IMFINZI Concentrate for solution for infusion Ref.[8680] Active ingredients: Durvalumab

Source: European Medicines Agency (EU)  Revision Year: 2024  Publisher: AstraZeneca AB, SE-151 85 Södertälje, Sweden

Pharmacodynamic properties

Pharmacotherapeutic group: Antineoplastic agents, monoclonal antibodies and antibody drug conjugates, PD-1/PDL-1 (Programmed cell death protein 1/death ligand 1) inhibitors
ATC code: L01FF03

Mechanism of action

Expression of programmed cell death ligand-1 (PD-L1) protein is an adaptive immune response that helps tumours evade detection and elimination by the immune system. PD-L1 can be induced by inflammatory signals (e.g., IFN-gamma) and can be expressed on both tumour cells and tumour-associated immune cells in tumour microenvironment. PD-L1 blocks T-cell function and activation through interaction with PD-1 and CD80 (B7.1). By binding to its receptors, PD-L1 reduces cytotoxic T-cell activity, proliferation and cytokine production.

Durvalumab is a fully human, immunoglobulin G1 kappa (IgG1κ) monoclonal antibody that selectively blocks the interaction of PD-L1 with PD-1 and CD80 (B7.1). Durvalumab does not induce antibody dependent cell-mediated cytotoxicity (ADCC). Selective blockade of PD-L1/PD-1 and PD-L1/CD80 interactions enhances antitumour immune responses and increases T-cell activation.

The combination of tremelimumab, a CTLA-4 inhibitor and durvalumab, a PD-L1 inhibitor functions to enhance anti-tumour T-cell activation and function at multiple stages of the immune response resulting in improved anti-tumour responses. In murine syngeneic tumour models, dual blockade of PD-L1 and CTLA-4 resulted in enhanced anti-tumour activity.

Clinical efficacy and safety

Durvalumab doses of 10 mg/kg every 2 weeks or 1 500 mg every 4 weeks were evaluated in NSCLC and ES-SCLC clinical studies. Based on the modeling and simulation of exposure, exposure-safety relationships and exposure-efficacy data comparisons, there are no anticipated clinically significant differences in efficacy and safety between durvalumab doses of 10 mg/kg every 2 weeks or 1 500 mg every 4 weeks.

NSCLC – PACIFIC Study

The efficacy of IMFINZI was evaluated in the PACIFIC Study, a randomised, double-blind, placebo-controlled, multicentre study in 713 patients with locally advanced, unresectable NSCLC. Patients had completed at least 2 cycles of definitive platinum-based chemotherapy with radiation therapy within 1 to 42 days prior to initiation of the study and had a ECOG performance status of 0 or 1. Ninety-two percent of patients had received a total dose of 54 to 66 Gy of radiation. The study excluded patients who had progressed following chemoradiation therapy, patients with prior exposure to any anti-PD-1 or anti-PD-L1 antibody, patients with active or prior documented autoimmune disease within 2 years of initiation of the study; a history of immunodeficiency; a history of severe immune-mediated adverse reactions; medical conditions that required systemic immunosuppression, except physiological dose of systemic corticosteroids; active tuberculosis or hepatitis B or C or HIV infection or patients receiving live attenuated vaccine within 30 days before or after the start of IMFINZI. Patients were randomised 2:1 to receive 10 mg/kg IMFINZI (n=476) or 10 mg/kg placebo (n=237) via intravenous infusion every 2 weeks for up to 12 months or until unacceptable toxicity or confirmed disease progression. Randomisation was stratified by gender, age (<65 years vs. ≥65 years) and smoking status (smoker vs. non-smoker). Patients with disease control at 12 months were given the option to be re-treated upon disease progression. Tumour assessments were conducted every 8 weeks for the first 12 months and then every 12 weeks thereafter.

Patients were enrolled regardless of their tumour PD-L1 expression level. Where available, archival tumour tissue specimens taken prior to chemoradiation therapy were retrospectively tested for PD-L1 expression on tumour cells (TC) using the VENTANA PD-L1 (SP263) IHC assay. Of the 713 patients randomised, 63% of patients provided a tissue sample of sufficient quality and quantity to determine PD-L1 expression and 37% were unknown.

The demographics and baseline disease characteristics were well balanced between study arms. Baseline demographics of the overall study population were as follows: male (70%), age ≥65 years (45%), age ≥75 years (8%), White (69%), Asian (27%), other (4%), current smoker (16%), past-smoker (75%), never smoker (9%), ECOG Performance Status 0 (49%), ECOG Performance Status 1 (51%). Disease characteristics were as follows: Stage IIIA (53%), Stage IIIB (45%), histological sub-groups of squamous (46%), non-squamous (54%). Of 451 patients with PD-L1 expression available, 67% were TC ≥1% [PD-L1 TC 1-24% (32%), PD-L1 TC ≥25% (35%)] and 33% were TC <1%.

The two primary endpoints of the study were progression-free survival (PFS) and overall survival (OS) of IMFINZI vs. placebo. Secondary efficacy endpoints included PFS at 12 months (PFS 12) and 18 months (PFS 18) from randomisation and Time from Randomisation to Second Progression (PFS2). PFS was assessed by Blinded Independent Central Review (BICR) according to RECIST v1.1.

The study demonstrated a statistically significant improvement in PFS in the IMFINZI-treated group compared with the placebo group [hazard ratio (HR)=0.52 (95% CI: 0.42, 0.65), p<0.0001]. The study demonstrated a statistically significant improvement in OS in the IMFINZI-treated group compared with the placebo group [HR=0.68 (95% CI: 0.53, 0.87), p=0.00251]. See Table 3 and Figures 1 and 2.

