INQOVI Film-coated tablet Ref.[51655] Active ingredients: Cedazuridine Decitabine Decitabine and Cedazuridine

Source: FDA, National Drug Code (US)  Revision Year: 2022 

12.1. Mechanism of Action

Decitabine is a nucleoside metabolic inhibitor that is believed to exert its effects after phosphorylation and direct incorporation into DNA and inhibition of DNA methyltransferase, causing hypomethylation of DNA and cellular differentiation and/or apoptosis. Decitabine inhibits DNA methylation in vitro, which is achieved at concentrations that do not cause major suppression of DNA synthesis. Decitabine-induced hypomethylation in cancer cells may restore normal function to genes that are critical for the control of cellular differentiation and proliferation. In rapidly dividing cells, the cytotoxicity of decitabine may also be attributed to the formation of covalent adducts between DNA methyltransferase and decitabine incorporated into DNA. Non-proliferating cells are relatively insensitive to decitabine.

Cytidine deaminase (CDA) is an enzyme that catalyzes the degradation of cytidine, including the cytidine analog decitabine. High levels of CDA in the gastrointestinal tract and liver degrade decitabine and limit its oral bioavailability. Cedazuridine is a CDA inhibitor. Administration of cedazuridine with decitabine increases systemic exposure of decitabine.

12.2. Pharmacodynamics

Decitabine induced hypomethylation both in vitro and in vivo. In patients administered the recommended dosage of INQOVI, the maximum change from baseline in the long interspersed nucleotide elements-1 (LINE-1) demethylation was observed at Day 8, with less than complete recovery of LINE-1 methylation to baseline at the end of the treatment cycle.

Based on the exposure-response analyses, a relationship between an increase in 5-day cumulative daily decitabine exposure and a greater likelihood of some adverse reactions (e.g., any grade neutropenias, thrombocytopenia) was observed in clinical studies.

12.3. Pharmacokinetics

The pharmacokinetics of decitabine and cedazuridine following administration of INQOVI at the recommended dosage in patients with MDS and CMML are shown in Table 4.

The geometric mean ratio (GMR) of decitabine area under the curve (AUC) following the first dose of INQOVI compared to that of intravenous decitabine on Day 1 was 60% (90% confidence intervals (CI): 55, 65) in patients with MDS and CMML [see Dosage and Administration (2.1)]. The GMR of decitabine AUC following 5 consecutive once daily doses of INQOVI compared to that of intravenous decitabine on Day 5 was 106% (90% CI: 98, 114) and the GMR of the 5-day cumulative decitabine AUC following 5 consecutive once daily doses of INQOVI compared to that of intravenous decitabine was 99% (90% CI: 93, 106).

An approximately dose-proportional increase in peak concentrations (Cmax) and AUC over the dosing interval was observed for decitabine following administration of oral decitabine at 20 mg to 40 mg once daily (0.6 to 1.1 times the recommended dose) in combination with 100 mg oral cedazuridine, and for cedazuridine following administration of oral cedazuridine at 40 to 100 mg once daily (0.4 to 1.0 times the recommended dose) in combination with 20 mg oral decitabine.

Table 4. Pharmacokinetics of the Components of INQOVI*:

Parameter Decitabine Cedazuridine
General Information
With the recommended dosage of INQOVI for 5 consecutive days:
5-day cumulative AUC, ng.hr/mL 851 (50%) --
Day 1 AUC, ng·hr/mL 103 (55%) 2950 (49%)
Steady state AUC, ng·hr/mL 178 (53%) 3291 (45%)
Time to steady state, days 2 2
Accumulation ratio based on AUC 1.7 (42%) 1.1 (63%)
Cmax, ng/mL 145 (55%) 371 (52%)
Absorption
Bioavailability Cedazuridine
increases oral
decitabine exposure
20% (23%)
Tmax, hours 1 (0.3 to 3.0) 3 (1.5 to 6.1)
Distribution
V/F at steady state, L 417 (54%) 296 (51%)
Fraction unbound, in vitro 96% (4%) to 94% (2%)
between 17 ng/mL
to 342 ng/mL
66% (6%) to 62% (2%)
between 1000 ng/mL
and 50000 ng/mL
Elimination
Half-life at
steady state, hours
1.5 (27%) 6.7 (19%)
CL/F at steady state,
L/hours
197 (53%) 30.3 (46%)
Metabolism
Primary Pathways Primarily by cytidine
deaminase (CDA) and
by physicochemical
degradation
Conversion to epimer
by physicochemical
degradation
Excretion§
Total (% unchanged) -- 46% (21%) in urine
and 51% (27%)
in feces

Cmax= maximum plasma concentration; AUC0-24h=area under the plasma concentration-time curve from time zero to 24 hours; CV=coefficient of variation; SD=standard deviation; Tmax= Time to maximum concentration; V/F=apparent volume of distribution; CL/F=apparent clearance
* Mean (%CV)
Median (range)
Mean (SD)
§ Healthy subjects

Specific Populations

Age (32 to 90 years), sex, and mild hepatic impairment (total bilirubin >1 to 1.5 × ULN or AST > ULN) did not have an effect on the pharmacokinetics of decitabine or cedazuridine after dosing with INQOVI.

