Pharmacology: Pharmacodynamics: Mechanism of Action: Tofacitinib is a potent, selective inhibitor of the Janus-associated kinase (JAK) family of kinases with a high degree of selectivity against other kinases in the human genome. In kinase assays, tofacitinib inhibits JAK1, JAK2, JAK3 and to a lesser extent TyK2. In cellular settings where JAK kinases signal in pairs, tofacitinib preferentially inhibits signaling by heterodimeric receptors associated with JAK3 and/or JAK1 with functional selectivity over receptors that signal via pairs of JAK2. Inhibition of JAK1 and JAK3 by tofacitinib blocks signaling through the common gamma (γ) chain-containing receptors for several cytokines, including interleukin (IL)-2, -4, -7, -9, -15 and -21. These cytokines are integral to lymphocyte activation, proliferation and function, and inhibition of their signaling may thus result in modulation of multiple aspects of the immune response. In addition, inhibition of JAK1 will result in attenuation of signaling by additional pro-inflammatory cytokines eg, IL-6 and type I interferons. At higher exposures, inhibition of erythropoietin signaling could occur via inhibition of JAK2 signaling.
Pharmacodynamic Effects: Treatment with Xeljanz was associated with dose-dependent reductions of circulating CD16/56+ natural killer cells, with estimated maximum reductions occurring at approximately 8-10 weeks after initiation of therapy. These changes generally resolved within 2-6 weeks after discontinuation of treatment. Treatment with Xeljanz was associated with dose-dependent increases in B cell counts. Changes in circulating T-lymphocyte counts and T-lymphocyte subsets (CD3+, CD4+ and CD8+) were small and inconsistent. The clinical significance of these changes is unknown.
Changes in total serum IgG, IgM and IgA levels over 6-month Xeljanz dosing in patients with rheumatoid arthritis were small, not dose-dependent and similar to those seen on placebo.
After treatment with Xeljanz in patients with rheumatoid arthritis, rapid decreases in serum C-reactive protein (CRP) were observed and maintained throughout dosing. Changes in CRP observed with Xeljanz treatment do not reverse fully within 2 weeks after discontinuation, indicating a longer duration of pharmacodynamic activity compared to the half-life (t
½).
Clinical Efficacy: The efficacy and safety of Xeljanz were assessed in 5 randomized, double-blind, placebo-controlled multicenter studies in patients >18 years with active rheumatoid arthritis diagnosed according to American College of Rheumatology (ACR) criteria. Patients had at least 6 tender and 6 swollen joints at randomization (4 swollen and tender joints for study II). Xeljanz, 5 or 10 mg twice daily, was given as monotherapy (study I) and in combination with disease-modifying antirheumatic drugs (DMARDs) (study II) in patients with an inadequate response to those drugs, and in combination with methotrexate (MTX) in patients with either an inadequate response to MTX (studies III and IV) or inadequate efficacy or lack of tolerance to at least 1 approved tumor necrosis factor (TNF)-inhibiting biologic agent (study V).
Study I was a 6-month monotherapy study in which 610 patients with moderate to severe active rheumatoid arthritis who had an inadequate response to a DMARD (nonbiologic or biologic) received Xeljanz 5 or 10 mg twice daily or placebo. At the month 3 visit, all patients randomized to placebo treatment were advanced in a blinded fashion to a 2nd predetermined treatment of Xeljanz 5 or 10 mg twice daily. The primary endpoints at month 3 were the proportion of patients who achieved an ACR20 response, changes in health assessment questionnaire-disability index (HAQ-DI) and rates of disease activity score (DAS)28-4(ESR) <2.6.
Study II was a 12-month study in which 792 patients with moderate to severe active rheumatoid arthritis who had an inadequate response to a nonbiologic DMARD received Xeljanz 5 or 10 mg twice daily or placebo added to background DMARD treatment (excluding potent immunosuppressive treatments eg, azathioprine or cyclosporine). At the month 3 visit, nonresponding patients randomized to placebo treatment were advanced in a blinded fashion to a 2nd predetermined treatment of Xeljanz 5 or 10 mg twice daily. At the end of month 6, all placebo patients were advanced to their 2nd predetermined treatment in a blinded fashion. The primary endpoints were the proportion of patients who achieved an ACR20 response at month 6, changes in HAQ-DI at month 3 and rates of DAS28-4(ESR) <2.6 at month 6.
Study III was a 12-month study in which 717 patients with moderate to severe active rheumatoid arthritis who had an inadequate response to MTX. Patients received Xeljanz 5 or 10 mg twice daily, adalimumab 40 mg SC every other week or placebo added to background MTX. Placebo patients were advanced as in study II. The primary endpoints were the proportion of patients who achieved an ACR20 response at month 6, HAQ-DI at month 3 and DAS28-4(ESR) <2.6 at month 6.