In the 5 year follow-up analysis, with a median follow-up of 34.2 months, IMFINZI continued to demonstrate improved OS and PFS compared to placebo. The OS and PFS results from the primary analysis and the follow-up analysis are summarized in Table 5.

Table 5. Efficacy results for the PACIFIC Study:

 Primary analysisa 5 year follow-up analysisb
IMFINZI
(n=476)
Placebo
(n=237)
IMFINZI
(n=476)
Placebo
(n=237)
OS
Number of deaths (%) 183 (38.4%) 116 (48.9%) 264 (55.5%) 155 (65.4%)
Median (months)
(95% CI)
NR
(34.7, NR)
28.7
(22.9, NR)
47.5
(38.1, 52.9)
29.1
(22.1, 35.1)
HR (95% CI) 0.68 (0.53, 0.87) 0.72 (0.59, 0.89)
2-sided p-value 0.00251 
OS at 24 months (%)
(95% CI)
66.3%
(61.7%, 70.4%)
55.6%
(48.9%, 61.3%)
66.3%
(61.8%, 70.4%)
55.3%
(48.6%, 61.4%)
p-value 0.005 
OS at 48 months (%)
(95% CI)
  49.7%
(45.0%, 54.2%)
36.3%
(30.1%, 42.6%)
OS at 60 months (%)
(95% CI)
  42.9%
(38.2%, 47.4%)
33.4%
(27.3%, 39.6%)
PFS
Number of events (%) 214 (45.0%) 157 (66.2%) 268 (56.3%) 175 (73.8%)
Median PFS (months)
(95% CI)
16.8
(13.0, 18.1)
5.6
(4.6, 7.8)
16.9
(13.0, 23.9)
5.6
(4.8, 7.7)
HR (95% CI) 0.52 (0.42, 0.65) 0.55 (0.45, 0.68)
p-value p<0.0001 
PFS at 12 months (%)
(95% CI)
55.9%
(51.0%, 60.4%)
35.3%
(29.0%, 41.7%)
55.7%
(51.0%, 60.2%)
34.5%
(28.3%, 40.8%)
PFS at 18 months (%)
(95% CI)
44.2%
(37.7%, 50.5%)
27.0%
(19.9%, 34.5%)
49.1%
(44.2%, 53.8%)
27.5%
(21.6%, 33.6%)
PFS at 48 months (%)
(95% CI)
  35.0%
(29.9%, 40.1%)
19.9%
(14.4%, 26.1%)
PFS at 60 months (%)
(95% CI)
  33.1%
(28.0%, 38.2%)
19.0%
(13.6%, 25.2%)
PFS2c
Median PFS2 (months)
(95% CI)
28.3
(25.1, 34.7)
17.1
(14.5, 20.7)
  
HR (95% CI) 0.58 (0.46, 0.73)  
p-value p<0.0001 

a Primary analysis of PFS at data cut-off 13 February 2017. Primary analysis of OS and PFS2 at data cut-off 22 March 2018.
b Follow-up OS and PFS analysis at data cut-off 11 January 2021.
c PFS2 is defined as the time from the date of randomisation until the date of second progression (defined by local standard clinical practice) or death.
NR: Not Reached

Figure 1. Kaplan-Meier curve of OS:

Figure 2. Kaplan-Meier curve of PFS:

The improvements in PFS and OS in favour of patients receiving IMFINZI compared to those receiving placebo were consistently observed in all predefined subgroups analysed, including ethnicity, age, gender, smoking history, EGFR mutation status and histology.

Post-hoc subgroup analysis by PD-L1 expression Additional subgroup analyses were conducted to evaluate the efficacy by tumour PD-L1 expression (≥25%, 1-24%, ≥1%, <1%) and for patients whose PD-L1 status cannot be established (PD-L1 unknown). PFS and OS results from the 5 year follow-up analysis are summarised in Figures 3, 4, 5 and 6.

Figure 3. Kaplan-Meier curve of OS for PD-L1 TC ≥1%:

Figure 4. Kaplan-Meier curve of PFS for PD-L1 TC ≥1%:

Figure 5. Forest plot of OS by PD-L1 expression:

Figure 6. Forest plot of PFS by PD-L1 expression:

Overall the safety profile of durvalumab in PD-L1 TC ≥1% subgroup was consistent with the intent to treat population, as was the PD-L1 TC <1% subgroup.

Patient-reported outcomes (PRO)

Patient-reported symptoms, function and health-related quality of life (HRQoL) were collected using the EORTC QLQ-C30 and its lung cancer module (EORTC QLQ-LC13). The LC13 and C30 were assessed at baseline, every 4 weeks for the first 8 weeks, followed by every 8 weeks until completion of the treatment period or discontinuation of IMFINZI due to toxicity or disease progression. Compliance was similar between the IMFINZI and placebo treatment groups (83% vs. 85.1% overall of evaluable forms completed).

At baseline, no differences in patient-reported symptoms, function and HRQoL were observed between IMFINZI and placebo groups. Throughout the duration of the study to Week 48, there was no clinically meaningful difference between IMFINZI and placebo groups in symptoms, functioning and HRQoL (as assessed by a difference of greater than or equal to 10 points).

NSCLC – POSEIDON Study

POSEIDON was a study designed to evaluate the efficacy of IMFINZI with or without tremelimumab in combination with platinum-based chemotherapy. POSEIDON was a randomised, open-label, multicenter study in 1013 metastatic NSCLC patients with no sensitising epidermal growth factor receptor (EGFR) mutation or anaplastic lymphoma kinase (ALK) genomic tumour aberrations. Patients with histologically or cytologically documented metastatic NSCLC were eligible for enrolment. Patients had no prior chemotherapy or any other systemic therapy for metastatic NSCLC. Prior to randomisation, patients had tumour PD-L1 status confirmed by using the Ventana PD-L1 (SP263) assay. Patients had a World Health Organization (WHO)/Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 at enrolment.