Decitabine exposure (AUC) increased with decreasing body surface area or body weight, and cedazuridine exposure increased with decreasing CLcr; however, body surface area (1.3 to 2.9 m²), body weight (41 to 158 kg), and mild to moderate renal impairment (CLcr 30 to 89 mL/min based on Cockcroft Gault) did not have a clinically meaningful effect on the pharmacokinetics of decitabine and cedazuridine after dosing with INQOVI.

The effects of moderate (total bilirubin >1.5 to 3 × ULN and any AST) and severe hepatic impairment (total bilirubin >3 × ULN and any AST) or severe renal impairment (CLcr 15 to <30 mL/min) and ESRD (CLcr <15 mL/min) on the pharmacokinetics of decitabine and cedazuridine are unknown.

Drug Interaction Studies

Clinical Studies

Decitabine had no clinically meaningful effect on the pharmacokinetics of cedazuridine. Cedazuridine increased the exposure of decitabine.

The coadministration of INQOVI with proton pump inhibitors had no clinically meaningful effect on exposure to decitabine or cedazuridine.

In vitro Studies

CYP Enzymes: Cedazuridine is not a substrate of cytochrome P450 (CYP) enzymes. Cedazuridine does not induce CYP1A, CYP2B6, CYP2C9, or CYP3A or inhibit CYP1A, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, or CYP3A.

Transporter Systems: Cedazuridine is not a substrate of P-glycoprotein (P-gp), MATE1, MATE2-K, OAT1, OAT3, OATP1B1, OAPT1B3, OATP2B1, OCT1, or OCT2, and does not inhibit P-gp, BCRP, MATE1, MATE2-K, OAT1, OAT3, OATP1B1, OATP1B3, or OCT2.

13.1. Carcinogenesis, Mutagenesis, Impairment of Fertility

Carcinogenicity studies with decitabine, cedazuridine, or their combination have not been conducted.

INQOVI is genotoxic. Decitabine increased mutation frequency in L5178Y mouse lymphoma cells, and mutations were produced in an E. coli lac-I transgene in colonic DNA of decitabine-treated mice. Decitabine also caused chromosomal rearrangements in larvae of fruit flies. Cedazuridine was genotoxic in a reverse bacterial mutation assay (Ames assay) and in an in vitro chromosomal aberration study using human lymphocytes.

Fertility and repeat-dose toxicity studies in animals showed adverse outcomes on reproductive function and fertility. In male mice given intraperitoneal injections of 0.15, 0.3, or 0.45 mg/m² decitabine (approximately 0.3% to 1% the recommended clinical dose) 3 times a week for 7 weeks, testes weights were reduced, abnormal histology was observed, and significant decreases in sperm number were found at doses ≥0.3 mg/m². In females mated to males dosed with ≥0.3 mg/m² decitabine, pregnancy rate was reduced, and preimplantation loss was significantly increased.

Decitabine was administered orally to rats at 0.75, 2.5, or 7.5 mg/kg/day in cycles of 5-days-on/23-days-off for a total of 90 days. Low testes and epididymis weights, abnormal histology, and reduced sperm number were observed at doses ≥0.75 mg/kg. The dose of 0.75 mg/kg resulted in exposures in animals that were approximately 3 times the exposure in patients at the recommended clinical dose based on AUC.

Cedazuridine was administered orally to mice at 100, 300, or 1,000 mg/kg/day in cycles of 7-days-on/21-days-off for a total of 91 days. Adverse findings in male and female reproductive organs were observed at the 1,000 mg/kg dose and included abnormal histology in the testes and epididymis, reduced sperm number, and abnormal histology in the ovary. The dose of 1,000 mg/kg/day resulted in exposures in animals that were approximately 108 times the exposure in patients at the recommended clinical dose. Adverse effects in male and female reproductive organs were reversible following a recovery period.

14. Clinical Studies

Study ASTX727-01-B

INQOVI was evaluated in Study ASTX727-01-B, an open-label, randomized, 2-cycle, 2-sequence crossover study (NCT02103478) that included 80 adult patients with MDS (International Prognostic Scoring System [IPSS] Intermediate-1, Intermediate-2, or high-risk) or CMML. Patients were randomized 1:1 to receive INQOVI (35 mg decitabine and 100 mg cedazuridine) orally in Cycle 1 and decitabine 20 mg/m 2 intravenously in Cycle 2 or the reverse sequence. Both INQOVI and intravenous decitabine were administered once daily on Days 1 through 5 of the 28-day cycle. Starting with Cycle 3, all patients received INQOVI orally once daily on Days 1 through 5 of each 28-day cycle until disease progression or unacceptable toxicity. Randomization was stratified by IPSS risk level. Twelve (15%) of the 80 patients went on to stem cell transplantation following INQOVI treatment.

