with glimepiride plus metformin, with a mean change (minus glimepiride) of −0.12% (95% CI −0.217 to −0.023, p<0.001). In DIA3006, mean change in HbA1c (minus placebo) was −0.627% (95% CI −0.758 to −0.481) for canagliflozin 100 mg plus metformin and −0.77% for canagliflozin 300 mg plus metformin (95% CI −0.914 to −0.636), compared with −0.66% (95% CI −0.795 to −0.516) for sitagliptin plus metformin (p<0.001 versus placebo for the canagliflozin arms).

3.6 In DIA3009, there was a greater improvement in systolic blood pressure at 52 weeks with both doses of canagliflozin compared with glimepiride (mean difference in systolic blood pressure reduction [minus glimepiride] −3.5 mmHg [95% CI −4.9 to −2.1] with canagliflozin 100 mg and −4.8 mmHg [95% CI −6.2 to −3.40 with canagliflozin 300 mg). In DIA3006, canagliflozin 100 mg and 300 mg decreased systolic blood pressure from baseline at 26 weeks with a difference in mean systolic blood pressure (minus placebo) of −5.36 mmHg (95% CI −7.280 to −3.439) and −6.58 mmHg (95% CI −8.504 to −4.653) respectively compared with −3.54 mmHg (95% CI −5.273 to −1.413) in the sitagliptin arm (p<0.001 for both canagliflozin doses compared with placebo).

3.7 In DIA3009 at week 52, both doses of canagliflozin were associated with a statistically greater change in body weight compared with glimepiride. Weight loss was −4.2 kg (standard error 0.2) for canagliflozin 100 mg and −4.7 kg (standard error 0.2) for canagliflozin 300 mg compared with a slight increase in weight with glimepiride of +1.0 kg (standard error 0.2). In DIA3006 at week 26, a weight loss of −3.7 kg (standard error 0.2) was observed with canagliflozin 100 mg and −4.2 kg (standard error: 0.2) with canagliflozin 300 mg compared with −1.2 kg (standard error 0.2) with sitagliptin. Both doses of canagliflozin demonstrated statistical superiority to sitagliptin up to week 52 with a difference in mean weight reduction (minus sitagliptin) of −2.1 kg for canagliflozin 100 mg and −2.5 kg for canagliflozin 300 mg (p<0.001 for both doses).

Manufacturer's results for triple therapy

3.8 In DIA3015 at week 52, canagliflozin 300 mg produced a statistically superior reduction in HbA1c compared with sitagliptin, with a difference in mean change in HbA1c for canagliflozin 300 mg (minus sitagliptin) of −0.37% (95% CI −0.50 to −0.25). DIA3002 and DIA3012 showed a statistically significant reduction in HbA1c at 26 weeks for both doses of canagliflozin compared with placebo. Mean reduction in HbA1c at week 26 with canagliflozin 100 mg (minus placebo) was −0.71% (95% CI −0.904 to −0.524, p<0.001) in DIA3002 and −0.62% (95% CI −0.811 to −0.437, p<0.001) in DIA3012. Mean reduction with canagliflozin 300 mg was −0.92% (95% CI −1.114 to −0.732, p<0.001) in DIA3012 and −0.76% (95% CI −0.951 to −0.575, p<0.001) in DIA3002.

3.9 Both doses of canagliflozin, when used in triple therapy, resulted in a statistically significant greater reduction in systolic blood pressure compared with sitagliptin or placebo in DIA3015 and DIA3012 but not DIA3002. In DIA3015, canagliflozin 300 mg statistically significantly decreased systolic blood pressure compared with sitagliptin with a difference in mean change (minus sitagliptin) in systolic blood pressure at week 52 of −5.9 mmHg (95% CI −7.642 to −4.175, p<0.001). Similarly, a statistically significant difference was seen in DIA3012 at 26 weeks, with a mean difference in systolic blood pressure (minus placebo) of −4.07 (95% CI −6.879 to −1.251, p=0.005) with canagliflozin 100 mg and −3.46 (95% CI −6.281 to −0.643, p=0.016) with canagliflozin 300 mg. No statistically significant difference in mean systolic blood pressure (minus placebo) was seen at 26 weeks in DIA3002 with canagliflozin 100 mg (−2.24 [95% CI −4.719 to 0.241, p=0.077]) or canagliflozin 300 mg (−1.62 [95% CI −4.111 to 0.866, p=0.201]).

3.10 Both doses of canagliflozin, when used in triple therapy, lowered body weight. In DIA3015 at week 52, canagliflozin 300 mg treatment resulted in statistically significant reductions in body weight relative to sitagliptin (−2.8 kg [95% CI −3.3 to −2], p<0.001). In DIA3002 at week 26, canagliflozin 100 mg and 300 mg (in combination with metformin and a sulfonylurea) resulted in a reduction in weight of −2.1 kg (standard error 0.3) and −2.6 kg (standard error 0.3) respectively compared with a weight change of −0.7 kg (standard error 0.3) in the placebo group. Changes in body weight were also evident at week 52 with a difference in mean weight change (minus placebo) of −1.0 kg (95% CI −1.8 to −1.2) for canagliflozin 100 mg and −2.1 kg (95% CI −2.9 to −1.2) for canagliflozin 300 mg. In DIA3012 at week 26, a change in weight of −2.7 kg (standard error 0.3) and −3.8 kg (standard error 0.3) was observed for canagliflozin 100 mg and 300 mg respectively (in combination with metformin and a thiazolidinedione).

Manufacturer's results for add-on treatment to insulin

3.11 At week 18 in the DIA3008 insulin sub-study, the difference in mean change in HbA1c versus placebo was −0.65% (95% CI −0.73 to −0.56) in the canagliflozin 100 mg group and −0.73% (95% CI −0.82 to −0.65) in canagliflozin 300 mg arm (p<0.001 both comparisons). Both doses of canagliflozin resulted in a significant reduction in systolic blood pressure (p<0.001) compared with placebo. The difference in mean systolic blood pressure change (minus placebo) at week 18 was −2.58 mmHg (95% CI −4.060 to 1.091; p<0.001) with canagliflozin 100 mg and −4.38 mmHg (95% CI −5.850 to −2.903; p<0.001) with canagliflozin 300 mg. Change in body weight (minus placebo) at week 18 was −1.8 kg (95% CI −2.2 to −1.6) in the canagliflozin 100 mg group and −2.3 kg (95% CI −2.7 to −2.1) in the canagliflozin 300 mg group (p<0.001 for both comparisons).

Manufacturer's meta-analysis

3.12 The manufacturer conducted a systematic literature review to identify randomised controlled trials that evaluated treatments relevant to the NICE scope for this appraisal in patients with type 2 diabetes. The manufacturer’s base-case network meta-analyses included 38 studies comparing treatments given in combination with metformin (metformin background), 10 studies comparing treatments given in combination with metformin and a sulfonylurea (metformin and sulfonylurea background), 2 studies comparing treatments given in combination with metformin and pioglitazone (metformin and pioglitazone background) and 14 studies comparing treatments given in combination with insulin (insulin background). The manufacturer used a Bayesian hierarchical model for the network meta-analyses. After assessing relative goodness of fit of fixed effects and random effects models using the deviance information criterion, the model associated with the lowest score was selected (with a difference of at least 3 points). All analyses were conducted by background therapy. The manufacturer conducted sensitivity analyses to determine the robustness of results. The manufacturer’s submission reported outcomes that were relevant to its economic model including change in HbA1c, weight, BMI, systolic blood pressure and incidence of hypoglycaemia.

Dual therapy with a metformin background

3.13 The manufacturer presented differences in HbA1c change at 52 weeks for canagliflozin 100 mg and 300 mg compared with the different comparators as dual therapy with a metformin background. Canagliflozin 100 mg produced a greater reduction in HbA1c than sitagliptin 100 mg (−0.01% [−0.48 to 0.44]) and dapagliflozin 10 mg (−0.14% [−0.81 to 0.47]), but not liraglutide 1.2 mg (0.40% [−0.33 to 1.11]), canagliflozin 300 mg (0.13% [−0.25 to 0.52]), pioglitazone 30 mg (0.11% [−0.44 to 0.84]), exenatide 10 micrograms (0.02% [−0.65 to 0.55]) or glimepiride (0.00% [−0.45 to 0.46]). Canagliflozin 300 mg was associated with a greater reduction in HbA1c than pioglitazone 30 mg (−0.02% [−0.57 to 0.72]), exenatide 10 micrograms (−0.11% [−0.78 to 0.42]), glimepiride (−0.13% [−0.58 to 0.33]), canagliflozin 100 mg (−0.13% [−0.52 to 0.25]), sitagliptin 100 mg (−0.14% [−0.61 to 0.31]) and dapagliflozin 10 mg (−0.27% [−0.94 to 0.34]). However, it was not associated with a greater reduction in HbA1c compared with liraglutide 1.2 mg (0.27% [−0.46 to 0.98]).

3.14 When given as part of dual therapy with a metformin background, canagliflozin 100 mg and 300 mg were associated with greater weight reductions at 52 weeks compared with a DPP‑4 inhibitor (sitagliptin 100 mg; differences of −2.12 kg and −2.48 kg respectively), a sulfonylurea (glimepiride; differences of −3.97 kg and −4.33 kg respectively) and pioglitazone 30 mg (differences of −4.57 kg and −4.93 kg respectively). The reduction in weight was at least similar for canagliflozin 100 mg and 300 mg compared with dapagliflozin 10 mg (differences of −0.11 kg and −0.48 kg respectively). When comparing canagliflozin with the GLP‑1 analogues, canagliflozin 100 mg and 300 mg gave a lesser weight reduction than exenatide 10 micrograms (differences of 1.47 kg and 1.11 kg respectively) but a greater weight reduction than liraglutide 1.2 mg (−0.49 and −0.85 kg respectively).