The study excluded patients with active or prior documented autoimmune disease; active and/or untreated brain metastases; a history of immunodeficiency; administration of systemic immunosuppression within 14 days before the start of IMFINZI or tremelimumab, except physiological dose of systemic corticosteroids; active tuberculosis or hepatitis B or C or HIV infection; or patients receiving live attenuated vaccine within 30 days before or after the start of IMFINZI and/or tremelimumab (see section 4.4).

Randomisation was stratified by tumour cells (TC) PD-L1 expression (TC ≥ 50% vs. TC < 50%), disease stage (Stage IVA vs. Stage IVB, per the 8th edition of American Joint Committee on Cancer), and histology (non-squamous vs. squamous).

Patients were randomised 1:1:1 to receive:

  • Arm 1: IMFINZI 1 500 mg with tremelimumab 75 mg and platinum-based chemotherapy every 3 weeks for 4 cycles followed by, IMFINZI 1 500 mg every 4 weeks as monotherapy. A fifth dose of tremelimumab 75 mg was given at Week 16 alongside IMFINZI dose 6.
  • Arm 2: IMFINZI 1 500 mg and platinum-based chemotherapy every 3 weeks for 4 cycles, followed by IMFINZI 1 500 mg every 4 weeks as monotherapy.
  • Arm 3: Platinum-based chemotherapy every 3 weeks for 4 cycles. Patients could receive 2 additional cycles (a total of 6 cycles post-randomisation), as clinically indicated, at the Investigator’s discretion.

In the 3 treatment arms, patients received one of the following histology-based chemotherapy regimens:

  • Non-squamous NSCLC
    • Pemetrexed 500 mg/m² with carboplatin AUC 5-6 or cisplatin 75 mg/m² every 3 weeks. Unless contraindicated by the investigator, pemetrexed maintenance could be given.
  • Squamous NSCLC
    • Gemcitabine 1 000 or 1 250 mg/m² on Days 1 and 8 with cisplatin 75 mg/m² or carboplatin AUC 5-6 on Day 1 every 3 weeks.
  • Non-squamous or squamous NSCLC
    • Nab-paclitaxel 100 mg/m² on Days 1, 8, and 15 with carboplatin AUC 5-6 on Day 1 every 3 weeks.

Tremelimumab was given up to a maximum of 5 doses unless there was disease progression or unacceptable toxicity. IMFINZI and histology-based pemetrexed maintenance therapy (when applicable) was continued until disease progression or unacceptable toxicity.

Tumour assessments were conducted at Week 6 and Week 12 from the date of randomisation, and then every 8 weeks until confirmed objective disease progression. Survival assessments were conducted every 2 months following treatment discontinuation.

The dual primary endpoints of the study were PFS and OS for IMFINZI + platinum-based chemotherapy vs. platinum-based chemotherapy alone. The key secondary endpoints of the study were PFS and OS for IMFINZI + tremelimumab + platinum-based chemotherapy and platinum-based chemotherapy alone. The secondary endpoints included objective response rate (ORR) and Duration of Response (DoR). PFS, ORR, and DoR, were assessed using BICR according to RECIST v1.1

The demographics and baseline disease characteristics were well-balanced between study arms. Baseline demographics of the overall study population were as follows: male (76.0%), age ≥65 years (47.1%), age ≥75 years (11.3%) median age 64 years (range: 27 to 87 years), White (55.9%), Asian (34.6%), Black or African American (2.0%), Other (7.6%), non-Hispanic or Latino (84.2%), current smoker or past-smoker (78.0%), WHO/ECOG PS 0 (33.4%), WHO/ECOG PS 1 (66.5%). Disease characteristics were as follows: Stage IVA (50.0%), Stage IVB (49.6%), histological sub-groups of squamous (36.9%), non-squamous (62.9%), brain metastases (10.5%), PD-L1 expression TC≥ 50% (28.8%), PD-L1 expression TC <50% (71.1%).

The study showed a statistically significant improvement in OS with IMFINZI + tremelimumab + platinum-based chemotherapy vs. platinum-based chemotherapy. IMFINZI + tremelimumab + platinum-based chemotherapy showed a statistically significant improvement in PFS vs. platinumbased chemotherapy alone. The results are summarised below.

Table 6. Efficacy results for the POSEIDON study:

 Arm 1: IMFINZI+tremelimumab
+platinum-based chemotherapy
(n=338)
Arm 3: Platinum-based
chemotherapy
(n=337)
OSa
Number of deaths (%) 251 (74.3) 285 (84.6)
Median OS (months)
(95% CI)
14.0
(11.7, 16.1)
11.7
(10.5, 13.1)
HR (95% CI)b 0.77 (0.650, 0.916)
p-valuec 0.00304
PFSa
Number of events (%) 238 (70.4) 258 (76.6)
Median PFS (months)
(95% CI)
6.2
(5.0, 6.5)
4.8
(4.6, 5.8)
HR (95% CI)b 0.72 (0.600, 0.860)
p-valuec 0.00031
ORR n (%) d,e 130 (38.8) 81 (24.4)
Complete Response n (%) 2 (0.6) 0
Partial Response n (%) 128 (38.2) 81 (24.4)
Median DoR (months)
(95% CI)d,e
9.5
(7.2, NR)
5.1
(4.4, 6.0)

a Analysis of PFS at data cut off 24 July 2019 (median follow up 10.15 months). Analysis of OS at data cut off 12 March 2021 (median follow up 34.86 months). The boundaries for declaring efficacy (Arm 1 vs. Arm 3: PFS 0.00735, OS 0.00797; 2-sided) were determined by a Lan-DeMets alpha spending function that approximates an O’Brien Fleming approach. PFS was assessed by BICR according to RECIST v1.1.
b HR are derived using a Cox pH model stratified by PD-L1, histology and disease stage.
c 2-sided p-value based on a log-rank test stratified by PD-L1, histology and disease stage.
d Confirmed Objective Response.
e Post-hoc analysis.
NR = Not Reached, CI = Confidence Interval