Study IV is an ongoing 2-year study with a planned analysis at 1 year in which 797 patients with moderate to severe active rheumatoid arthritis who had an inadequate response to MTX received Xeljanz 5 or 10 mg twice daily or placebo added to background MTX. Placebo patients were advanced as in study II. The primary endpoints were the proportion of patients who achieved an ACR20 response at month 6, mean change from baseline in van der Heijde-modified total sharp score (mTSS) at month 6, HAQ-DI at month 3 and DAS28-4(ESR) <2.6 at month 6.
Study V was a 6-month study in which 399 patients with moderate to severe active rheumatoid arthritis who had an inadequate response to at least one approved TNF-inhibiting biologic agent received Xeljanz 5 or 10 mg twice daily or placebo added to background MTX. At the month 3 visit, all patients randomized to placebo treatment were advanced in a blinded fashion to a 2nd predetermined treatment of Xeljanz 5 or 10 mg twice daily. The primary endpoints at month 3 were the proportion of patients who achieved an ACR20 response, HAQ-DI and DAS28-4(ESR) <2.6.
Clinical Response: The percentages of Xeljanz-treated patients achieving ACR20, ACR50 and ACR70 responses in studies I, IV and V are shown in Table 1. In all studies, patients treated with either Xeljanz 5 or 10 mg twice daily had statistically significant ACR 20, ACR50 and ACR70 response rates at month 3 and month 6 versus placebo-treated patients.
In studies I, II and V, improvement in ACR20 response rate versus placebo was observed within 2 weeks. In studies II, III and IV, ACR response rates were maintained to 12 months in Xeljanz-treated patients. ACR response was maintained for 3 years in the ongoing open-label extension studies.
During the 3-month (studies I and V) and 6-month (studies II, III, and IV) controlled portions of the studies, patients treated with Xeljanz at a dose of 10 mg twice daily generally had higher response rates compared to patients treated with Xeljanz 5 mg twice daily. In study III, the primary endpoints were the proportion achieving an ACR20 response at month 6; change in HAQ-DI at month 3 and DAS28-4(ESR) <2.6 at month 6. The data for these primary outcomes were 51.5%, 52.6%, 47.2% and 28.3%; -0.55, -0.61, -0.49; and -0.24; and 6.2%, 12.5%, 6.7% and 1.1% for the Xeljanz 5 mg twice daily, Xeljanz 10 mg twice daily, adalimumab 40 mg SC every other week and placebo groups, respectively. For a pre-specified secondary endpoint, the ACR70 response rates at month 6 for the Xeljanz 5 mg twice daily and 10 mg twice daily groups were significantly greater than adalimumab (19.9%, 21.9% and 9.1%, respectively).
The treatment effect was similar in patients independent of rheumatoid factor status, age, gender, race or disease status. Time to onset was rapid (as early as week 2 in studies I, II and V) and the magnitude of response continued to improve with duration of treatment. As with the overall ACR response in patients treated with Xeljanz 5 or 10 mg twice daily, each of the components of the ACR response was consistently improved from baseline including: Tender and swollen joint counts; patient and physician global assessment; disability index scores; pain assessment and CRP compared to patients receiving placebo plus MTX or other DMARDs in all studies.
Patients in the phase 3 studies had a mean DAS28-4(ESR) of 6.1-6.7 at baseline. Significant reductions in DAS28-4(ESR) from baseline (mean improvement) of 1.8-2 and 1.9-2.2 were observed in 5 mg and 10 mg Xeljanz-treated patients, respectively, compared to placebo-treated patients (0.7–1.1) at 3 months. The proportion of patients achieving a DAS28 clinical remission [DAS28-4(ESR) <2.6] in studies II, III and IV was significantly higher in patients receiving Xeljanz 5 mg or 10 mg (6-9% and 13-16%, respectively) compared to 1-3% of placebo patients at 6 months. In study III, similar percentages of patients achieving DAS28-4(ESR) <2.6 were observed for adalimumab and Xeljanz 5 mg twice daily or 10 mg twice daily at month 6.
In a pooled analysis of the phase 3 studies, the 10 mg twice daily dose provided increased benefit over the 5 mg twice daily dose in multiple measures of signs and symptoms: Improvement from baseline (ACR20, ACR50 and ACR70 response rates) and achievement of targeted disease activity state [either DAS28-4(ESR) <2.6 or ≤3.2]. Greater benefits of 10 mg versus 5 mg were shown in the more stringent measures [ie, ACR70 and DAS28-4(ESR) <2.6 response rates].