3.15 The manufacturer reported that canagliflozin 100 mg and 300 mg were associated with higher reductions in systolic blood pressure compared with glimepiride (differences of −3.52 mmHg and −4.73 mmHg respectively), liraglutide 1.2 mg (differences of −3.50 mmHg and −4.71 mmHg respectively) and sitagliptin 100 mg (differences of −2.84 mmHg and −4.04 mmHg respectively), and smaller reductions in systolic blood pressure than pioglitazone 30 mg (differences of 2.05 mmHg and 0.83 mmHg respectively).

3.16 The manufacturer reported that canagliflozin 100 mg and 300 mg were associated with a lower risk of hypoglycaemia compared with a sulfonylurea (odds ratios ranging from 0.10 to 0.11) and a higher risk of hypoglycaemia compared with dapagliflozin 10 mg (odds ratios ranging from 3.43 to 3.65) and a DPP‑4 inhibitor (odds ratios ranging from 1.19 to 3.62). The manufacturer advised that the difference in risk of hypoglycaemia compared with dapagliflozin in this network meta-analysis was likely to be because of differences in how hypoglycaemic events were reported in the clinical trials, but did not provide any further explanation for this.

Triple therapy with a metformin plus sulfonylurea background

3.17 Comparisons for canagliflozin and comparators with a metformin and sulfonylurea background were provided by the manufacturer. Compared with a DPP‑4 inhibitor (sitagliptin 100 mg), canagliflozin 100 mg produced a similar reduction in HbA1c (0.07%), whereas canagliflozin 300 mg was associated with a slightly greater HbA1c reduction (−0.17%). Canagliflozin 100 mg gave a similar HbA1c reduction (difference of 0.03%) compared with a GLP‑1 analogue (exenatide 10 micrograms), and canagliflozin 300 mg gave a higher HbA1c reduction (difference of −0.21%). Canagliflozin 100 mg and 300 mg were associated with greater weight reductions than a DPP‑4 inhibitor (difference of −2.03 kg and −2.64 kg respectively) and similar weight reductions to a GLP‑1 analogue (difference of −0.14 kg and 0.47 kg respectively). Canagliflozin 100 mg and 300 mg were associated with a higher reduction in systolic blood pressure than sitagliptin 100 mg (differences of −5.76 mmHg and −5.16 mmHg respectively). Canagliflozin 100 mg and 300 mg were associated with a similar risk of hypoglycaemia as sitagliptin 100 mg (odds ratios of 0.75 and 0.96 respectively) and exenatide 10 micrograms (odds ratios of 0.96 and 1.23 respectively).

Triple therapy with a metformin plus thiazolidinedione background

3.18 The manufacturer's submission presented comparisons for canagliflozin and comparators with a metformin and thiazolidinedione background. Compared with sitagliptin 100 mg, canagliflozin 100 mg was associated with a smaller reduction in HbA1c at 26 weeks (difference of 0.07%) and canagliflozin 300 mg was associated with a greater reduction (difference of −0.07%). Canagliflozin 100 mg and 300 mg were associated with weight reductions compared with sitagliptin 100 mg (differences of −2.70 kg and −3.65 kg respectively). No statistical analysis was conducted for changes in systolic blood pressure because of limitations in the evidence base. Canagliflozin 100 mg was associated with a similar risk of hypoglycaemic events as sitagliptin 100 mg (odds ratio of 0.86), whereas canagliflozin 300 mg was associated with a higher risk (odds ratio of 1.89).

Add-on to insulin with or without an antihyperglycaemic background

3.19 When given as an add-on treatment to insulin, the reduction in HbA1c at 26 weeks with canagliflozin 100 mg and 300 mg was greater than with sitagliptin 100 mg (differences of −0.01% and −0.15% respectively) and dapagliflozin 10 mg (differences of −0.01% and −0.15% respectively). The higher dose of canagliflozin gave a greater HbA1c reduction than exenatide 10 micrograms (difference of −0.05%) but the lower dose did not (difference of 0.09%). A similar pattern was observed for the comparison of canagliflozin and pioglitazone 30 mg (difference of −0.10% for canagliflozin 300 mg and 0.04% for canagliflozin 100 mg). Pioglitazone 45 mg produced a greater HbA1c reduction than either canagliflozin 100 mg or 300 mg.

3.20 When given as an add-on treatment to insulin, canagliflozin 100 mg and 300 mg were associated with greater weight reduction than a DPP‑4 inhibitor (differences of −2.10 kg and −2.67 kg respectively), pioglitazone 30 mg (differences of −3.28 kg and −3.85 kg respectively) and dapagliflozin 10 mg (differences of −0.41 kg and −0.98 kg respectively) but not exenatide 10 micrograms (differences of 0.64 kg and 0.07 kg respectively). Canagliflozin 100 mg and 300 mg were associated with higher reductions in systolic blood pressure compared with dapagliflozin 10 mg (differences of −0.92 mmHg and −2.62 mmHg respectively). When comparing canagliflozin and exenatide 10 micrograms, canagliflozin 300 mg was associated with a lower reduction in systolic blood pressure (difference of −1.33 mmHg) but canagliflozin 100 mg was not (0.38 mmHg). No analysis of BMI results or all hypoglycaemic events was conducted because of lack of data at 26 weeks for the canagliflozin trial.

Adverse events

3.21 The canagliflozin clinical trials included in the submission evaluated the safety of canagliflozin in 10,285 people with type 2 diabetes. The manufacturer provided data from 3 pre-specified pooled safety datasets, focusing on a broad dataset that included data for canagliflozin 100 mg (n=3092), canagliflozin 300 mg (n=3085) and all non-canagliflozin arms (placebo, glimepiride and sitagliptin [n=3262]).

3.22 In the broad dataset, the incidence of any adverse event was similar across groups (76.6% for canagliflozin 100 mg, 77.0% for canagliflozin 300 mg and 75.8% in the non-canagliflozin group). The incidence of adverse events leading to discontinuation in the broad dataset was higher in the canagliflozin 300 mg group (7.3%) than the canagliflozin 100 mg (5.6%) and non-canagliflozin (5.0%) groups. Adverse events that led to discontinuation of more than 0.2% of patients in the canagliflozin 300 mg group were decreased glomerular filtration rate, renal impairment and increased blood creatinine. The incidence of adverse events considered related to the study drug by the investigator was slightly higher in the canagliflozin 100 mg and 300 mg groups (33.6% and 29.4% respectively) than the non-canagliflozin group (21.8%). The incidence of serious adverse events and deaths was similar in the canagliflozin and non-canagliflozin groups. Adverse events occurring in at least 5% of subjects in a canagliflozin group were nasopharyngitis, hypoglycaemia, upper respiratory tract infection, urinary tract infection, diarrhoea, arthralgia, back pain and headache.

3.23 The manufacturer noted that sodium–glucose cotransporter-2 (SGLT-2) inhibitors are associated with a higher incidence of genital mycotic infections because of urinary glucose excretion. In the broad dataset, the incidence of genital mycotic infection adverse events in women was higher in those receiving canagliflozin 100 mg (14.7%) and canagliflozin 300 mg (13.9%) than in those taking placebo (3.1%). In men, the incidence was 7.3% in the canagliflozin 100 mg group and 9.3% in the canagliflozin 300 mg groups, compared with 1.6% of men in the non-canagliflozin group.

ERG's comments on the manufacturer's clinical-effectiveness evidence

3.24 The ERG considered that only the trials with active comparator arms were relevant to the NICE scope (DIA3009 and DIA3006 [dual therapy] and DIA3015 [triple therapy]) and judged all 3 to be of generally good methodological quality. The ERG considered the manufacturer’s submission to provide a generally unbiased estimate of canagliflozin’s treatment effect within the stated scope of the decision problem, and that the manufacturer’s interpretation of the evidence was largely appropriate and justified.

3.25 However, the ERG identified some exceptions and uncertainties:

  • A lack of direct evidence for comparisons with treatments other than sitagliptin and glimepiride.
  • No discussion ofthe implications of differences in results between the modified intention-to-treat analyses (reported in the manufacturer’s submission) and per protocol analyses (reported in the trial journal publications and clinical study reports). The ERG advised that the per protocol analyses did not show superiority for canagliflozin 300 mg in the 2 dual therapy trials (unlike the modified intention-to-treat analyses).
  • A low volume of direct evidence for some of the loops in the network meta-analyses, which led the ERG to conclude that the results should be interpreted with caution.
  • It was not convinced by the manufacturer's justification for network meta-analysis for triple therapy assessing effects at 26 weeks, rather than at 52 weeks.
  • The exclusion of background treatments that did not include metformin (because including these would better reflect all the potential uses of canagliflozin within its European marketing authorisation).
  • Lack of evidence for the longer-term efficacy and safety of canagliflozin (because most outcomes were measured up to 52 weeks in the clinical trials).
  • It did not have confidence in the manufacturer’s favourable interpretation of adverse events related to canagliflozin compared with other treatments, and the discontinuation rates compared with sitagliptin and glimepiride.

Cost effectiveness

3.26 The manufacturer’s submission included de novo economic analyses of canagliflozin in combination therapy for type 2 diabetes using the following regimens:

  • dual therapy in combination with metformin
  • triple therapy in combination with metformin and a sulfonylurea
  • triple therapy in combination with metformin and a thiazolidinedione
  • add-on treatment to insulin (with or without other antihyperglycaemic agents).

3.27 The manufacturer advised that these populations reflected the licensed indications for canagliflozin combination treatment and patients in the clinical trials, and it considered these patients to be representative of the population most likely to receive canagliflozin in clinical practice in England. The 100 mg and 300 mg doses of canagliflozin were considered separately in the base-case analyses (increasing the dose from 100 mg to 300 mg according to the summary of product characteristics [see section 2.2] was explored in a scenario analysis). Comparator treatments used at the different points in the treatment pathway were sulfonylureas, thiazolidinediones, DPP‑4 inhibitors, GLP‑1 analogues, dapagliflozin and insulin. The manufacturer selected a comparator to represent each treatment class in its economic model (based on frequency of use in clinical practice, comparators in the canagliflozin clinical trials and available data).