Figure 7. Kaplan-Meier curve of OS:

Figure 8. Kaplan-Meier curve of PFS:

Figure 9 summarises efficacy results of OS by tumour PD-L1 expression in prespecified subgroup analyses.

Figure 9. Forest plot of OS by PD-L1 expression for IMFINZI+tremelimumab+platinum-based chemotherapy vs. platinum-based chemotherapy:

Elderly population

A total of 75 patients aged ≥75 years were enrolled in the IMFINZI in combination with tremelimumab and chemotherapy (n=35) and platinum-based chemotherapy only (n=40) arms of the POSEIDON study. An exploratory HR of 1.05 (95% CI: 0.64, 1.71) for OS was observed for the IMFINZI in combination with tremelimumab and platinum-based chemotherapy vs. platinum-based chemotherapy within this study subgroup. Due to the exploratory nature of this subgroup analysis no definitive conclusions can be drawn, but caution is suggested when considering this regimen for elderly patients.

SCLC – CASPIAN Study

CASPIAN was a study designed to evaluate the efficacy of IMFINZI with or without tremelimumab in combination with etoposide and either carboplatin or cisplatin. CASPIAN was a randomized, openlabel, multicentre study in 805 treatment naïve ES-SCLC patients with WHO/ECOG Performance status of 0 or 1, body weight >30 kg, suitable to receive a platinum-based chemotherapy regimen as first-line treatment for SCLC, with life expectancy ≥12 weeks, at least one target lesion by RECIST 1.1 and adequate organ and bone marrow function. Patients with asymptomatic or treated brain metastases were eligible. The study excluded patients with a history of chest radiation therapy; a history of active primary immunodeficiency; autoimmune disorders including paraneoplastic syndrome (PNS); active or prior documented autoimmune or inflammatory disorders; use of systemic immunosuppressants within 14 days before the first dose of the treatment except physiological dose of systemic corticosteroids; active tuberculosis or hepatitis B or C or HIV infection; or patients receiving live attenuated vaccine within 30 days before or after the start of IMFINZI.

Randomisation was stratified by the planned platinum-based (carboplatin or cisplatin) therapy in cycle 1.

Patients were randomised 1:1:1 to receive:

  • Arm 1: IMFINZI 1 500 mg + tremelimumab 75 mg + etoposide and either carboplatin or cisplatin.
  • Arm 2: IMFINZI 1 500 mg + etoposide and either carboplatin or cisplatin.
  • Arm 3: Either carboplatin (AUC 5 or 6 mg/ml/min) or cisplatin (75-80 mg/m²) on Day 1 and etoposide (80-100 mg/m²) intravenously on Days 1, 2, and 3 of each 21-day cycle for between 4–6 cycles.

For patients randomised to Arm 1 and 2, etoposide and either carboplatin or cisplatin was limited to 4 cycles on an every 3-week schedule subsequent to randomisation. IMFINZI monotherapy continued every 4 weeks until disease progression or unacceptable toxicity. Administration of IMFINZI monotherapy was permitted beyond disease progression if the patient was clinically stable and deriving clinical benefit as determined by the investigator.

Patients randomised to Arm 3 were permitted to receive a total of up to 6 cycles of etoposide and either carboplatin or cisplatin. After completion of etoposide + platinum, PCI was permitted only in Arm 3 per investigator discretion.

Tumour assessments were conducted at Week 6 and Week 12 from the date of randomisation, and then every 8 weeks until confirmed objective disease progression. Survival assessments were conducted every 2 months following treatment discontinuation.

The primary endpoints of the study were OS of IMFINZI + etoposide + platinum (Arm 2) vs. etoposide + platinum alone (Arm 3) and IMFINZI + tremelimumab + etoposide + platinum (Arm 1) vs. etoposide + platinum alone (Arm 3). The key secondary endpoint was PFS. Other secondary endpoints were ORR, OS and PFS landmarks and PRO. PFS and ORR were assessed using Investigator assessments according to RECIST v1.1.

The demographics and baseline disease characteristics were well balanced between the two study arms (268 patients in Arm 2 and 269 patients in Arm 3). Baseline demographics of the overall study population were as follows: male (69.6%), age ≥ 65 years (39.6%), median age 63 years (range: 28 to 82 years), white (83.8%), Asian (14.5%), Black or African American (0.9%), other (0.6%), non-Hispanic or Latino (96.1%), current or past-smoker (93.1%), never smoker (6.9%), WHO/ECOG PS 0 (35.2%), WHO/ECOG PS 1 (64.8%), Stage IV 90.3%, 24.6% of the patients received cisplatin and 74.1% of the patients received carboplatin. In Arm 3, 56.8% of the patients received 6 cycles of etoposide + platinum and 7.8% of the patients received PCI.