Click on icon to see table/diagram/image
The results of the components of the ACR response criteria for studies IV and V are shown in Table 2. Similar results were observed in studies I, II and III.
Click on icon to see table/diagram/image
The percent of ACR20 responders by visit for study IV is shown in Figure 1. Similar responses were observed in studies I, II, III and V.
Click on icon to see table/diagram/image
Radiographic Response: In study IV, structural joint damage was assessed radiographically and expressed as change in mTSS score and its components, the erosion score and joint space narrowing (JSN) score. The results from baseline to month 6 are shown in Table 3.
Xeljanz, 10 mg twice daily, inhibited the progression of structural damage compared to placebo plus MTX at 6 and 12 months. When given at a dose of 5 mg twice daily, Xeljanz exhibited similar effects on mean progression of structural damage at 6 and 12 months (not statistically significant). The proportion of patients with no radiographic progression (defined as ≤0.5 unit increase from baseline in mTSS) at months 6 and 12 was significantly greater in both the Xeljanz 5 mg and 10 mg twice daily groups compared with placebo.
Click on icon to see table/diagram/image
Physical Function Response and Health-Related Outcomes: Improvement in physical functioning was measured by the HAQ-DI. Patients receiving Xeljanz 5 or 10 mg twice daily demonstrated significantly greater improvement from baseline in physical functioning compared to placebo at month 3 (studies I, II, III and V) and month 6 (studies II and III). Xeljanz 5 or 10 mg twice daily-treated patients exhibited significantly greater improved physical functioning compared to placebo as early as week 2 in studies I and II. In study III, mean HAQ-DI improvements were maintained to 12 months in Xeljanz-treated patients. Mean HAQ-DI improvements were maintained for 36 months in the ongoing open-label extension studies. Compared with adalimumab-treated patients, at month 3, patients in the Xeljanz 5 mg twice daily had similar decreases from baseline in HAQ-DI values and patients in the 10 mg twice daily group had significantly greater decreases in HAQ-DI.
Health-related quality of life was assessed by the short form health survey (SF-36) in all 5 studies. In these studies, patients receiving Xeljanz 10 mg twice daily demonstrated significantly greater improvement from baseline compared to placebo in all 8 domains of the SF-36 as well as the physical component summary (PCS) and the mental component summary (MCS) at month 3. Both Xeljanz-treated groups exhibited significantly greater improvement from baseline compared to placebo in all 8 domains as well as PCS and MCS at month 3 in studies I, IV and V. In studies III and IV, mean SF-36 improvements were maintained to 12 months in Xeljanz-treated patients.
Improvement in fatigue was evaluated by the functional assessment of chronic illness therapy-fatigue (FACIT-F) scale at month 3 in all studies. Patients receiving Xeljanz 5 or 10 mg twice daily demonstrated significantly greater improvement from baseline in fatigue compared to placebo in all 5 studies. In studies III and IV, mean FACIT-F improvements were maintained to 12 months in Xeljanz-treated patients.
Improvement in sleep was assessed using the sleep problems index I and II summary scales of the medical outcomes study sleep (MOS-Sleep) measure at month 3 in all studies. Patients receiving Xeljanz 5 or 10 mg twice daily demonstrated significantly greater improvement from baseline in both scales compared to placebo in studies II, III and IV. In studies III and IV, mean improvements in both scales were maintained to 12 months in Xeljanz-treated patients.
Improvement in productivity was evaluated using the work limitations questionnaire (WLQ) scale at month 3 in all studies. Patients receiving Xeljanz 10 mg twice daily demonstrated significantly greater improvement from baseline in the overall output summary scale compared to placebo in studies III, IV, and V. In studies III and IV, mean overall output improvements were maintained to 12 months in Xeljanz 10 mg twice daily-treated patients.
Pharmacokinetics: The pharmacokinetic profile of tofacitinib is characterized by rapid absorption [peak plasma concentrations (C
max) are reached within 0.5-1 hr], rapid elimination (t
½ of approximately 3 hrs) and dose-proportional increases in systemic exposure. Steady-state concentrations are achieved in 24-48 hrs with negligible accumulation after twice daily administration.
Absorption and Distribution: Tofacitinib is well-absorbed, with an oral bioavailability of 74%. Co-administration of Xeljanz with a high-fat meal resulted in no changes in area under the concentration-time curve (AUC) while C
max was reduced by 32%. In clinical trials, tofacitinib was administered without regard to meal.