3.28 The manufacturer's systematic literature review of potentially relevant published cost-effectiveness analyses identified 52 analyses, and a similar systematic literature review found 21 economic evaluation models of type 2 diabetes. The manufacturer chose the ECHO-T2DM model, which was a stochastic micro-simulation model in which cohorts of individual hypothetical patients were created and simulated over time using Monte Carlo (first-order uncertainty) techniques. Second-order (parameter) uncertainty was captured by hypothetical cohorts of 1000 patients defined by baseline characteristics sourced from the canagliflozin clinical trials including demographics (for example, age and sex), biomarker values (for example, HbA1c, systolic blood pressure and BMI, which are short-term outcomes used to predict the likelihood of longer-term events in the model) and disease indicators (for example, disease duration and history of complications). Individual patient outcomes were simulated over time through health states capturing micro- and macrovascular complications and death. There were 1000 patient cohorts for each model run for the base-case comparisons, and patient cohorts were simulated until death or the end of the designated follow-up period. Results of key parameter values (including treatment effects, risk equation coefficients, and quality-adjusted life year [QALY] disutility weights) were then aggregated. The model used a lifetime time horizon (40 years), the cycle length was 1 year, health benefits and costs were each discounted at 3.5% and the analysis was from an NHS perspective.

3.29 The manufacturer's model included 3 parallel sets of microvascular complications (to reflect increasing severity of retinopathy, chronic kidney disease and neuropathy) and 4 types of macrovascular complications (ischaemic heart disease, myocardial infarction, stroke, and congestive heart failure; because of interdependence with neuropathic outcomes, peripheral vascular disease was classified as a microvascular neuropathy complication). These were represented by Markov health states and were associated with costs, utility values and, in some cases, a possible treatment contraindication (for example, end-stage renal disease for pioglitazone) or excess risk of death (for example, myocardial infarction or stroke). Annual probabilities of experiencing worsening microvascular complications were derived primarily from the Wisconsin Epidemiologic Study of Diabetic Retinopathy, the Rochester Epidemiology Project, and the Centers for Disease Control model of chronic kidney disease. Risk equations from the original UKPDS Outcomes Model were used to simulate macrovascular complications in the base case. The manufacturer mainly used published utility values that had been elicited from CODE-2, which was a non-interventional, observational study. Utility decrements were applied to the baseline quality of life value (estimated at 1.027 using multivariate regression techniques) for patient characteristics (for example, age and duration of disease), microvascular and macrovascular complications, hypoglycaemic events, obesity and adverse events.

3.30 The manufacturer explained that diabetes treatments have an initial effect followed by annual 'drift', where the effect lessens over time. Biomarker values after the first cycle were estimated using annual drift values for each treatment. Based on published values, HbA1c drift was assumed to be 0.14% for canagliflozin, dapagliflozin, DPP‑4 inhibitors and GLP‑1 analogues, 0.07% for thiazolidinediones, 0.24% for sulfonylureas and 0.15% for insulin. The drift values for systolic blood pressure, lipids, weight and eGFR were assumed to be the same for all treatments. The manufacturer’s model allowed patients to take anti-dyslipidaemia and anti-hypertension medications, and applied rescue treatments when biomarker thresholds for dyslipidaemia and hypertension were exceeded.

Manufacturer's cost-effectiveness results

3.31 The manufacturer presented pairwise comparisons using cohorts of simulated patients from the probability distribution of patient characteristics for canagliflozin 100 mg and canagliflozin 300 mg in dual therapy, triple therapy and as an add-on to insulin. At the clarification stage, the manufacturer provided some updated cost-effectiveness results after identifying that, where BMI was not available in the network meta-analysis, the change in actual BMI had been used instead of being calculated using a weighted average of height in the UK general population. The updated incremental cost-effectiveness ratios (ICERs) were generally more favourable for canagliflozin than the ones they replaced.

Manufacturer’s base case – dual therapy

3.32 Canagliflozin 100 mg as part of a dual-therapy regimen showed QALY gains and increased costs compared with a sulfonylurea (ICER of £1537 per QALY gained [incremental costs £288; incremental QALYs 0.188]), a DPP‑4 inhibitor (ICER of £97 per QALY gained [incremental costs £1; incremental QALYs 0.013]), and dapagliflozin (£8674 per QALY gained [incremental costs £63; incremental QALYs 0.007]). The ICER provided by the manufacturer at the clarification stage for the comparison with dapagliflozin was £2993 per QALY gained (incremental costs and QALYs not provided). Compared with a GLP‑1 analogue, canagliflozin 100 mg was less effective and less costly (incremental costs −£2424; incremental QALYs −0.048). Canagliflozin 100 mg continued to be less effective and less costly than a GLP‑1 analogue after correcting the BMI data (incremental costs and QALYs not provided). Canagliflozin 100 mg was dominated by pioglitazone (that is, canagliflozin was more costly and less effective [incremental costs £2833; incremental QALYs −0.159]).

3.33 Canagliflozin 300 mg as part of a dual-therapy regimen, when compared with a sulfonylurea, produced higher costs and greater QALY gains (ICER of £4899 per QALY gained [incremental costs £976; incremental QALYs 0.199]), a DPP‑4 inhibitor (ICER of £18,349 per QALY gained [incremental costs £576; incremental QALYs 0.031]), and dapagliflozin (ICER of £27,419 per QALY gained [incremental costs £625; incremental QALYs 0.023]. The ICER provided at the clarification stage for the comparison with dapagliflozin was £21,626 per QALY gained (incremental costs and QALYs not provided). Similar to the 100 mg dose, canagliflozin 300 mg was less costly and less effective than a GLP‑1 analogue, giving an ICER of £76,214 per QALY gained for a GLP-1 analogue to replace canagliflozin 300 mg (incremental costs −£1892; incremental QALYs −0.025). Canagliflozin 300 mg continued to be less effective and less costly than a GLP‑1 analogue after correcting the BMI data (incremental costs and QALYs not provided). Like the 100 mg dose, canagliflozin 300 mg was also dominated by pioglitazone (incremental costs £3353; incremental QALYs −0.141).

Manufacturer’s base case – triple therapy

3.34 The manufacturer also presented cost-effectiveness results for triple-therapy regimens containing canagliflozin 100 mg and 300 mg. In a triple-therapy combination with metformin and a sulfonylurea, canagliflozin 100 mg dominated a DPP‑4 inhibitor (incremental costs −£42; incremental QALYs 0.016), and also dominated a GLP‑1 analogue (incremental costs −£1297; incremental QALYs 0.001). Adjusting the change in BMI at the clarification stage caused canagliflozin to become less costly and less effective than a GLP‑1 analogue, giving an updated ICER of £265,928 per QALY gained for a GLP‑1 analogue to replace canagliflozin 100 mg. Canagliflozin 100 mg was associated with greater costs and QALYs than long-acting insulin, with an ICER of £263 per QALY gained (incremental costs £135; incremental QALYs 0.514). An updated ICER provided at the clarification stage was £183 per QALY gained. In a triple-therapy combination with metformin and a thiazolidinedione, canagliflozin 100 mg had higher costs and QALY gains than a DPP‑4 inhibitor, producing an ICER of £1095 per QALY gained (incremental costs £7; incremental QALYs 0.007).

3.35 In combination with metformin and a sulfonylurea, canagliflozin 300 mg compared with a DPP‑4 inhibitor had an ICER of £13,287 per QALY gained (incremental costs £461; incremental QALYs 0.035) and dominated a GLP‑1 analogue (incremental costs −£685; incremental QALYs 0.004). Canagliflozin 300 mg continued to dominate a GLP‑1 analogue after updating the BMI data at the clarification stage. In combination with metformin and a thiazolidinedione, the ICER for canagliflozin 300 mg compared with a DPP‑4 inhibitor was £21,430 per QALY gained (incremental costs £691; incremental QALYs 0.032). Canagliflozin 300 mg compared with long-acting insulin gave an ICER of £607 per QALY gained (incremental costs £379; incremental QALYs 0.624). After updating the BMI data at the clarification stage, the ICER increased slightly to £671 per QALY gained.

Manufacturer’s base case – add-on to insulin

3.36 Canagliflozin 100 mg as an add-on treatment to insulin dominated dapagliflozin (incremental costs −£72; incremental QALYs 0.003). Canagliflozin 100 mg was associated with lower costs and lower QALY gains compared with a DPP‑4 inhibitor (incremental costs −£13; incremental QALYs −0.010), with an ICER of £1340 per QALY gained for canagliflozin 100 mg to be replaced by a DPP-4 inhibitor. Compared with a GLP‑1 analogue, canagliflozin 100 mg was also associated with lower costs and lower QALY gains (incremental costs −£836; incremental QALYs −0.065) with an ICER of £12,915 per QALY gained for canagliflozin 100 mg to be replaced by a GLP-1 analogue.

3.37 Canagliflozin 300 mg as an add-on treatment to insulin produced greater costs and QALY gains compared with a DPP‑4 inhibitor (ICER of £7975 per QALY gained [incremental costs £322; incremental QALYs 0.040]), and dapagliflozin (ICER of £5992 per QALY gained [incremental costs £327; incremental QALYs 0.055]). Compared with a GLP‑1 analogue, canagliflozin 300 mg was associated with lower costs and lower QALY gains (incremental costs −£526; incremental QALYs −0.015), with an ICER of £35,575 per QALY gained for canagliflozin to be replaced by a GLP-1 analogue.