At a planned interim (primary) analysis the study demonstrated a statistically significant improvement in OS with IMFINZI + etoposide + platinum (Arm 2) vs. etoposide + platinum alone (Arm 3) [HR=0.73 (95% CI: 0.591, 0.909), p=0.0047]. Although not formally tested for significance, IMFINZI + etoposide + platinum demonstrated an improvement in PFS vs. etoposide + platinum alone [HR=0.78 (95% CI: 0.645, 0.936)].

The PFS, ORR and DoR results from the planned final analysis (DCO: 27 Jan 2020) are summarized in Table 7. Kaplan-Meier curve for PFS is presented in Figure 11.

The OS results with the planned long-term OS follow-up analysis (DCO: 22 March 2021) (median follow-up: 39.3 months) are presented in Table 7. IMFINZI + etoposide + platinum (Arm 2) vs. etoposide + platinum (Arm 3) continued to demonstrate sustained improvement in OS. Kaplan-Meier curve for OS is presented in Figure 10.

Table 7. Efficacy Results for the CASPIAN Study:

 Final analysisa Long-term follow-up analysisb
Arm 2:
IMFINZI +
etoposide and
either
carboplatin or
cisplatin
(n=268)
Arm 3:
etoposide + and
either
carboplatin or
cisplatin
(n=269)
Arm 2:
IMFINZI +
etoposide
and either
carboplatin
or cisplatin
(n=268)
Arm 3:
etoposide +
and either
carboplatin or
cisplatin
(n=269)
OS
Number of deaths (%) 210 (78.4) 231 (85.9) 221 (82.5) 248 (92.2)
Median OS (months)
(95% CI)
12.9
(11.3, 14.7)
10.5
(9.3, 11.2)
12.9
(11.3, 14.7)
10.5
(9.3, 11.2)
HR (95% CI)b,c 0.75 (0.625, 0.910) 0.71 (0.595, 0.858)
p-valued 0.00320.0003
OS at 18 months (%)
(95% CI)
32.0
(26.5, 37.7)
24.8
(19.7, 30.1)
32.0
(26.5, 37.7)
24.8
(19.7, 30.1)
OS at 36 months (%)
(95% CI)
  17.6
(13.3, 22.4)
5.8
(3.4, 9.1)
PFS
Number of events (%) 234 (87.3) 236 (87.7)   
Median PFS (months)
(95% CI)
5.1
(4.7, 6.2)
5.4
(4.8, 6.2)
  
HR (95% CI)c 0.80 (0.665, 0.959)  
PFS at 6 months (%)
(95% CI)
45.4
(39.3, 51.3)
45.8
(39.5, 51.9)
  
PFS at 12 months
(%) (95% CI)
17.9
(13.5, 22.8)
5.3
(2.9, 8.8)
  
ORR n (%)
(95% CI)e
182 (67.9)
(62.0, 73.5)
156 (58.0)
(51.8, 64.0)
  
Complete Response n
(%)
7 (2.6) 2 (0.7)   
Partial Response n
(%)
175 (65.3) 154 (57.2)   
Median DoR (months)
(95% CI)e,f
5.1
(4.9, 5.3)
5.1
(4.8, 5.3)
  

a Final PFS, ORR and DoR analysis at data cut-off 27 January 2020.
b Long-term follow-up OS analysis at data cut-off 22 March 2021.
c The analysis was performed using the stratified log-rank test, adjusting for planned platinum therapy in Cycle 1 (carboplatin or cisplatin), and using the rank tests of association approach.
d At the interim analysis (data cut-off 11 March 2019) the OS p-value was 0.0047, which met the boundary for declaring statistical significance of 0.0178 for a 4% overall 2-sided alpha, based on a Lan-DeMets alpha spending function with O’Brien Fleming type boundary with the actual number of events observed.
e Confirmed Objective Response.
f Post-hoc analysis.

Figure 10. Kaplan-Meier curve of OS:

Figure 11. Kaplan-Meier curve of PFS:

Subgroup analysis

The improvements in OS in favour of patients receiving IMFINZI + etoposide + platinum compared to those receiving etoposide + platinum alone, were consistently observed across the prespecified subgroups based on demographics, geographical region, carboplatin or cisplatin use and disease characteristics.

BTC – TOPAZ-1 Study

TOPAZ-1 was a study designed to evaluate the efficacy of IMFINZI in combination with gemcitabine and cisplatin. TOPAZ-1 was a randomised, double-blind, placebo-controlled, multicentre study in 685 patients with unresectable or metastatic BTC (including intrahepatic and extrahepatic cholangiocarcinoma and gallbladder carcinoma) and ECOG Performance status of 0 or 1. Patients had not received previous therapy in the advanced/unresectable setting. Patients who developed recurrent disease >6 months after surgery and/or completion of adjuvant therapy were included. Patients must have had an adequate organ and bone marrow function, and have had acceptable serum bilirubin levels (≤2.0 x the upper limit of normal (ULN)), and any clinically significant biliary obstruction had to be resolved before randomisation.

The study excluded patients with ampullary carcinoma, with brain metastases, active or prior documented autoimmune or inflammatory disorders, HIV infection or active infections, including tuberculosis or hepatitis C or patients with current or prior use of immunosuppressive medication within 14 days before the first dose of IMFINZI. Patients with active HBV were allowed to participate if they were on antiviral therapy.

Randomisation was stratified by disease status (initially unresectable vs. recurrent) and primary tumour location (intrahepatic cholangiocarcinoma vs. extrahepatic cholangiocarcinoma vs. gallbladder carcinoma).

