Abstract
Background
WNT974 is a small molecule inhibitor of Wnt signaling that specifically inhibits porcupine O-acyltransferase. This phase Ib dose-escalation study evaluated the maximum tolerated dose of WNT974 in combination with encorafenib and cetuximab in patients with BRAF V600E-mutant metastatic colorectal cancer with RNF43 mutations or RSPO fusions.
Patients and Methods
Patients received once-daily encorafenib and weekly cetuximab, in addition to once-daily WNT974, in sequential dosing cohorts. In the first cohort, patients received 10-mg WNT974 (COMBO10), which was reduced in subsequent cohorts to 7.5-mg (COMBO7.5) or 5-mg (COMBO5) after dose–limiting toxicities (DLTs) were observed. Primary endpoints were incidence of DLTs and exposure to WNT974 and encorafenib. Secondary endpoints were anti-tumor activity and safety.
Results
Twenty patients were enrolled (COMBO10, n = 4; COMBO7.5, n = 6; COMBO5, n = 10). DLTs were observed in 4 patients, including grade 3 hypercalcemia (COMBO10, n = 1; COMBO7.5, n = 1), grade 2 dysgeusia (COMBO10, n = 1), and lipase increased (COMBO10, n = 1). A high incidence of bone toxicities (n = 9) was reported, including rib fracture, spinal compression fracture, pathological fracture, foot fracture, hip fracture, and lumbar vertebral fracture. Serious adverse events were reported in 15 patients, most frequently bone fracture, hypercalcemia, and pleural effusion. The overall response rate was 10% and disease control rate 85%; most patients achieved stable disease as their best response.
Conclusion
Concerns surrounding the safety and lack of preliminary evidence of improved anti-tumor activity of WNT974 + encorafenib + cetuximab, compared with previous encorafenib + cetuximab data, ultimately led to study discontinuation. Phase II was not initiated.
Trial registration
ClinicalTrials.gov, NCT02278133
Keywords: WNT974, encorafenib, cetuximab, metastatic, colorectal cancer
This phase Ib dose-escalation study evaluated the maximum tolerated dose of WNT974 in combination with encorafenib and cetuximab in patients with BRAF V600E-mutant metastatic colorectal cancer with RNF43 mutations or RSPO fusions.
Implications for Practice.
This phase Ib dose-escalation study evaluates the maximum tolerated dose of WNT974, a Wnt inhibitor, when combined with encorafenib + cetuximab in patients with BRAF V600E-mutant metastatic colorectal cancer with RNF43 mutations or RSPO fusions. The addition of WNT974 to encorafenib + cetuximab was limited by severe toxicities. This study provides important information for future studies that include the addition of a Wnt inhibitor for the treatment of BRAF V600E-mutant metastatic colorectal cancer.
Introduction
Colorectal cancer (CRC) is the third most common cancer worldwide and the second most common cause of cancer deaths.1 The Wnt signaling pathway is altered in most CRCs, most commonly due to inactivating adenomatous polyposis coli (APC) mutations.2-4 In addition, some CRCs carry mutations in other genes, including RNF43 mutations and RSPO fusions, which also activate the Wnt pathway and are mutually exclusive to APC mutations.3,5-7RNF43 mutations are also associated with high microsatellite instability.8 These mutations serve as potential therapeutic targets for upstream inhibition of the Wnt pathway.9
RNF43 and RSPO alterations in CRCs are associated with concurrent activating mutations in BRAF, a potent modulator of MAPK signaling.4 Approximately 10% of patients with metastatic CRC (mCRC) have a BRAF mutation, most commonly at the V600 codon.10,11 Patients with BRAFV600E-mutant mCRC typically have a worse prognosis than those with mCRC without BRAF mutations.12 BRAF inhibitor monotherapy shows little clinical benefit in patients with BRAF V600E-mutant mCRC due to EGFR-mediated reactivation of the MAPK pathway in response to BRAF inhibition.13-15 Combining the EGFR inhibitor cetuximab with the BRAF inhibitor encorafenib has been proven successful in overcoming this feedback loop and improving survival outcomes in patients with BRAF V600E-mutant mCRC.16
Crosstalk between the MAPK and Wnt signaling pathways suggests that they may act together to drive progression of CRC.17 Combining agents that target the aberrant signaling through both pathways may potentially improve outcomes in patients with CRC.18 WNT974 (formerly LGK974) is a small-molecule inhibitor of Wnt signaling, specifically inhibiting porcupine O-acyltransferase (PORCN).19 In vitro and in vivo, WNT974 demonstrated anti-tumor activity in CRC preclinical models.20-22 A phase I study of WNT974 reported a manageable safety profile in patients with advanced solid tumors, with the most frequently reported adverse events (AEs) across all doses being dysgeusia (50%), decreased appetite (45.7%), nausea (33.0%), fatigue (31.9%), and vomiting (30.9%).23
Here we report safety, pharmacokinetics (PK), and efficacy results from a phase Ib study to evaluate the combination of WNT974 + encorafenib + cetuximab in patients with BRAF V600E-mutant mCRC with RNF43 mutations or RSPO fusions.