Manufacturer’s sensitivity analyses

3.38 The manufacturer conducted deterministic and probabilistic sensitivity analyses for canagliflozin compared with a DPP‑4 inhibitor (in dual therapy, triple therapy and as an add-on to insulin therapy) and with a sulfonylurea in dual therapy, because it perceived these to be the key comparators for canagliflozin. The manufacturer did not report results for other comparators used in its base case, including dapagliflozin.

3.39 The manufacturer’s deterministic sensitivity analyses showed that the results from the model were most sensitive to varying the metabolic drift of HbA1c associated with canagliflozin and its comparators. The manufacturer commented that, except for HbA1c drift, the ICERs for the comparisons of both doses of canagliflozin with a sulfonylurea in dual therapy were largely insensitive to parameter changes. However, the manufacturer noted the ICERs for canagliflozin compared with a DPP‑4 inhibitor were generally less stable.

3.40 The manufacturer presented results of probabilistic sensitivity analyses for canagliflozin compared with some of the comparators used in its base case and commented that there was greater uncertainty where the incremental changes in costs and QALYs were small. It noted that, at a maximum acceptable ICER of £20,000 per QALY gained, the probability of canagliflozin being cost effective compared with a DPP‑4 inhibitor for the different comparisons was 45–56% for the 100 mg dose and 52–61% for the 300 mg dose. The manufacturer stated that there was less uncertainty in the dual therapy comparisons of canagliflozin (100 mg and 300 mg) with a sulfonylurea, with the probability of cost effectiveness being around 90% for both doses (at maximum acceptable ICERs of £20,000 or £30,000 per QALY gained).

3.41 The manufacturer undertook scenario analyses that explored dose escalation from the recommended starting dose of 100 mg to 300 mg (see section 2.1). Modelling techniques were used because this had not been studied in clinical trials. If patients in the simulated cohort had tolerated the 100 mg dose of canagliflozin but had not reached an HbA1c of less than 7.5% at 6 months, then the canagliflozin dose was increased to 300 mg in cycle 2. The manufacturer based this assumption on clinical specialist opinion. Switching to the higher dose was only permitted at the end of the first cycle: patients who tolerated treatment and had satisfactory glycaemic control continued the 100 mg dose and switched to standard rescue therapy when needed. It was assumed that patients who switched to the higher dose experienced the same treatment effects as patients treated with 300 mg for the full 12 months. The manufacturer stated that the dose escalation scenario improved the cost effectiveness of canagliflozin for 11 out of 12 comparisons made. Only the ICER comparing canagliflozin with a sulfonylurea in dual therapy increased (from £1537 per QALY gained to £1721 per QALY gained) and canagliflozin was dominant in 7 of the scenarios.

ERG's comments on the manufacturer’s cost-effectiveness analyses

3.42 The ERG concluded that the methods and inputs in the manufacturer’s economic evaluation were generally in line with the NICE reference case, but noted:

  • Not all comparators in the NICE scope had been included in the manufacturer’s decision problem.
  • Comparators used were not always the most widely prescribed.
  • The NICE reference case states that data from head-to-head trials should be presented in the reference-case analysis if possible, but the manufacturer had instead sometimes used results from the meta-analyses. The ERG indicated that although the manufacturer had stated this was for consistency, this was not properly justified because the manufacturer had not provided a fully incremental cost-effectiveness analysis (that is, only pairwise comparisons had been generated).
  • It was not clear if the preference data for utility values wholly represented the population of England because they were derived from a European study (CODE-2).

The ERG considered the model to be internally consistent and very well validated.

3.43 Overall, the ERG considered the clinical-effectiveness data in the model to be appropriate (although sometimes network meta-analysis estimates were used instead of head-to-head trial data). It found the equations used for the extrapolation of biomarker outcomes to be well-established and appropriate for the patient population in England, and the drift assumptions for the biomarkers to be reasonable. However, the ERG was concerned that using only 1000 patients per cohort in the base case might not robustly capture the ICERs.

3.44 The ERG commented that health benefits had been measured and evaluated in line with the NICE reference case and that utility values had been appropriately incorporated into the manufacturer’s model. The ERG generally agreed with the costs used by the manufacturer for drugs, adverse events and health states (complications and comorbidities), although there was some uncertainty in the costs used for insulin.

3.45 Although the ERG agreed with the manufacturer’s conclusions from the results of the deterministic analyses as presented, the ERG considered that the uncertainty in the decision problem had not been fully explored, and that the manufacturer did not present the results in the most informative way. The ERG was unclear why second order uncertainty was not switched off in the deterministic sensitivity analyses (that is, by specifying only 1 patient cohort). Consequently, the ERG believed that the manufacturer’s results were partly confounded by stochasticity in other parameters (that is, they included uncertainty from more than 1 source and did not truly reflect the uncertainty associated with varying 1 parameter).

3.46 The ERG reviewed the manufacturer’s scenario analyses, focusing on the dose escalation scenario. It noted that, like the deterministic sensitivity analyses, second-order uncertainty was not switched off in the scenario analyses, meaning that the results were again partly confounded by parameter stochasticity. The ERG noted that the manufacturer’s submission concluded from the dose escalation scenario analysis that the dose escalation schedule from 100 mg to 300 mg was cost effective, but the ERG did not consider that this conclusion applied to all treatment comparisons, noting that:

  • Canagliflozin 100 mg compared with a thiazolidinedione in dual therapy remained dominated in the scenario analyses as well as in the base case.
  • Canagliflozin 100 mg as an add-on to insulin was cheaper and less effective than a DPP‑4 inhibitor in the base case, but associated with an ICER of £503 per QALY gained in the scenario analysis.
  • Canagliflozin 100 mg as an add-on to insulin was cheaper and less effective than a GLP‑1 analogue in the base case and remained so in the scenario analysis.

3.47 The ERG concluded that although all relevant variables appeared to have been included in the manufacturer’s probabilistic sensitivity analyses, it was unclear if uncertainty in the decision problem had been sufficiently explored because the distributional assumptions were not well described in the manufacturer’s submission and were not transparent in the model. The ERG also commented that standard errors used for the HbA1c treatment effect parameters were too small and that cost-effectiveness acceptability data had not been presented for all base-case comparisons.

ERG’s exploratory analyses

3.48 The ERG did exploratory work to:

  • examine the variation in base-case ICERs by re-running some of the manufacturer’s analyses
  • examine the variation in final ICERs for dual therapy with no parameter uncertainty and 100,000 patients
  • produce incremental analyses for dual therapy
  • calculate the probability that canagliflozin is cost effective compared with dapagliflozin
  • determine the effect of modifying the clinical efficacy of glimepiride and exenatide.

3.49 The ERG found there was minimal variation in the dual-therapy ICERs when it re-ran the manufacturer’s base-case analyses. However, it was unsure if the manufacturer’s use of 1000 cohorts of 1000 patients estimated the ICERs robustly, and how the base-case results might change if parameter uncertainty was removed. Its exploratory analyses with a single cohort of 100,000 patients and no parameter uncertainty had a small impact on the different cost and QALY outcomes compared with the manufacturer’s base-case analyses (typically hundreds of pounds or less, and thousandths of a QALY). The ERG noted, however, that the small differences in incremental QALYs could in some cases drive an apparently large variation in the ICER compared with the manufacturer’s ICERs described in sections 3.32 and 3.33:

  • Canagliflozin 100 mg was dominated by a thiazolidinedione (incremental costs £2929; incremental QALYs −0.166).
  • Canagliflozin 100 mg compared with a sulfonylurea had an ICER of £1579 per QALY gained (incremental costs £274; incremental QALYs 0.174).
  • Canagliflozin 100 mg compared with dapagliflozin had an ICER of £100,719 per QALY gained (incremental costs £193; incremental QALYs 0.002).
  • Canagliflozin 100 mg compared with a DPP‑4 inhibitor had an ICER of £12,938 per QALY gained (incremental costs £211; incremental QALYs 0.016).
  • Canagliflozin 100 mg was less costly and less effective than a GLP‑1 analogue, with an ICER of £67,414 per QALY lost (incremental costs −£2381; incremental QALYs −0.035).
  • Canagliflozin 300 mg was dominated by a thiazolidinedione (incremental costs £3245; incremental QALYs −0.14).
  • Canagliflozin 300 mg compared with a sulfonylurea had an ICER of £5368 per QALY gained (incremental costs £1121; incremental QALYs 0.21).
  • Canagliflozin 300 mg compared with a DPP‑4 inhibitor had an ICER of £9246 per QALY gained (incremental costs £412; incremental QALYs 0.04).
  • Canagliflozin 300 mg compared with dapagliflozin had an ICER of £17,161 per QALY gained (incremental costs £751; incremental QALYs 0.04).
  • Canagliflozin 300 mg was less costly and less effective than a GLP‑1 analogue, with an ICER of £86,412 per QALY lost (incremental costs −£1867; incremental QALYs −0.0216).

3.50 The ERG did a fully incremental exploratory analysis for canagliflozin 100 mg in dual therapy in combination with metformin (excluding thiazolidinediones because their use in clinical practice in England is declining). In the ERG’s exploratory analysis using the manufacturer’s base-case inputs, canagliflozin 100 mg had an ICER of £507 per QALY gained (incremental costs £8; incremental QALYs 0.015) compared with the next best option, which was dapagliflozin. When using the ERG’s preferred inputs (excluding parameter uncertainty and with 100,000 patients), canagliflozin 100 mg had an ICER of £84,800 per QALY gained (incremental costs £245; incremental QALYs 0.003) compared with the next best option, a DPP‑4 inhibitor. The ERG noted that dapagliflozin, canagliflozin 100 mg and a DPP‑4 inhibitor were associated with similar total costs and similar overall total QALYs.