Patients were randomised 1:1 to receive:

  • Arm 1: IMFINZI 1 500 mg administered on Day 1 + gemcitabine 1 000 mg/m² and cisplatin 25 mg/m² (each administered on Days 1 and 8) every 3 weeks (21 days) for up to 8 cycles, followed by IMFINZI 1 500 mg every 4 weeks until disease progression or unacceptable toxicity, or
  • Arm 2: Placebo administered on Day 1 + gemcitabine 1 000 mg/m² and cisplatin 25 mg/m² (each administered on Days 1 and 8) every 3 weeks (21 days) for up to 8 cycles, followed by placebo every 4 weeks until disease progression or unacceptable toxicity.

Tumour assessments were conducted every 6 weeks for the first 24 weeks after the date of randomisation, and then every 8 weeks until confirmed objective disease progression.

The primary endpoint of the study was OS, the key secondary endpoint was PFS. Other secondary endpoints were ORR, DoR and PRO. PFS, ORR and DoR were investigator-assessed according to RECIST v1.1.

The demographics and baseline disease characteristics were well balanced between the two study arms (341 patients in Arm 1 and 344 patients in Arm 2). Baseline demographics of the overall study population were as follows: male (50.4%), age <65 years (53.3%), white (37.2%), Asian (56.4%), Black or African American (2.0%), other (4.2%), non-Hispanic or Latino (93.1%), ECOG PS 0 (49.1%), vs. PS 1 (50.9%), primary tumour location (intrahepatic bile duct 55.9%, extrahepatic bile duct 19.1% and gallbladder 25.0%), disease status [recurrent (19.1%) vs. unresectable (80.7%), metastatic (86.0%) vs. locally advanced (13.9%)]. PD-L1 expression was evaluated on tumour and immune cells using the Ventana PD-L1 (SP263) assay and the TAP (tumour area positivity) algorithm, 58.7% patients had TAP ≥1% and 30.1% TAP <1%.

OS and PFS were formally tested at a pre-planned interim analysis (data cut-off 11 Aug 2021) after a median follow-up of 9.8 months. Efficacy results are shown in Table 8 and Figure 13. The maturity for OS was 62% and the maturity for PFS was 84%. IMFINZI + chemotherapy (Arm 1) showed statistically significant improvement vs. placebo + chemotherapy (Arm 2) in OS and in PFS.

Table 8. Efficacy Results for the TOPAZ-1 Studya:

 IMFINZI + gemcitabine
and cisplatin
(n=341)
Placebo + gemcitabine and
cisplatin
(n=344)
OS   
Number of deaths (%) 198 (58.1) 226 (65.7)
Median OS (months)
(95% CI)b
12.8
(11.1, 14.0)
11.5
(10.1, 12.5)
HR (95% CI)c 0.80 (0.66, 0.97)
p-valuec,d 0.021
Median follow-up in all patients
(months)
10.2 9.5
PFS   
Number of events (%) 276 (80.9) 297 (86.3)
Median PFS (months)
(95% CI)b
7.2
(6.7, 7.4)
5.7
(5.6, 6.7)
HR (95% CI)c 0.75 (0.63, 0.89)
p-valuec,e 0.001
Median follow-up in all patients
(months)
7.2 5.6
ORRf 91 (26.7) 64 (18.7)
Complete Response n (%) 7 (2.1) 2 (0.6)
Partial Response n (%) 84 (24.6) 62 (18.1)
DoR   
Median DoR (months)
(95% CI)b
6.4 (5.9, 8.1) 6.2 (4.4, 7.3)

a Analysis at data cut-off 11 August 2021.
b Calculated using the Kaplan-Meier technique. CI for median derived based on Brookmeyer-Crowley method.
c The analysis for HR was performed using a stratified Cox proportional hazards model and 2-sided p-value is based on a stratified log-rank test, both are adjusted for disease status and primary tumour location.
d At the interim analysis (data cut-off 11 August 2021) the OS p-value was 0.021, which met the boundary for declaring statistical significance of 0.03 for a 4.9% overall 2-sided alpha, based on a Lan-DeMets alpha spending function with O’Brien Fleming type boundary with the actual number of events observed.
e At the interim analysis (data cut-off 11 August 2021) the PFS p-value was 0.001, which met the boundary for declaring statistical significance of 0.0481 for a 4.9% overall 2-sided alpha, based on a Lan-DeMets alpha spending function with Pocock-type boundary with the actual number of events observed.
f Confirmed objective response.

An additional planned follow-up analysis of OS (data cut-off 25 Feb 2022) was performed 6.5 months after the interim analysis with an OS maturity of 77%. IMFINZI + chemotherapy continued to demonstrate improved OS vs. chemotherapy alone [HR=0.76, (95% CI: 0.64, 0.91)] and the median follow-up increased to 12 months.

Figure 12. Kaplan-Meier curve of OS, follow-up OS analysis at data cut-off 25 February 2022:

Figure 13. Kaplan-Meier curve of PFS, inferential (primary) analysis at data cut-off 11 August 2021:

HCC – HIMALAYA Study

The efficacy of IMFINZI as monotherapy and given in combination with a single dose of tremelimumab 300 mg was evaluated in the HIMALAYA Study, a randomised, open-label, multicentre study in patients with confirmed uHCC who did not receive prior systemic treatment for HCC. The study included patients with Barcelona Clinic Liver Cancer (BCLC) Stage C or B (not eligible for locoregional therapy) and Child-Pugh Score Class A.

The study excluded patients with brain metastases or a history of brain metastases, co-infection of viral hepatitis B and hepatitis C; active or prior documented gastrointestinal (GI) bleeding within 12 months; ascites requiring non-pharmacologic intervention within 6 months; hepatic encephalopathy within 12 months before the start of treatment; active or prior documented autoimmune or inflammatory disorders.

Patients with oesophageal varices were included except those with active or prior documented GI bleeding within 12 months prior to study entry.