The baseline demographic and disease characteristics are shown in Table 5.

Table 5. Demographics and Baseline Disease Characteristics for Study ASTX727-01-B:

Characteristic N=80
Age
Median (min, max) (years) 71 (32, 90)
Sex (%)
Male 76
Female 24
Race (%)
White 93
Black or African American 3
Asian 1
Other or Not Reported 4
ECOG Performance Score (%)
0 44
1 48
2 9
Disease Category / IPSS (%)
MDS INT-1 44
MDS INT-2 24
MDS High-Risk 11
CMML 21
Prior HMA Therapy* (%)
Prior Azacitidine 4
Prior Decitabine 4
Transfusion Dependence (%)
RBC Transfusion Dependence 48
Platelet Transfusion Dependence 15

* One cycle only, per the Exclusion Criteria.
Defined as documentation of ≥ 2 units of transfusion within 56 days prior to the first day of study treatment.

Efficacy was established on the basis of complete response (CR) and the rate of conversion from transfusion dependence to transfusion independence. Efficacy results are shown in Table 6. The median follow-up time was 24.0 months (range: 12.0 to 28.8 months) and median treatment duration was 6.6 months (range <0.1 to 27.9).

Table 6. Efficacy Results in Patients with MDS or CMML from Study ASTX727-01-B:

Efficacy Endpoint INQOVI
N=80
Complete Response () (95 CI) 18 (10, 28)
Median Duration of CR – months (range)* 8.7 (1.1, 18.2)
Median Time to CR – months (range) 4.8 (1.7, 10.0)

Among the 41 patients who were dependent on red blood cell (RBC) and/or platelet transfusions at baseline, 20 (49%) became independent of RBC and platelet transfusions during any consecutive 56-day post-baseline period. Of the 39 patients who were independent of both RBC and platelet transfusions at baseline, 25 (64%) remained transfusion-independent during any consecutive 56-day post-baseline period.

Study ASTX727-02

INQOVI was evaluated in ASTX727-02, an open-label, randomized, 2-cycle, 2-sequence crossover study (NCT03306264) that included 133 adult patients with MDS or CMML, including all French-American-British (FAB) classification criteria and IPSS Intermediate-1, Intermediate-2, or high-risk prognostic scores. Patients were randomized 1:1 to receive INQOVI (35 mg decitabine and 100 mg cedazuridine) orally in Cycle 1 and decitabine 20 mg/m 2 intravenously in Cycle 2 or the reverse sequence. Both INQOVI and intravenous decitabine were administered once daily on Days 1 through 5 of the 28-day cycle. Starting with Cycle 3, all patients received INQOVI orally once daily on Days 1 through 5 of each 28-day cycle until disease progression or unacceptable toxicity. No stratification was performed. Twenty-seven (20%) of the 133 patients went on to stem cell transplantation following INQOVI treatment.

The baseline demographic and disease characteristics are shown in Table 7.

Table 7. Demographics and Baseline Disease Characteristics for Study ASTX727-02:

Characteristic N=133
Age (years)
Median (min, max) 71 (44, 88)
Sex (%)
Male 65
Female 35
Race (%)
White 91
Black or African American 3
Asian 2
Other or Not Reported 4
ECOG Performance Score (%)
0 41
1 59
Disease Category / IPSS (%)
MDS INT-1 44
MDS INT-2 20
MDS High Risk 16
MDS Low Risk 8
CMML 12
Prior HMA Therapy* (%)
Prior Azacitidine 5
Prior Decitabine 3
Transfusion Dependence (%)
RBC Transfusion Dependence 39
Platelet Transfusion Dependence 8

* One cycle only, per the Exclusion Criteria.
Defined as documentation of ≥2 units of transfusion within 56 days prior to the first day of study treatment.

The primary outcome measure was comparison of the 5-day cumulative decitabine AUC between INQOVI and intravenous decitabine [see Clinical Pharmacology (12.3)]. Efficacy was established on the basis of complete response (CR) and the rate of conversion from transfusion dependence to transfusion independence. Efficacy results are shown in Table 8. The median follow-up time was 12.6 months (range: 9.3 to 20.5) and median treatment duration was 8.2 months (range 0.2 to 19.7).

Table 8. Efficacy Results in Patients with MDS or CMML from Study ASTX727-02:

Efficacy Endpoints INQOVI
(N=133)
Complete Response () (95 CI) 21 (15, 29)
Median Duration of CR – months (range)* 7.5 (1.6, 17.5)
Median Time to CR – months (range) 4.3 (2.1, 15.2)

* From start of CR until relapse or death.

Among the 57 patients who were dependent on RBC and/or platelet transfusions at baseline, 30 (53%) became independent of RBC and platelet transfusions during any 56-day post-baseline period. Of the 76 patients who were independent of both RBC and platelet transfusions at baseline, 48 (63%) remained transfusion-independent during any 56-day post-baseline period.

© 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.