3.51 The ERG did a fully incremental exploratory analysis (excluding thiazolidinediones) for canagliflozin 300 mg in dual therapy in combination with metformin. The ICER for canagliflozin 300 mg compared with the next best option, dapagliflozin, was £17,639 per QALY gained (incremental costs £587; incremental QALYs 0.033) for the manufacturer’s base-case inputs of 1000 cohorts of 1000 patients, and £17,903 per QALY gained (incremental costs £585; incremental QALYs 0.033) compared with the next best option, a DPP‑4 inhibitor, using data from the ERG’s preferred input of a single cohort of 100,000 patients.

3.52 The ERG noted that the manufacturer’s submission did not report the probability of canagliflozin being cost effective compared with dapagliflozin. The ERG’s exploratory analyses for dual therapy with metformin and an SGLT-2 inhibitor showed that the probability of canagliflozin 100 mg being cost effective compared with dapagliflozin was 52.5% at maximum acceptable ICERs of £20,000 and £30,000 per QALY gained. For canagliflozin 300 mg, the probability of cost effectiveness was 46.7% at a maximum acceptable ICER of £20,000 per QALY gained and 50% at £30,000 per QALY gained.

3.53 The ERG was aware that the manufacturer had chosen a single comparator to be representative of each treatment class, but considered that there was variation in specific parameters between drugs within each treatment class that might not be fully captured by the manufacturer’s deterministic sensitivity analyses. The ERG explored greater variation in hypoglycaemia event rate for sulfonylureas and agreed with the manufacturer that this parameter was not very influential. Similarly, the ERG found that using cost and efficacy data for liraglutide instead of exenatide 10 micrograms gave results that were consistent with the manufacturer’s deterministic sensitivity analyses.

3.54 Full details of all the evidence are in the manufacturer’s submission and the ERG report.

4 Consideration of the evidence

4.1 The Appraisal Committee reviewed the data available on the clinical and cost effectiveness of canagliflozin, having considered evidence on the nature of type 2 diabetes and the value placed on the benefits of canagliflozin by people with the condition, those who represent them, and clinical specialists. It also took into account the effective use of NHS resources.

4.2 The Committee discussed the clinical treatment pathway for type 2 diabetes. It heard from the clinical specialists that although treatment for type 2 diabetes is individualised for each patient, current clinical practice in England broadly follows NICE guidance (see section 6). The Committee heard from the clinical specialists that all of the existing treatments have advantages and disadvantages, and that they do not enable everyone with type 2 diabetes to achieve target HbA1c levels. It further heard that HbA1c values tended to drift upwards over time (even after a good initial response to treatment), with sulfonylureas known to have a relatively high drift rate, and that a particular treatment aim is maintaining consistently tight glycaemic control to produce better long-term outcomes. The Committee heard from the clinical specialists that around 85% of people start treatment with metformin (with many of the remaining 15% starting treatment with a sulfonylurea) and that a sulfonylurea is often subsequently added to metformin for dual therapy. If patients are unable to take a sulfonylurea because of concerns about weight gain or hypoglycaemia, the clinical specialists stated that alternatives such as pioglitazone (a thiazolidinedione), sitagliptin (a DPP‑4 inhibitor) and dapagliflozin (a sodium–glucose cotransporter-2 [SGLT-2] inhibitor, like canagliflozin) may be used in combination with metformin. The Committee heard from the clinical specialists that the use of DPP‑4 inhibitors was increasing and that the use of pioglitazone was decreasing because of concerns about weight gain and safety. The clinical specialists also said that the same treatments could be used in triple therapy and as add-on to insulin therapy. The Committee heard that when considering triple therapy options, many people prefer to take a third oral agent rather than starting insulin (which is recommended in NICE clinical guideline 87) because of fear of hypoglycaemia, the need for injections, and the possible impact on their lifestyle (for example, concerns about keeping their driving licence, or employment). The Committee also noted that increased monitoring is needed with insulin, usually involving secondary care in addition to primary care. The Committee was aware that injectable GLP‑1 analogues were also recommended in NICE guidance as part of dual therapy for a very small proportion of patients who were unable to take several other oral options. They are also recommended as part of triple therapy for a specific population (that is, a high BMI and associated problems, or if insulin treatment would have significant occupational implications, or if weight loss would be beneficial).

4.3 The Committee discussed the most likely place for canagliflozin in the treatment pathway, and which treatments in the NICE scope were the key comparators. The Committee heard from the clinical specialists that they would like to have the option of using canagliflozin as part of dual and triple therapy, and as add-on treatment to insulin. It was aware from the clinical specialists’ input that evidence showed intensive early treatment of type 2 diabetes was associated with reduced macrovascular and microvascular complications, and therefore canagliflozin might be of most benefit early in the pathway. However, it also heard from the clinical specialists that there was extensive experience of using sulfonylureas, and that it was unlikely that healthcare professionals would imminently stop using them in favour of drugs with little or no long-term evidence of their efficacy and safety. However, the Committee further heard from the clinical specialists that this extensive experience with sulfonylureas has shown that they are not an appropriate treatment for all patients (for example, if weight gain is a concern or in older people who are more at risk of injury from a fall secondary to hypoglycaemia). On the basis of the evidence from the clinical specialists, the Committee agreed that canagliflozin was most likely to be used in dual therapy in combination with metformin only for those people for whom a sulfonylurea was not appropriate. Consequently, the Committee concluded that the principal comparator to consider at this place in the pathway would be a DPP‑4 inhibitor (such as sitagliptin). The Committee noted that dapagliflozin, which is an SGLT-2 inhibitor like canagliflozin, is recommended by NICE for use in dual therapy in combination with metformin and that this was also a key comparator. The Committee heard that although pioglitazone is still prescribed, and is suitable for some patients, safety concerns have reduced the use of thiazolidinediones as a class in clinical practice. The Committee concluded that for dual therapy in combination with metformin, DPP‑4 inhibitors and dapagliflozin were the key comparators.

4.4 The Committee heard from the clinical specialists that canagliflozin would also potentially be used as part of triple therapy, principally in combination with metformin and a sulfonylurea. It heard that the clinical specialists considered that the key comparator here would be a DDP-4 inhibitor. The Committee noted that dapagliflozin was not recommended in triple therapy by NICE in technology appraisal guidance 288 except as part of a clinical trial (that is, not in routine clinical practice) because there was no direct trial evidence for dapagliflozin in triple therapy at that time and dapagliflozin’s manufacturers had highlighted limitations in the indirect comparison. The Committee also heard from the clinical specialists that canagliflozin had a place as an add-on treatment to insulin because of its different mechanism of action and the potential for an insulin-sparing effect, which could decrease the rate of hypoglycaemic events. Taking all the evidence into account, including current clinical practice, the Committee concluded that a DPP‑4 inhibitor was the key comparator in triple therapy, and a DPP‑4 inhibitor and dapagliflozin were the key comparators as add-on treatment to insulin.

Clinical effectiveness

4.5 The Committee considered the evidence on the clinical effectiveness of canagliflozin compared with other antidiabetic treatments and noted that the manufacturer’s submission contained the results from 3 head-to-head trials (DIA3006, DIA3009 and DIA3015) comparing canagliflozin with an active comparator: a DPP‑4 inhibitor (sitagliptin) or a sulfonylurea (glimepiride) in dual therapy and a DPP‑4 inhibitor (sitagliptin) in triple therapy. The Committee recalled that the ERG had judged these trials to be of generally good methodological quality. The Committee was aware, however, that most data came from the manufacturer’s network meta-analyses. The Committee heard from the ERG that the manufacturer’s searches had been well conducted and that it was generally satisfied with the manufacturer’s approach to the network meta-analyses, although it was unclear why the triple therapy meta-analyses had been conducted with 26-week data instead of 52-week data. The manufacturer clarified that this was because there were more comparators with data available at 26 weeks than 52 weeks. The Committee accepted this explanation and, although it had some reservations about the duration of the treatment, concluded that the results of the manufacturer’s network meta-analyses provided an appropriate basis for making decisions about the clinical effectiveness of canagliflozin compared with other antidiabetic therapies.

4.6 The Committee discussed the generalisability of the clinical trial populations to the NHS patient population in England that was potentially eligible for treatment with canagliflozin. The Committee was satisfied that the populations in the dual therapy and triple therapy trials were generalisable, but had concerns about the population in the DIA3008 insulin sub-study. This was because the sub-study was part of the CANVAS trial, which only included patients with a history of, or estimated high risk of cardiovascular disease. The Committee heard from the clinical specialists that, in general, by the time patients have tried different oral treatments and progressed to insulin (around 8–10 years from diagnosis), they would be likely to have an increased risk of cardiovascular disease compared with the general population. The Committee was persuaded that patients on insulin for type 2 diabetes would be at increased risk of cardiovascular disease as a result of the condition, and concluded that the results of all the canagliflozin trials (including the DIA3008 insulin sub-study) were generalisable to the population likely to receive canagliflozin in clinical practice in England.

4.7 The Committee considered the clinical effectiveness of canagliflozin as dual therapy in combination with metformin. The Committee noted that the evidence came from 2 clinical trials and a network meta-analysis. It noted that head-to-head trials had compared canagliflozin at doses of 100 mg and 300 mg with glimepiride (a sulfonylurea) and with sitagliptin (a DPP‑4 inhibitor). The Committee considered that, on the basis of current clinical guidelines and clinical practice, SGLT-2 inhibitors would only be considered for use as dual therapy in combination with metformin when sulfonylureas were unsuitable, and that therefore sulfonylureas were not a relevant comparator. However the comparison with sitagliptin was relevant to the decision problem. For sitagliptin, the Committee considered that the data suggested that canagliflozin has a broadly comparable efficacy to sitagliptin in reducing HbA1c, and greater efficacy in reducing body weight and systolic blood pressure. The Committee concluded that, on the basis of the results of the clinical trials and network meta-analyses (see section 3.13–3.16), canagliflozin as part of dual therapy in combination with metformin appeared to provide broadly comparable glycaemic control to the other antidiabetic drugs, including the key comparators DPP‑4 inhibitors and dapagliflozin, and may result in greater weight loss and lowering of blood pressure than DPP‑4 inhibitors.