Randomisation was stratified by macrovascular invasion (MVI) (yes vs. no), aetiology of liver disease (confirmed hepatitis B virus vs. confirmed hepatitis C virus vs. others) and ECOG performance status (0 vs. 1). The HIMALAYA study randomized 1171 patients 1:1:1 to receive:

  • IMFINZI: durvalumab 1 500 mg every 4 weeks.
  • Tremelimumab 300 mg as a single dose + IMFINZI 1 500 mg; followed by IMFINZI 1 500 mg every 4 weeks.
  • Sorafenib 400 mg twice daily.

Tumour assessments were conducted every 8 weeks for the first 12 months and then every 12 weeks thereafter. Survival assessments were conducted every month for the first 3 months following treatment discontinuation and then every 2 months.

The primary endpoint was OS superiority for the comparison of IMFINZI given in combination with a single dose of tremelimumab vs. Sorafenib. The key secondary objectives were OS non-inferiority followed by superiority for the comparison of IMFINZI vs. Sorafenib. Other secondary endpoints included PFS, Investigator-assessed ORR and DoR according to RECIST v1.1.

The demographics and baseline disease characteristics were well balanced among study arms. The baseline demographics of the overall study population were as follows: male (83.7%), age <65 years (50.4%) White (44.6%), Asian (50.7%), Black or African American (1.7%), Other race (2.3%), ECOG PS 0 (62.6%); Child-Pugh Class score A (99.5%), macrovascular invasion (25.2%), extrahepatic spread (53.4%), baseline AFP < 400 ng/ml (63.7%), baseline AFP ≥400 ng/ml (34.5%), viral aetiology; hepatitis B (30.6%), hepatitis C (27.2%), uninfected (42.2%), evaluable PD-L1 data (86.3%), PD-L1 Tumour area positivity (TAP) ≥1% (38.9%), PD-L1 TAP <1% (48.3%) [Ventana PD-L1 (SP263) assay].

Results are presented in Table 9, Figure 14 and Figure 15.

Table 9. Efficacy Results for the HIMALAYA Study for IMFINZI given in combination with a single dose of tremelimumab 300 mg and IMFINZI as monotherapy vs. Sorafenib:

 IMFINZI +
tremelimumab
300 mg
(n=393)
Sorafenib
(n=389)
IMFINZI
(n=389)
Follow up duration
Median follow up (months)a 33.2 32.2 32.6
OS
Number of deaths (%) 262 (66.7) 293 (75.3) 280 (72.0)
Median OS (months)
(95% CI)
16.4
(14.2, 19.6)
13.8
(12.3, 16.1)
16.6
(14.1, 19.1)
HR (95% CI)b,c 0.78 (0.66, 0.92) -
p-valued 0.0035-
HR (95% CI)b,c,e - 0.86 (0.73, 1.03)
PFS
Number of events (%) 335 (85.2) 327 (84.1) 345 (88.7)
Median PFS (months) (95% CI) 3.78
(3.68-5.32)
4.07
(3.75-5.49)
3.65
(3.19-3.75)
HR (95% CI) 0.90 (0.77, 1.05) -
HR (95% CI) - 1.02 (0.88, 1.19)
ORR
ORR n (%) f 79 (20.1) 20 (5.1) 66 (17.0)
Complete Response n (%) 12 (3.1) 0 6 (1.5)
Partial Response n (%) 67 (17.0) 20 (5.1) 60 (15.4)
DoR
Median DoR (months) 22.3 18.4 16.8

a Calculated using reverse the Kaplan-Meier technique (with censor indicator reversed).
b Based on stratified Cox-model adjusting for treatment, etiology of liver disease (HBV versus HCV versus others), ECOG (0 versus 1).
c Performed using stratified log-rank test adjusting for treatment, etiology of liver disease (HBV versus HCV versus others), ECOG (0 versus 1), and macro-vascular invasion (yes versus no).
d Based on a Lan-DeMets alpha spending function with O’Brien Fleming type boundary and the actual number of events observed, the boundary for declaring statistical significance for IMFINZI + tremelimumab 300 mg vs. Sorafenib was 0.0398 (Lan◦and◦DeMets 1983).
e Non-inferiority margin for HR (IMFINZI vs Sorafenib) is 1.08 using a 95.67% confidence interval based on a Lan-DeMets alpha spending function with O’Brien Fleming type boundary and the actual number of events observed (Lan◦and◦DeMets 1983). P-value based on superiority testing of IMFINZI vs. Sorafenib was 0.0674 and did not reach statistical significance.
f Confirmed complete response.
CI=Confidence Interval

Figure 14. Kaplan-Meier curve of OS of IMFINI given in combination with a single dose of tremelimumab 300 mg:

Figure 15. Kaplan-Meier curve of OS of IMFINZI given as monotherapy:

Paediatric population

The safety and efficacy of IMFINZI in combination with tremelimumab in children and adolescents aged less than 18 years has not been established. Study D419EC00001 was a multi-centre, open-label dose finding and dose expansion study to evaluate the safety, preliminary efficacy and pharmacokinetics of IMFINZI in combination with tremelimumab followed by IMFINZI monotherapy, in paediatric patients with advanced malignant solid tumours (except primary central nervous system tumours) who had disease progression and for whom no standard of care treatment exists. The study enrolled 50 paediatric patients with an age range from 1 to 17 years with primary tumour categories: neuroblastoma, solid tumour and sarcoma. Patients received either IMFINZI 20 mg/kg in combination with tremelimumab 1 mg/kg or IMFINZI 30 mg/kg in combination with tremelimumab 1 mg/kg intravenously every 4 weeks for 4 cycles, followed by IMFINZI as monotherapy every 4 weeks. In the dose finding phase, IMFINZI and tremelimumab combination therapy was preceded by a single cycle of IMFINZI monotherapy; 8 patients in this phase however discontinued treatment prior to receiving tremelimumab. Thus, of the 50 patients enrolled in the study, 42 received IMFINZI in combination with tremelimumab and 8 received IMFINZI only. In the doseexpansion phase, an ORR of 5.0% (1/20 patients) was reported in the evaluable for response analysis set. No new safety signals were observed relative to the known safety profiles of IMFINZI and tremelimumab in adults. See section 4.2 for information on paediatric use.

