Pharmacology: Mechanism of Action: Larotrectinib is an orally-bioavailable, adenosine triphosphate (ATP)-competitive, potent and highly selective
Tropomyosin Receptor Kinase (TRK) kinase inhibitor. Larotrectinib targets the TRK family of proteins inclusive of TRKA, TRKB, and TRKC that are encoded by
NTRK1,
NTRK2, and
NTRK3 genes, respectively. Larotrectinib has minimal activity with off-target kinases tested.
In-frame gene fusion events resulting from chromosomal rearrangements of the human genes
NTRK1,
NTRK2, and
NTRK3 lead to the formation of oncogenic TRK fusion proteins. These resultant novel chimeric oncogenic proteins are aberrantly expressed driving constitutive kinase activity subsequently activating downstream cell signalling pathways involved in cell proliferation and survival leading to TRK fusion cancer.
Larotrectinib demonstrated potent inhibition of TRK proteins and inhibition of proliferation of cell lines containing
NTRK gene fusions in a concentration-dependent manner. In TRK fusion-driven mouse xenograft models larotrectinib treatment induced significant tumour growth inhibition.
Larotrectinib had minimal activity in cell lines with point mutations in the TRKA kinase domain, including the clinically identified acquired resistance mutation, G595R. Point mutations in the TRKC kinase domain with clinically identified acquired resistance to larotrectinib include G623R, G696A, and F617L.
Pharmacodynamics: Cardiac Electrophysiology: Potential effects of larotrectinib on the QT interval were examined with the use of concentration-response modeling of the QTc data. The model was built on a single data set containing 36 healthy adult subjects receiving single doses ranging from 100 mg to 900 mg (n=6 per treatment arm receiving larotrectinib). Based on the model, larotrectinib at C
max did not prolong the QT interval to any clinically relevant extent.
Clinical Trials: Trial Design and Study Demographics: Three ongoing multicenter, open-label, single-arm clinical studies in patients with advanced cancer contributed patients to a pooled efficacy analysis evaluating VITRAKVI for the treatment of adult and pediatric patients with unresectable or metastatic solid tumours with a Neurotrophic Tyrosine Receptor Kinase (
NTRK) gene fusion, as follows (see Table 2): 1) Phase 1 adult dose-finding study (Study 1 [LOXO-TRK-14001] [n=
13]); 2) Phase 2 adult and pediatric "basket" study (Study 2 [NAVIGATE] [n=
98]); and 3) Phase 1/2 pediatric dose-finding/efficacy and safety study (Study 3 [SCOUT] [n=
53]).
Enrollment to Study 1 and the Phase 1 portion of Study 3 was not restricted to patients with a documented
NTRK gene fusion but patients with prospectively identified
NTRK gene fusions were included in the pooled efficacy analysis. Patients enrolled to Study 2 were required to have tropomyosin receptor kinase (TRK) fusion cancer. All patients were required to have progressed following systemic therapy for their disease, if available, or would have required surgery with significant morbidity. Protocol amendments excluded patients with prior progression on approved or investigational kinase inhibitors with anti-TRK activity from Studies 2 and 3.
The assessment of efficacy is based on an analysis of
164 patients comprising an extended primary analysis set (ePAS). The ePAS includes the first 55 patients with solid tumours with an
NTRK gene fusion who were enrolled across the three clinical studies (the primary analysis set [PAS])
plus additional patients who subsequently started treatment and with ≥ 6 months follow up at the July 15, 2019 cutoff date. Patients in the ePAS were required to have a documented
NTRK gene fusion as determined by local testing; a non-Central Nervous System (non-CNS) primary tumour with ≥ 1 measurable lesion at baseline, per investigator assessment based on Response Evaluation Criteria in Solid Tumours (RECIST), version 1.1 (v1.1); and to have received ≥ 1 dose of VITRAKVI.
Identification of
NTRK gene fusions was prospectively determined in local certified laboratories primarily using next generation sequencing (NGS), in some cases, by fluorescence in situ hybridization (FISH) and by reverse transcription-polymerase chain reaction (RT-PCR) in one case.
The majority of adult patients received a starting dose of VITRAKVI of 100 mg orally twice daily and the majority of pediatric patients received VITRAKVI 100 mg/m
2 up to a maximum dose of 100 mg orally twice daily until unacceptable toxicity or disease progression.