4.8 The Committee discussed an alternative dual therapy combination that was specified in the NICE scope: canagliflozin in combination with a sulfonylurea. The Committee noted that the manufacturer had not made a case for canagliflozin in combination with sulfonylurea in its submission because most people with diabetes would initially use metformin. The Committee agreed that most patients would start on metformin monotherapy, but was aware that, if metformin was unsuitable, some patients would receive a sulfonylurea instead. However, the Committee concluded that it was unable to make a recommendation on the dual therapy combination of canagliflozin and a sulfonylurea because the manufacturer had not provided any clinical data.

4.9 The Committee discussed the clinical effectiveness of canagliflozin as part of triple therapy in combination with metformin and a sulfonylurea. The Committee noted that the evidence came from 2 clinical trials and a network meta-analysis. It acknowledged the availability of head-to-head trial results for canagliflozin compared with a DPP‑4 inhibitor (sitagliptin), but was aware that DIA3015 only investigated the higher dose of canagliflozin. The Committee noted that DIA3015 showed that canagliflozin 300 mg statistically significantly reduced HbA1c, systolic blood pressure and body weight compared with sitagliptin. The Committee was aware that in DIA3002, which compared canagliflozin with placebo in triple therapy, benefits were seen in reducing HbA1c and body weight, but not systolic blood pressure. The Committee concluded that, on the basis of the clinical trials and the manufacturer’s meta-analysis (see section 3.17), canagliflozin as part of triple therapy in combination with metformin and a sulfonylurea gave a comparable HbA1c reduction compared with a DPP‑4 inhibitor. It further concluded that reduction in weight and systolic blood pressure was greater with canagliflozin than a DPP‑4 inhibitor.

4.10 The Committee discussed the clinical effectiveness of canagliflozin as part of triple therapy in combination with metformin and a thiazolidinedione. The Committee noted that the evidence came from 1 clinical trial and a network meta-analysis. The Committee noted that the clinical trial compared canagliflozin with placebo and not an active comparator, and that it was more effective than placebo in lowering HbA1c, body weight and blood pressure. It heard from the clinical specialists that the use of thiazolidinediones was decreasing in clinical practice in England because of safety concerns about cardiac problems and bladder cancer, as well as its association with weight gain. It concluded that although there was some evidence for the clinical effectiveness of canagliflozin as part of triple therapy in combination with metformin and a thiazolidinedione, this particular triple therapy combination was unlikely to be used routinely in clinical practice. The Committee did not consider this combination further.

4.11 The Committee considered the clinical effectiveness of canagliflozin as an add-on treatment to insulin, noting that the evidence came from 1 clinical trial (the DIA3008 insulin sub-study) and a network meta-analysis. The Committee noted that the trial was placebo controlled and was only 18 weeks long. Despite these limitations, the Committee concluded that, on the basis of the results of the DIA3008 insulin sub-study and the network meta-analysis, canagliflozin as add-on therapy to insulin appeared to be slightly more effective in reducing HbA1c and body weight than DPP‑4 inhibitors and dapagliflozin.

4.12 In light of the dosing options specified in canagliflozin’s marketing authorisation, the Committee discussed how dose escalation with canagliflozin might be implemented in clinical practice in England, and how it might relate to its clinical effectiveness. It was aware that canagliflozin 100 mg and 300 mg had been studied separately in the clinical trials, and that there was no clinical trial evidence for the clinical effectiveness of canagliflozin after dose escalation. It heard from the clinical specialists that there was no experience in escalating the drug dose, and that it was not clear how many patients would need dose escalation, or when this might occur. The Committee concluded that there was uncertainty about the precise clinical effectiveness of canagliflozin when escalating the dose in clinical practice.

4.13 The Committee discussed the adverse events associated with canagliflozin. It heard from clinical specialists that the risk of hypoglycaemia and weight gain were relatively low compared with some other antihyperglycaemic treatments, which was important to people with type 2 diabetes and their healthcare professionals. Focusing on the adverse events that were typically associated with SGLT-2 inhibitors, the Committee heard from the clinical specialists that although rates of genital mycotic infections in the clinical trials were much higher than with placebo, these had generally been resolved by topical treatment and that recurrence rates were low. The Committee did not perceive there to be any major short-term safety concerns and noted that the CANVAS safety trial was ongoing. The Committee concluded that canagliflozin (100 mg and 300 mg doses) had an acceptable safety profile.

Cost effectiveness

4.14 The Committee discussed the manufacturer’s ECHO-T2DM model, which had not been used in previous NICE technology appraisals of technologies used to treat type 2 diabetes. The Committee observed that the manufacturer’s model followed the NICE reference case and methodology, noting that the utility values had mainly been derived from a large European dataset using EQ-5D. It noted the ERG’s view that the model had been well validated, but was aware that the ERG had concerns about the stability of the incremental cost-effectiveness ratios (ICERs) generated using the model, especially when probabilistic uncertainty was included (as it was in the manufacturer’s base case). The Committee heard from the manufacturer that although the values may vary slightly in each model run, the incremental values were highly consistent. The Committee concluded that, despite some uncertainty in the stability of the results, the manufacturer’s economic model was suitable for assessing the cost effectiveness of canagliflozin in combination therapy for treating type 2 diabetes.

4.15 The Committee discussed the cost-effectiveness analyses presented by the manufacturer, noting that these did not include all the comparators in the NICE scope. The Committee was aware that more comparators had been included in the network meta-analyses but then subsequently excluded from the cost-effectiveness analysis because the manufacturer had chosen a single comparator to represent each class. On balance, the Committee concluded that the manufacturer had included an adequate range of comparators for the cost-effectiveness analysis of canagliflozin in dual therapy, triple therapy and as an add-on to insulin therapy.

4.16 The Committee reviewed the manufacturer’s approach to including clinical data in the economic model. It was aware that the manufacturer had used a mixture of data from its meta-analyses and clinical trials, where available. The Committee noted that there was a lack of data in certain loops of the network meta-analyses and that these sometimes indicated slightly more efficacious estimates in favour of canagliflozin compared with some of the direct trial evidence. However, the Committee heard from the ERG that the meta-analyses of clinical effectiveness of canagliflozin and its comparators were sufficiently robust and generally consistent with the direct trial evidence, and had been appropriately incorporated into the model. The Committee therefore concluded that the clinical data incorporated into the manufacturer’s model were acceptable for informing the cost-effectiveness estimates.

4.17 The Committee discussed how the manufacturer had modelled changes in clinical effectiveness over time. It noted that the annual drift values for biochemical and other risk factors had been based on available data (see section 3.30), but that the same annual drift values for HbA1c were used for the SGLT-2 inhibitors, DPP‑4 inhibitors and GLP‑1 analogues. The manufacturer clarified that although there were 4-year data for the DPP‑4 inhibitors and GLP‑1 analogues, only 52-week data were available for canagliflozin. The Committee considered this to have significant implications for the economic modelling, because the manufacturer’s deterministic sensitivity analyses showed that results from the model were most sensitive to varying HbA1c drift. The Committee was aware that the maximum trial duration with canagliflozin was 104 weeks, with most trials being shorter than this, and that extrapolating short-term data over a lifetime time horizon contributed some uncertainty to the cost-effectiveness estimates. However, in the absence of any long-term clinical data, the Committee concluded that the manufacturer’s approach to modelling the long-term effectiveness of canagliflozin was acceptable.

4.18 The Committee discussed the manufacturer’s scenario for dose escalation. It heard from the clinical specialists that it was impossible to predict with any certainty the number of patients who would be eligible for dose escalation. The Committee observed that the summary of product characteristics stated that the higher dose could be used if tighter glycaemic control was needed, but did not provide a definition of this, or specify a time when dose escalation should take place. In the absence of any other information, the Committee accepted that the manufacturer’s assumptions for dose escalation in its scenario analysis were not unreasonable. However, it considered that the results were associated with significant uncertainty and should be interpreted with caution, especially as the manufacturer had assumed that clinical effectiveness was the same in patients who had started on the 100 mg dose and escalated to 300 mg as those who had started and continued on the 300 mg dose. It concluded that the cost effectiveness of canagliflozin in combination therapy for treating type 2 diabetes should instead be informed by considering the cost-effectiveness estimates for the separate 100 mg and 300 mg doses for each treatment comparison.

4.19 The Committee considered the most plausible ICERs for canagliflozin in combination with metformin as dual therapy. Based on clinical specialist opinion, the Committee decided that thiazolidinediones and sulfonylureas were not key comparators in this setting (see section 4.3). The Committee also believed that the GLP‑1 analogues were not key comparators because they are recommended only for a very small population (see section 4.2–4.3). The Committee noted that the manufacturer’s and ERG’s analyses showed that canagliflozin was associated with higher costs and QALYs than DPP‑4 inhibitors and dapagliflozin, but that these incremental differences were small (see sections 3.32–3.37 and 3.49–3.52).The Committee understood that these low incremental costs and health benefits meant the ICERs could vary dramatically in response to even small changes because of a pronounced effect on the cost–effectiveness ratio. The Committee noted that the ERG had carried out a fully incremental analysis including all dual therapy combinations. However, it noted that this included treatment options that would not be appropriate for all patients in clinical practice. The Committee therefore examined appropriate parts of the incremental analysis, together with relevant pairwise analyses, to evaluate the cost effectiveness of canagliflozin in dual therapy compared with the key comparators (DPP-4 inhibitors and dapagliflozin). The Committee saw that in some cases there was a very wide range of estimates of the ICER for the same drug comparison. After exploring the reasons for this, the Committee noted that the differences in costs over a 40-year time horizon were modest (from £1 to £751) and the differences in QALYs were extremely small (from 0.002 to 0.04). The Committee considered that it was important to take these small incremental differences into account when interpreting the ICERs. Overall, the Committee concluded that because of the very small differences in costs and QALYs between canagliflozin and DPP‑4 inhibitors, and between canagliflozin and dapagliflozin, canagliflozin 100 mg and 300 mg in a dual therapy regimen in combination with metformin had been shown to be a cost-effective use of NHS resources. The Committee therefore recommended canagliflozin 100 mg and 300 mg as a treatment option when the alternative treatments would be a DPP-4 inhibitor or dapagliflozin in line with the recommendations in NICE clinical guideline 87 and technology appraisal guidance 288 (that is, if they areat significant risk of hypoglycaemia or its consequences or if a sulfonylurea is not tolerated or contraindicated).