Pharmacokinetic properties

The pharmacokinetics (PK) of durvalumab was assessed for IMFINZI as a single agent, in combination with chemotherapy, in combination with tremelimumab and platinum-based chemotherapy and in combination with tremelimumab.

The PK of durvalumab was studied in 2903 patients with solid tumours with doses ranging from 0.1 to 20 mg/kg administered intravenously once every two, three or four weeks as monotherapy. PK exposure increased more than dose-proportionally (non-linear PK) at doses <3 mg/kg, and dose proportionally (linear PK) at doses ≥3 mg/kg. Steady state was achieved at approximately 16 weeks. Based on population PK analysis that included 1878 patients who received durvalumab monotherapy in the dose range of ≥10 mg/kg every 2 weeks, the geometric mean steady state volume of distribution (Vss) was 5.64 L. Durvalumab clearance (CL) decreased over time resulting in a geometric mean steady state clearance (CLss) of 8.16 ml/h at Day 365; the decrease in CLss was not considered clinically relevant. The terminal half-life (t1/2), based on baseline CL, was approximately 18 days. There was no clinically meaningful difference between the PK of durvalumab as a single agent, in combination with chemotherapy, in combination with tremelimumab and platinum-based chemotherapy and in combination with tremelimumab. The primary elimination pathways of durvalumab are protein catabolism via reticuloendothelial system or target mediated disposition.

Special populations

Age (19-96 years), body weight (31-149 kg), gender, positive anti-drug antibody (ADA) status, albumin levels, LDH levels, creatinine levels, soluble PD-L1, tumour type, race or ECOG status had no clinically significant effect on the PK of durvalumab.

Renal impairment

Mild (creatinine clearance (CrCL) 60 to 89 ml/min) and moderate renal impairment (creatinine clearance (CrCL) 30 to 59 ml/min) had no clinically significant effect on the PK of durvalumab. The effect of severe renal impairment (CrCL 15 to 29 ml/min) on the PK of durvalumab is unknown; however, as IgG monoclonal antibodies are not primarily cleared via renal pathways, a change in renal function is not expected to influence durvalumab exposure.

Hepatic impairment

Mild hepatic impairment (bilirubin ≤ ULN and AST > ULN or bilirubin >1.0 to 1.5 x ULN and any AST) or moderate hepatic impairment (bilirubin >1.5 to 3 x ULN and any AST) had no clinically significant effect on the PK of durvalumab. The effect of severe hepatic impairment (bilirubin >3.0 x ULN and any AST) on the pharmacokinetics of durvalumab is unknown; however, as IgG monoclonal antibodies are not primarily cleared via hepatic pathways, a change in hepatic function is not expected to influence durvalumab exposure.

Paediatric population

The PK of durvalumab in combination with tremelimumab was evaluated in a study of 50 paediatric patients with an age range from 1 to 17 years in study D419EC00001. Patients received either durvalumab 20 mg/kg in combination with tremelimumab 1 mg/kg or durvalumab 30 mg/kg in combination with tremelimumab 1 mg/kg intravenously every 4 weeks for 4 cycles, followed by durvalumab as monotherapy every 4 weeks. Based on population PK analysis, durvalumab systemic exposure in paediatric patients ≥35kg receiving durvalumab 20 mg/kg every 4 weeks was similar to exposure in adults receiving durvalumab 20 mg/kg every 4 weeks, whereas in paediatric patients (≥35kg) receiving durvalumab 30mg/kg every 4 weeks, exposure was approximately 1.5 fold higher compared to exposure in adults receiving durvalumab 20 mg/kg every 4 weeks. In paediatric patients <35kg receiving durvalumab 30 mg/kg every 4 weeks, the systemic exposure was similar to exposure in adults receiving durvalumab 20 mg/kg every 4 weeks.

Preclinical safety data

Carcinogenicity and mutagenicity

The carcinogenic and genotoxic potential of durvalumab has not been evaluated.

Reproductive toxicology

As reported in the literature, the PD-1/PD-L1 pathway plays a central role in preserving pregnancy by maintaining maternal immune tolerance to the foetus, and in mouse allogeneic pregnancy models disruption of PD-L1 signalling was shown to result in an increase in foetal loss. In animal reproduction studies, administration of durvalumab to pregnant cynomolgus monkeys from the confirmation of pregnancy through delivery, at exposure levels approximately 18-times higher than those observed at the clinical dose of 10 mg/kg of durvalumab (based on AUC), was associated with placental transfer but not with maternal toxicity or effects on embryofoetal development, pregnancy outcome or postnatal development. Negligible levels of durvalumab was found in milk of cynomolgous monkey on Day 28 after birth.

© All content on this website, including data entry, data processing, decision support tools, "RxReasoner" logo and graphics, is the intellectual property of RxReasoner and is protected by copyright laws. Unauthorized reproduction or distribution of any part of this content without explicit written permission from RxReasoner is strictly prohibited. Any third-party content used on this site is acknowledged and utilized under fair use principles.