(See Table 2.)
Click on icon to see table/diagram/image
For the pooled efficacy analysis, the primary endpoint was overall response rate (ORR), while the duration of response (DOR) was a secondary endpoint, both determined by a blinded Independent Review Committee (IRC) according to RECIST, v1.1. Additional secondary efficacy outcomes assessed included time to first response. A lower boundary of 30% for ORR, considered to be clinically meaningful, was predefined as statistically significant for response. The ORR was defined as the proportion of patients with the best overall response of confirmed complete response (CR) or confirmed partial response (PR).
Baseline characteristics for the pooled
164 patients with solid tumours harbouring an
NTRK gene fusion were as follows: median age
42 years (range 0.1-
84 years);
34% < 18 years of age,
66% ≥ 18 years, and 21% > 65 years;
77% white and
49% male; and Eastern Cooperative Oncology Group (ECOG) Performance Status (ECOG PS) 0-1
(86%), 2
(12%), or
3 (2%).
Seventy-four percent of patients had metastatic disease and
26% of patients had locally advanced, unresectable disease. Median time from diagnosis was
1.7 years (range: 0.02-31.5 years).
Ninety-four percent of patients had received prior treatment for their cancer, defined as surgery, radiotherapy, or systemic therapy. Of these,
77% had received prior systemic therapy with a median of
1 prior regimen received (range: 0-10).
Twenty-seven percent of all patients had received 3 or more prior systemic therapies and
51% of all patients had received 1-2 prior systemic therapies.
Twenty-two percent of all patients had received no prior systemic therapy.
The most common tumour types represented were soft tissue sarcoma
(22%), infantile fibrosarcoma
(20%), thyroid cancer
(16%), salivary gland tumour
(13%) and
lung (8%).
NTRK gene fusions were identified by NGS, FISH, and RT-PCR in
92.3%, 6.4%, and 1.3% of patients, respectively. The TRK fusions involved
NTRK1 (in
41% of patients),
NTRK2 (in
2%), or
NTRK3 (in
56%) and
31 unique upstream fusion partners. In
nine patients
(5%) with infantile fibrosarcoma who had a documented
ETV6 translocation identified by FISH,
NTRK3 gene fusions were inferred.
Study Results: In the ePAS
(n=164),
with a median duration of follow-up of 15.7 months, the overall response rate (ORR) was
73% (95% confidence interval [CI]:
65, 79). The ORR included a CR in
31 patients
(19%), a surgical CR (sCR) in
8 (5%), and a PR in
80 (49%).
Seventy-six percent of responders (
90 of 164 patients) were still in response, and the duration of response exceeded 6 months in
89%; the median duration of response (DOR) was not yet estimable. The pooled efficacy results for overall response rate and best overall response are presented in Table 3.
(See Table 3.)
Click on icon to see table/diagram/image
The median time to first response was 1.8 months (range:
0.9 to 14.6 months) and
81% of responses occurred within the first 2 months of treatment which coincides with the timing of the first assessment protocol. ORR in the adult sub-population (n=
109) was
63% and 91% in the pediatric sub-population (n=
55).
In an updated analysis, with a median duration of follow-up of 23.3 months, the median DOR for the ePAS (n=164) was 34.5 months (95% CI: 27.6, 54.7).
Changes in target lesion size for individual patients are illustrated in the Waterfall plot in the figure.
(See figure.)
Click on icon to see table/diagram/image
Additional efficacy results by tumour type and by
NTRK gene fusion partner are presented in Table 4 and Table 5, respectively.
(See Tables 4 and 5.)
Click on icon to see table/diagram/image
Click on icon to see table/diagram/image
Patients with CNS Tumors: Twenty-four patients with primary CNS tumors and measurable disease at baseline were enrolled in study 2 ("NAVIGATE") and in study 3 ("SCOUT"). Baseline characteristics for the 24 patients with primary CNS tumors with an NTRK gene fusion assessed by investigator were as follows: median age 8 years (range 1.3-79 years); 20 patients < 18 years of age and 4 patients ≥ 18 years, and 19 patients white and 11 patients male; and ECOG PS 0-1 (22 patients), or 2 (1 patient). All CNS tumor patients had received prior cancer treatment (surgery, radiotherapy and/or previous systemic therapy). There was a median of 1 prior systemic treatment regimen received. Tumor responses for primary CNS tumors were assessed by the investigator using Response Assessment in Neuro Oncology (RANO) Criteria or Response Evaluation Criteria in Solid Tumors (RECIST v1.1).