4.20 The Committee considered the most plausible ICERs for canagliflozin in combination with metformin and a sulfonylurea as triple therapy. Based on clinical specialist opinion, the Committee decided that the injectable therapies insulin and GLP‑1 analogues were not key comparators because canagliflozin would be most suitable for people who prefer to add a third oral treatment rather than an injectable one, and also because GLP‑1 analogues are only recommended for a specific patient population (see section 4.2). The Committee noted that the manufacturer’s results showed that there were small incremental differences in costs and QALYs between canagliflozin and the DPP‑4 inhibitors. Although there was considerable uncertainty around the precise ICERs (especially because only canagliflozin 300 mg was investigated in DIA3015), the Committee concluded that because of the small differences in costs and QALYs between canagliflozin and DPP‑4 inhibitors, canagliflozin 100 mg and 300 mg in combination with metformin and a sulfonylurea in a triple therapy regimen had been shown to be a cost-effective use of NHS resources and should be recommended as a treatment option for people with type 2 diabetes.

4.21 The Committee considered the most plausible ICERs for canagliflozin as an add-on treatment to insulin. It noted that the NICE scope specified that the comparators were oral agents, and so disregarded the cost-effectiveness estimates for a GLP‑1 analogue. The Committee concluded that canagliflozin 100 mg and 300 mg had been shown to be a cost-effective use of NHS resources compared with DPP‑4 inhibitors and dapagliflozin as an add-on treatment to insulin because of its very small incremental costs and incremental QALYs. The Committee recommended canagliflozin as a treatment option for people with diabetes that is inadequately controlled by insulin with or without other oral antidiabetic drugs.

4.22 The Committee discussed whether canagliflozin was an innovative treatment and if there were any additional QALYs that had not been included in the manufacturer’s model. The manufacturer said that that there were no health benefits that had not been captured in the model; however, it did confirm that the model did not incorporate disutilities for pioglitazone’s long-term adverse effects, including fractures and bladder cancer. The Committee acknowledged that this omission could influence the cost-effectiveness estimates for the comparisons of canagliflozin and a thiazolidinedione. The Committee had heard from clinical specialists that pioglitazone use is declining because of safety concerns, meaning it is not a key comparator, and concluded that omitting long-term disutilities for pioglitazone was unlikely to have an impact on its decision-making. The Committee concluded that all substantial benefits related to treatment with canagliflozin had been captured in the QALY calculation.

Summary of Appraisal Committee's key conclusions

TAXXX

Appraisal title: Canagliflozin in combination therapy

for treating type 2 diabetes

Section
Key conclusion

Canagliflozin in a dual therapy regimen in combination with metformin is recommended as an option for treating type 2 diabetes, only if the person is at significant risk of hypoglycaemia or its consequences, or a sulfonylurea is contraindicated or not tolerated.

Canagliflozin in a triple therapy regimen in combination with metformin and a sulfonylurea is recommended as an option for treating type 2 diabetes.

Canagliflozin in combination with insulin with or without other antidiabetic drugs is recommended as an option for treating type 2 diabetes.

The Committee concluded that because of the very small differences in costs and QALYs between canagliflozin and its key comparators, canagliflozin 100 mg and 300 mg had been shown to be a cost-effective use of NHS resources as dual therapy in combination with metformin, triple therapy in combination with metformin and a sulfonylurea, and as an add-on treatment to insulin.

1.1, 1.2, 1.3, 4.19, 4.20, 4.21
Current practice
Clinical need of patients, including the availability of alternative treatments

The Committee heard from the clinical specialists that treatment for type 2 diabetes is individualised for each patient and that all existing treatments had advantages and disadvantages. It further heard that current treatments do not enable everyone with type 2 diabetes to achieve and maintain target HbA1c levels.

The Committee noted that some people with type 2 diabetes are unable to take a sulfonylurea because of concerns about weight gain or hypoglycaemia, that the use of pioglitazone was decreasing because of concerns about weight gain and safety and that many people prefer to delay starting insulin in favour of other options because of fear of hypoglycaemia and its consequences, and the need for injections.

4.2, 4.3
The technology

Proposed benefits of the technology

How innovative is the technology in its potential to make a significant and substantial impact on health-related benefits?

Canagliflozin (Invokana, Janssen-Cilag) is an orally administered selective sodium–glucose cotransporter-2 (SGLT-2) inhibitor. It lowers blood glucose in people with type 2 diabetes by blocking the reabsorption of glucose in the kidneys and promoting excretion of excess glucose in the urine.

The Committee concluded that all substantial benefits related to treatment with canagliflozin had been captured in the QALY calculation.

2.1

4.22

What is the position of the treatment in the pathway of care for the condition?

On the basis of the evidence from the clinical specialists, the Committee agreed that canagliflozin in dual therapy in combination with metformin was likely to be used in people for whom a sulfonylurea was not appropriate. The Committee concluded that DPP 4 inhibitors and dapagliflozin were the key comparators.

The Committee heard from the clinical specialists that canagliflozin would also potentially be used as part of triple therapy, principally in combination with metformin and a sulfonylurea, and concluded a DPP 4 inhibitor was the key comparator.

The Committee also heard from the clinical specialists that canagliflozin had a place as an add-on treatment to insulin, and concluded that DPP-4 inhibitors and dapagliflozin were the key comparators.

4.3, 4.4
Adverse reactions The summary of product characteristics states the following adverse reactions for canagliflozin are the most commonly reported: hypoglycaemia in combination with insulin or a sulphonylurea, vulvovaginal candidiasis, urinary tract infection, and polyuria or pollakiuria (that is, urinary frequency). The Committee did not perceive there to be any major short-term safety concerns and noted that the CANVAS safety trial was ongoing. The Committee concluded that canagliflozin (100 mg and 300 mg doses) had an acceptable safety profile. 2.3, 4.13
Evidence for clinical effectiveness
Availability, nature and quality of evidence The Committee noted that the manufacturer’s submission described the results from 6 randomised controlled trials, including 3 head-to-head trials (DIA3006, DIA3009 and DIA3015) comparing canagliflozin with an active comparator: a DPP-4 inhibitor (sitagliptin) or a sulfonylurea (glimepiride) in dual therapy and a DPP-4 inhibitor (sitagliptin) in triple therapy. It concluded that the results of the manufacturer’s network meta-analyses provided an appropriate basis for making decisions about the clinical effectiveness of canagliflozin compared with other antidiabetic therapies. However, the Committee concluded that it was unable to make a recommendation on the dual therapy combination of canagliflozin and a sulfonylurea because the manufacturer had not provided any clinical data. 4.5, 4.8
Relevance to general clinical practice in the NHS The Committee was persuaded by the clinical specialists’ opinion that patients on insulin for type 2 diabetes would be at increased risk of cardiovascular disease as a result of the condition, and concluded the results from the DIA3008 insulin sub-study, as well as those from the other trials, were generalisable to the population likely to receive canagliflozin in clinical practice in England. 4.6
Uncertainties generated by the evidence The Committee was aware that there was no evidence for the clinical effectiveness of canagliflozin after dose escalation, as described in canagliflozin’s marketing authorisation, because canagliflozin 100 mg and 300 mg had been studied separately in the clinical trials. The Committee concluded that there was uncertainty about the precise clinical effectiveness of canagliflozin when escalating the dose in clinical practice. 4.12
Are there any clinically relevant subgroups for which there is evidence of differential effectiveness? Not applicable.  
Estimate of the size of the clinical effectiveness including strength of supporting evidence For dual therapy in combination with metformin, the Committee concluded that, on the basis of the results of the clinical trials and network meta-analyses, canagliflozin appeared to provide broadly comparable glycaemic control to the other antidiabetic drugs, including the key comparators DPP-4 inhibitors and dapagliflozin, and may result in greater weight loss and lowering of blood pressure than DPP-4 inhibitors. 4.7
For triple therapy in combination with metformin and a sulfonylurea, the Committee concluded that, on the basis of the clinical trials and the manufacturer’s meta-analysis, canagliflozin as part of triple therapy in combination with metformin and a sulfonylurea gave a comparable HbA1c reduction compared with a DPP 4 inhibitor. It further concluded that reduction in weight and systolic blood pressure was greater with canagliflozin than a DPP 4 inhibitor. 4.9
For triple therapy in combination with metformin and a thiazolidinedione, the Committee noted that there was some evidence of canagliflozin’s clinical effectiveness. However, it concluded that this particular triple therapy combination was unlikely to be used routinely in clinical practice and did not consider this combination further. 4.10
For add-on treatment to insulin, the Committee concluded that, on the basis of the results of the DIA3008 insulin sub-study and the network meta-analysis, canagliflozin as add-on therapy to insulin appeared to be slightly more effective in reducing HbA1c and body weight than a DPP-4 inhibitor and dapagliflozin. 4.11
Evidence for cost effectiveness
Availability and nature of evidence The Committee observed that the manufacturer’s ECHO-T2DM model followed the NICE reference case and methodology and noted the ERG’s view that the model had been well validated. The Committee concluded that the manufacturer’s economic model was suitable for assessing the cost effectiveness of canagliflozin in combination therapy for treating type 2 diabetes. 4.14
Uncertainties around and plausibility of assumptions and inputs in the economic model The Committee heard from the ERG that, despite some uncertainty, the manufacturer’s meta-analyses were sufficiently robust and generally consistent with the direct trial evidence, and had been appropriately incorporated into the model. The Committee concluded that the clinical data incorporated into the manufacturer’s model were acceptable for informing the cost-effectiveness estimates. 4.16
The Committee had some concerns about how the manufacturer had modelled changes in clinical effectiveness over time (especially the annual drift values for HbA1c, because this was a key driver of the model). However, in the absence of any long-term clinical data, the Committee concluded that the manufacturer’s approach to modelling the long-term effectiveness of canagliflozin was acceptable. 4.17
The Committee accepted the manufacturer’s assumptions for dose escalation in its scenario analysis but concluded they were associated with significant uncertainty and the decision-making on cost effectiveness should instead be informed by considering the cost-effectiveness estimates for the separate 100 mg and 300 mg doses for each treatment comparison’. 4.18

Incorporation of health-related quality-of-life benefits and utility values

Have any potential significant and substantial health-related benefits been identified that were not included in the economic model, and how have they been considered?