After IV administration, the volume of distribution is 87 L. The protein-binding of tofacitinib is approximately 40%. Tofacitinib binds predominantly to albumin and does not appear to bind to α
1-acid glycoprotein. Tofacitinib distributes equally between red blood cells and plasma.
Metabolism and Elimination: Clearance mechanisms for tofacitinib are approximately 70% hepatic metabolism and 30% renal excretion of the parent drug. The metabolism of tofacitinib is primarily mediated by CYP3A4 with minor contribution from CYP2C19. In a human radiolabeled study, >65% of the total circulating radioactivity was accounted for by unchanged drug, with the remaining 35% attributed to 8 metabolites, each accounting for <8% of total radioactivity. The pharmacologic activity of tofacitinib is attributed to the parent molecule.
Pharmacokinetic data and dosing recommendations for special populations and drug interactions are provided in Figure 2.
Pharmacokinetics in Rheumatoid Arthritis Patients: Population pharmacokinetic analysis in rheumatoid arthritis patients indicated that systemic exposure (AUC) of tofacitinib in the extremes of body weight (40 kg, 140 kg) were similar to that of a 70-kg patient. Elderly patients 80 years were estimated to have <5% higher AUC relative to the mean age of 55 years. Women were estimated to have 7% lower AUC compared to men. The available data have also shown that there are no major differences in tofacitinib AUC between White, Black and Asian patients. An approximate linear relationship between body weight and volume of distribution was observed, resulting in higher peak (C
max) and lower trough (C
min) concentrations in lighter patients. However, this difference is not considered to be clinically relevant. The between-subject variability (percentage coefficient of variation) in AUC of Xeljanz is estimated to be approximately 27%.
Renal Impairment: Subjects with mild, moderate and severe renal impairment had 37%, 43% and 123% higher AUC, respectively, compared with healthy subjects (see Dosage & Administration). In subjects with end-stage renal disease, the contribution of dialysis to the total clearance of tofacitinib was relatively small.
Hepatic Impairment: Subjects with mild and moderate hepatic impairment had 3% and 65% higher AUC, respectively, compared with healthy subjects. Subjects with severe hepatic impairment were not studied (see Dosage & Administration).
Pediatric Population: The pharmacokinetics, safety and efficacy of tofacitinib in pediatric patients have not been established.
Click on icon to see table/diagram/image
Reference values for weight, age, gender and race comparisons are 70 kg, 55 years, male and White, respectively; reference groups for renal and hepatic impairment data are subjects with normal renal or hepatic function, respectively; reference group for drug interaction and food effect studies is administration of Xeljanz alone: Mod=moderate; Sev=severe; Imp=impairment.
Toxicology: Preclinical Safety Data: In nonclinical studies, effects were observed on the immune and hematopoietic systems that were attributed to the pharmacological properties (JAK inhibition) of tofacitinib. Secondary effects from immunosuppression eg, bacterial and viral infections and lymphoma were observed at clinically relevant doses. Other findings at doses well above human exposures included effects on the hepatic and gastrointestinal systems.
Lymphoma was observed in 3 of 8 adult and 0 of 14 juvenile monkeys dosed with tofacitinib at 5 mg/kg twice daily. The no observed adverse effect level (NOAEL) for the lymphomas was 1 mg/kg twice daily. The unbound AUC at 1 mg/kg twice daily was 341 ng·hr/mL, which is approximately half of the unbound AUC at 10 mg twice daily and similar to the unbound AUC at 5 mg twice daily in humans.
Tofacitinib is not mutagenic or genotoxic based on the results of a series of
in vitro and
in vivo tests for gene mutations and chromosomal aberrations.
The carcinogenic potential of tofacitinib was assessed in 6-month rasH2 transgenic mouse carcinogenicity and 2-year rat carcinogenicity studies. Tofacitinib was not carcinogenic in mice up to a high dose of 200 mg/kg/day (unbound drug AUC of approximately 19-fold the human AUC at 10 mg twice daily). Benign Leydig cell tumors were observed in rats: Benign Leydig cell tumors in rats are not associated with a risk of Leydig cell tumors in humans. Hibernomas (malignancy of brown adipose tissue) were observed in female rats at doses ≥30 mg/kg (unbound drug AUC of approximately 41-fold the human AUC at 10 mg twice daily). Benign thymomas were observed in female rats dosed only at the 100 reduced to 75 mg/kg/day dose (unbound drug AUC of approximately 94-fold the human AUC at 10 mg twice daily).
Tofacitinib was shown to be teratogenic in rats and rabbits, and have effects in rats on female fertility, parturition and peri/postnatal development. Tofacitinib had no effects on male fertility, sperm motility or sperm concentration. Tofacitinib was secreted in milk of lactating rats.