Of the 24 patients with primary CNS tumors, confirmed response was observed in 5 patients (21%) with 2 of the 24 patients (8%) being complete responders and 3 (12.5%) being partial responders. At a median follow-up time of 10.1 months, the median duration of response was 4.9 months (1.7+ months, 10.1+ months). Further, 17 patients (71%) had stable disease. Two patients (8%) had a progressive disease.
Pharmacokinetics: See Table 6.
Click on icon to see table/diagram/image
Absorption: VITRAKVI is available as a capsule and oral solution formulation. In healthy adult subjects, the AUC of larotrectinib in the oral solution formulation was similar to the capsule; C
max was 36% higher with the oral solution formulation.
The mean absolute bioavailability of larotrectinib was 34% (range: 32% to 37%) following a single 100 mg oral dose.
C
max and AUC in the capsule formulation were dose proportional in healthy adult subjects up to 400 mg and slightly greater than proportional at doses of 600 to 900 mg. Systemic accumulation is 1.6 fold at steady state.
Effect of Food: Larotrectinib C
max was reduced by approximately 35% and there was no effect on AUC in healthy subjects administered VITRAKVI after a high-fat and high-calorie meal compared to the C
max and AUC after overnight fasting.
Distribution: Binding of larotrectinib to human plasma proteins
in vitro was approximately 70% and was independent of drug concentration. The blood-to-plasma concentration ratio was approximately 0.9.
Metabolism: Larotrectinib is metabolized predominantly by CYP3A4/5 (see Interactions). Following oral administration of a single 100 mg dose of radiolabeled larotrectinib to healthy adult subjects, unchanged larotrectinib
(19%) and O-glucuronide larotrectinib that is formed following loss of the hydroxypyrrolidine-urea moiety
(26%) were the major circulating radioactive drug components in plasma.
Elimination: Following oral administration of 100 mg radiolabeled larotrectinib as an oral solution to healthy adult subjects, 58% (5% unchanged) of the administered radioactivity was recovered in feces and 39% (20% unchanged) was recovered in urine.
Special Populations and Conditions: Pediatrics: Based on population pharmacokinetic analyses exposure (C
max and AUC) in pediatric patients (1 month to < 3 months of age) at the recommended dose of 100 mg/m
2 with a maximum of 100 mg BID was 3-fold higher than in adults (≥ 18 years of age) given the dose of 100 mg BID. At the recommended dose, the C
max in pediatric patients (≥ 3 months to < 12 years of age) was higher than in adults, but the AUC was similar to that in adults. For pediatric patients older than 12 years of age, the recommended dose is likely to give similar C
max and AUC as observed in adults.
Geriatrics: Based on population pharmacokinetic analyses, C
max and AUC in patients > 65 years were similar to those in younger patients (< 65 years).
Sex: Gender had no significant effect on the systemic exposure of larotrectinib based on population pharmacokinetic analyses.
Ethnic Origin: Race had no significant effect on the systemic exposure of larotrectinib based on population pharmacokinetic analyses. Caucasians accounted for 72% of the analysis population.
Hepatic Insufficiency: A pharmacokinetic study was conducted in subjects with mild (Child-Pugh A), moderate (Child-Pugh B) and severe (Child-Pugh C) hepatic impairment, and in healthy adult control subjects with normal hepatic function matched for age, body mass index and sex. All subjects received a single 100 mg dose of larotrectinib. An increase in larotrectinib AUC
0-inf was observed in subjects with mild, moderate and severe hepatic impairment of 1.3, 2 and 3.2-fold respectively versus those with normal hepatic function. C
max was observed to increase slightly by 1.1, 1.1 and 1.5-fold respectively. Reduce the starting dose of VITRAKVI by 50% in patients with moderate (Child-Pugh B) to severe (Child-Pugh C) hepatic impairment. No dose adjustment is recommended for patients with mild (Child-Pugh A) hepatic impairment.