The Committee observed that the utility values in the manufacturer’s model had mainly been derived from a large European dataset using EQ-5D, in line with the NICE reference case. 4.14
Although Committee heard that the manufacturer’s model did not incorporate disutilities for pioglitazone’s long-term adverse effects, it concluded that this omission was unlikely to impact on its decision-making because of declining use of pioglitazone in England (partly because of safety concerns). The Committee concluded that all substantial benefits related to treatment with canagliflozin had been captured in the QALY calculation. 4.22
Are there specific groups of people for whom the technology is particularly cost effective? Not applicable.  
What are the key drivers of cost effectiveness? The Committee concluded that the manufacturer’s deterministic sensitivity analyses showed that HbA1c drift was a key driver of the model. 4.17
Most likely cost-effectiveness estimate (given as an ICER) The Committee concluded that because of the very small differences in costs and QALYs between canagliflozin and its key comparators, canagliflozin 100 mg and 300 mg had been shown to be a cost-effective use of NHS resources as dual therapy in combination with metformin, triple therapy in combination with metformin and a sulfonylurea, and as an add-on treatment to insulin. 4.19, 4.20, 4.21
Additional factors taken into account
Patient access schemes (PPRS) Not applicable.  
End-of-life considerations Not applicable.  
Equalities considerations and social value judgements No issues relating to equality considerations were raised in the submissions or the Committee meeting.  
       

5 Implementation

5.1 Section 7(6) of the National Institute for Health and Care Excellence (Constitution and Functions) and the Health and Social Care Information Centre (Functions) Regulations 2013 requires clinical commissioning groups, NHS England and, with respect to their public health functions, local authorities to comply with the recommendations in this appraisal within 3 months of its date of publication.

5.2 When NICE recommends a treatment ‘as an option’, the NHS must make sure it is available within the period set out in the paragraph above. This means that, if a patient has type 2 diabetes and the doctor responsible for their care thinks that canagliflozin is the right treatment, it should be available for use, in line with NICE’s recommendations.

5.3 NICE has developed tools [link to www.nice.org.uk/guidance/TAXXX] to help organisations put this guidance into practice (listed below). [NICE to amend list as needed at time of publication]

  • Slides highlighting key messages for local discussion.
  • Costing template and report to estimate the national and local savings and costs associated with implementation.
  • Implementation advice on how to put the guidance into practice and national initiatives that support this locally.
  • A costing statement explaining the resource impact of this guidance.
  • Audit support for monitoring local practice.

6 Related NICE guidance

Details are correct at the time of consultation. Further information is available on the NICE website.

Related NICE guidance

Published

Under development

  • Type 2 diabetes in adults: management of type 2 diabetes in adults. NICE clinical guideline, publication expected August 2015.

NICE pathways

7 Proposed date for review of guidance

7.1 NICE proposes that the guidance on this technology is considered for review by the Guidance Executive in May 2017. NICE welcomes comment on this proposed date. The Guidance Executive will decide whether the technology should be reviewed based on information gathered by NICE, and in consultation with consultees and commentators.

Jane Adam
Chair, Appraisal Committee
January 2014

8 Appraisal Committee members, guideline representatives and NICE project team

Appraisal Committee members

The Appraisal Committees are standing advisory committees of NICE. Members are appointed for a 3-year term. A list of the Committee members who took part in the discussions for this appraisal appears below. There are 4 Appraisal Committees, each with a chair and vice chair. Each Appraisal Committee meets once a month, except in December when there are no meetings. Each Committee considers its own list of technologies, and ongoing topics are not moved between Committees.

Committee members are asked to declare any interests in the technology to be appraised. If it is considered there is a conflict of interest, the member is excluded from participating further in that appraisal.

The minutes of each Appraisal Committee meeting, which include the names of the members who attended and their declarations of interests, are posted on the NICE website.

Dr Jane Adam (Chair)
Department of Diagnostic Radiology, St George’s Hospital, London

Dr Graham Ash
Consultant in General Adult Psychiatry, Lancashire Care NHS Foundation Trust

Dr Jeremy Braybrooke
Consultant Medical Oncologist, University Hospitals Bristol NHS Foundation Trust

Dr Simon Bond
Senior Statistician, Cambridge Clinical Trials Unit

Professor Aileen Clarke
Professor of Public Health & Health Services Research, University of Warwick

Mr Andrew England
Lecturer in Medical Imaging, NIHR Fellow, University of Liverpool

Dr Peter Heywood
Consultant Neurologist, Frenchay Hospital, Bristol

Dr Ian Lewin
Consultant Endocrinologist, North Devon District Hospital

Dr Louise Longworth
Reader in Health Economics, HERG, Brunel University

Dr Anne McCune
Consultant Hepatologist, University Hospitals Bristol NHS Foundation Trust

Professor John McMurray
Professor of Medical Cardiology, University of Glasgow

Dr Alec Miners
Lecturer in Health Economics, London School of Hygiene and Tropical Medicine

Dr Mohit Misra
GP, Queen Elizabeth Hospital, London

Ms Sarah Parry
CNS Paediatric Pain Management, Bristol Royal Hospital for Children

Ms Pamela Rees
Lay Member

Dr Ann Richardson
Lay Member

Ms Ellen Rule
Programme Director, NHS Bristol

Mr Stephen Sharp
Senior Statistician, MRC Epidemiology Unit, University of Cambridge

Dr Brian Shine
Consultant Chemical Pathologist, John Radcliffe Hospital

Dr Peter Sims
GP, Devon

Dr Eldon Spackman
Research Fellow, Centre for Health Economics, University of York

Mr David Thomson
Lay Member

Dr John Watkins
Clinical Senior Lecturer, Cardiff University; Consultant in Public Health Medicine, National Public Health Service Wales

Dr Olivia Wu
Reader in Health Economics, University of Glasgow

NICE project team

Each technology appraisal is assigned to a team consisting of 1 or more health technology analysts (who act as technical leads for the appraisal), a technical adviser and a project manager.

Linda Landells
Technical Lead

Joanna Richardson
Technical Adviser

Bijal Joshi
Project Manager

9 Sources of evidence considered by the Committee

A. The Evidence Review Group (ERG) report for this appraisal was prepared by Southampton Health Technology Assessments Centre (SHTAC), University of Southampton:

  • Copley V, Pickett K, Shepherd J, et al. Canagliflozin in combination therapy for treating type 2 diabetes, December 2013

B. The following organisations accepted the invitation to participate in this appraisal as consultees and commentators. They were invited to comment on the draft scope, the ERG report and the appraisal consultation document (ACD). Organisations listed in I were also invited to make written submissions. Organisations listed in II and III had the opportunity to give their expert views. Organisations listed in I, II and III also have the opportunity to appeal against the final appraisal determination.

I. Manufacturer/sponsor:

  • Janssen

II. Professional/specialist and patient/carer groups:

  • Association of British Clinical Diabetologists
  • Diabetes UK
  • National Diabetes Nurse Consultant Group
  • Royal College of Physicians

III. Other consultees:

  • Department of Health
  • NHS England
  • Welsh Government

IV. Commentator organisations (did not provide written evidence and without the right of appeal):

  • Boehringer Ingelheim
  • Commissioning Support Appraisal Service
  • Department of Health, Social Services and Public Safety for Northern Ireland
  • Eli Lilly UK
  • Healthcare Improvement Scotland
  • Medicines and Healthcare Products Regulatory Agency (MHRA)
  • Merck Sharp and Dohme
  • National Clinical Guidelines Centre
  • National Institute for Health Research Health Technology Assessment Programme
  • Novo Nordisk
  • Sanofi
  • Southampton Health Technology Assessments Centre (SHTAC), University of Southampton

C. The following individuals were selected from clinical specialist and patient expert nominations from the consultees and commentators. They gave their expert personal view on canagliflozin by attending the initial Committee discussion and providing written evidence to the Committee. They are invited to comment on the ACD.

  • Professor David Matthews, Professor of Diabetes Medicine, nominated by an organisation representing Janssen – clinical specialist
  • Dr Thozhukat Sathyapalan, Reader/Honorary Consultant medical Consultant Endocrinologist, nominated by organisation representing Royal College of Physicians and Association of British Clinical Diabetologists – clinical specialist

E. Representatives from the following manufacturer/sponsor attended Committee meetings. They contributed only when asked by the Committee chair to clarify specific issues and comment on factual accuracy.

  • Janssen

This page was last updated: 17 March 2014