Renal Insufficiency: A pharmacokinetic study was conducted in subjects with end stage renal disease requiring dialysis, and in healthy adult control subjects with normal renal function matched for age, body mass index and sex. All subjects received a single 100 mg dose of larotrectinib. An increase in larotrectinib C
max and AUC
0-inf, of 1.25 and 1.46-fold respectively was observed in renally impaired subjects versus those with normal renal function. No dose adjustment is recommended for patients with renal impairment of any severity.
Body Weight: Body weight from 5.0 kg to 179.4 kg had no significant effect on the AUC of larotrectinib based on population pharmacokinetic analyses. The mean AUC of larotrectinib may be increased in children weighing < 5.0 kg (see Pediatrics as previously mentioned).
Toxicology: Non-Clinical Toxicology: General Toxicity: Repeated-dose toxicity was assessed in studies with daily oral administration up to 13-weeks in rats and monkey. Dose limiting skin lesions were only seen in rats and were primarily responsible for mortality and morbidity. In rats, severe toxicity was observed at doses corresponding to human AUC at the recommended clinical dose. Clinical signs of gastrointestinal toxicity including emesis, were dose limiting in monkeys. No relevant systemic toxicity were observed in monkeys at exposures which correspond to > 10-times the human AUC at the recommended clinical dose.
Increased body weight, increased food consumption, and elevated serum liver enzymes (ALT and/or AST) are additional relevant findings that were observed in both species.
Genotoxicity and Carcinogenicity: Larotrectinib was not mutagenic in bacterial reverse mutation (Ames) assays and in
in vitro mammalian mutagenesis assays. Larotrectinib was negative in the
in vivo mouse micronucleus test.
Carcinogenicity studies have not been performed with larotrectinib.
Reproductive and Developmental Toxicology: Reproduction Toxicity: Fertility studies with larotrectinib have not been conducted. In 13-week repeated-dose studies, larotrectinib had no effects on spermatogenesis in rats and on the histopathology of male reproductive organs in rats and monkeys at doses corresponding to approximately 7-times (rats) and 10-times (monkeys) the human AUC at the recommended clinical dose.
In a 1-month study in female rats, fewer corpora lutea, increased incidence of anestrus and decreased uterine weight with uterine atrophy were observed at doses corresponding to approximately 8-times the human AUC at the recommended clinical dose; these effects were reversible. No effects on reproductive organs were seen in the 13-week study in rats and monkeys at doses corresponding to approximately 3 times (rats) and approximately 17-times (monkeys) the human AUC at the recommended clinical dose.
Development: In embryo-fetal development studies where pregnant rats and rabbits were dosed with larotrectinib during the period of organogenesis, malformations were observed at maternal exposures that were approximately 9- and 0.6-times, respectively, those observed at the clinical dose of 100 mg twice daily. Larotrectinib was not embryotoxic up to maternally toxic doses. Larotrectinib crosses the placenta in both species and can be detected in blood samples obtained from fetuses at termination.
Juvenile Toxicity: Larotrectinib was administered in a juvenile toxicity study in rats at twice daily doses of 0.2, 2 and 7.5 mg/kg from postnatal day (PND) 7 to 27 and at twice daily doses of 0.6, 6 and 22.5 mg/kg between PND 28 and 70. The dosing period was equivalent to human pediatric populations from newborn to adulthood. The lowest dose (0.2/0.6 mg/kg BID), equivalent to 0.02-fold the recommended clinical exposure, was considered the NOAEL. At doses ≥ 2/6 mg/kg BID (0.5-fold the recommended clinical exposure), increased mortality, neuronal effects (increased incidence of partially closed eyelids, lower hindlimb grip strength and foot splay), decreased growth (shorter tibial length and lower body weight gain with lower food intake) and delay in sexual development were noted. At doses 7.5/22.5 mg/kg BID (3-fold the recommended clinical exposure), central nervous system-related signs including head flick and circling, increased escape time and number of errors in a maze swim test when the original path is reversed, skin lesions, and swollen abdomen (females) were noted. Lower fertility was noted in animals at 3-times the recommended clinical exposure.
Microbiology: No microbiological information is required for this drug product.