Methods
Study Design and Patients
This open-label, multicenter, phase Ib dose-escalation study aimed to estimate the maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D) of WNT974 + encorafenib + cetuximab in patients with BRAF V600E-mutant mCRC harboring upstream Wnt pathway mutations. A single-arm phase II part of this study was planned to estimate the preliminary anti-tumor activity of the RP2D. The study protocol, informed consent form, and printed patient information materials were reviewed and approved by the independent ethics committee and/or local institutional review board for each site before any study procedures were performed (Supplemental Table S1). This study was conducted according to International Council for Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use guidelines concerning Good Clinical Practice, the Declaration of Helsinki, the European Union Clinical Trials Directive (2001/20/EC), Title 21 of the United States Code of Federal Regulations, and the practices and regulations of each participating nation. Written informed consent to participate in the study was obtained from each patient before any study-specific procedures were performed.
Eligible patients were 18 years or older; had mCRC with documented KRAS wild-type (WT) status, BRAF V600E mutation, and RNF43 mutation and/or RSPO fusions detected by molecular testing; experienced disease progression after one or more prior standard-of-care regimens for mCRC; and had an Eastern Cooperative Oncology Group (ECOG) performance status ≤2 and evidence of measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. A tumor biopsy (primary or metastatic, archival or newly obtained) was required at baseline. Patients with symptomatic brain metastases, symptomatic or untreated leptomeningeal disease, known acute or chronic pancreatitis, clinically significant cardiac disease, impaired hepatic function, or gastrointestinal disease or impaired gastrointestinal function that could significantly alter the absorption of WNT974 or encorafenib, as well as those receiving treatment with medication or consuming foods that were strong inhibitors/inducers of cytochrome CYP3A4/5, were excluded. Patients with prior treatment, including RAF inhibitors, Wnt pathway inhibitors, cetuximab, panitumumab, and/or other EGFR inhibitors were excluded from the phase II study only.
Study Treatment and Procedures
Patients were enrolled sequentially into cohorts, and treatments were administered in 28-day cycles. At least 15 patients were planned for enrollment and 20 patients were enrolled in the phase Ib study. In all cohorts, 200 mg of encorafenib was administered orally once daily and cetuximab was administered intravenously once weekly starting with 400 mg/m2 in week 1 and 250 mg/m2 per week thereafter. In the first cohort, patients received 10 mg of oral WNT975 once daily (COMBO10). However, after 2 of the 4 patients in this cohort experienced dose-limiting toxicities (DLTs), the second cohort was initiated with a dose of 5 mg of WNT974 once daily (COMBO5). No patients in the second cohort reported a DLT. Therefore, cohort 3 was initiated with a dose of 7.5 mg of WNT974 once daily (COMBO7.5), and patient reported a DLT. Once the COMBO7.5 cohort was completed, an enrichment cohort was re-opened at the COMBO5 dose. Patients were treated until they experienced unacceptable toxicity that precluded any further treatment, disease progression, or treatment was discontinued at the investigator’s discretion or the patient’s request.
Patients who developed hypercalcemia (any grade) were treated with bisphosphonates per institutional standards. Due to emerging bone toxicity, patients were monitored by DEXA scan at screening, cycle 3 day 1, every 16 weeks, and as clinically indicated (following protocol amendment); patients who demonstrated bone loss either discontinued WNT974 treatment (in the case of bone fractures or new DEXA T-score of less than −2.5) or were treated with bisphosphonates and were monitored by a DEXA scan every 16 weeks.
Endpoints
The primary endpoints of the phase Ib study were the incidence of DLTs and exposure to WNT974 and encorafenib as measured by PK parameters. A DLT was defined as an AE or abnormal laboratory value assessed as unrelated to disease, disease progression, intercurrent illness, or concomitant medications that occurred within the first 28 days of treatment with WNT974 + encorafenib + cetuximab. The secondary endpoints for this study included the objective response rate (ORR; RECIST 1.1), disease control rate (DCR), and frequency and severity of AEs (Common Terminology Criteria for Adverse Events v4.03).
Tumor assessments were performed at baseline (screening) and every 6 weeks after starting study treatment until disease progression, initiation of subsequent antineoplastic therapy, or death, whichever occurred first. End-of-treatment tumor assessment was conducted within 14 days of receiving the last dose or treatment discontinuation. Safety was assessed throughout the study and included physical examinations, vital signs, weight, performance status evaluation, laboratory evaluations, dermatologic evaluations, ophthalmological examinations, bone loss assessments, cardiac assessments, and documentation of all non-serious and serious AEs (SAEs).
Pharmacokinetics
To assess plasma concentrations of WNT974, its active metabolite LHA333, and encorafenib, serial blood samples were collected on days 1 and 15 of cycle 1, and pre-dose blood samples were collected on days 2, 8, 16, and 22 of cycle 1, and on day 1 of cycles 2, 3, and 4. PK parameters were determined using noncompartmental methods. The PK analysis set included all patients with at least one available valid PK concentration measurement who received at least one dose of study drug.
Statistical Analysis
Approximately 15 patients were planned to be enrolled in the phase Ib dose-escalation part of the study for the model to have reasonable operating characteristics relating to its MTD recommendation. The full analysis set (FAS) comprised all patients who received at least one full or partial dose of the assigned combination of drugs. The safety analysis set included all patients from the FAS who had at least one valid post-baseline safety assessment. The dose-determining set consisted of all patients in the safety analysis set who either met the minimum exposure criterion (met if the patient received at least 21 of the 28 planned daily oral doses of WNT974 and encorafenib in the first 28 days of dosing, at least 50% [14 out of 28 days] of the planned combination doses of WNT974 and encorafenib administered together, and must have received the planned cetuximab loading dose and 2 additional planned cetuximab doses during the first 28 days of the study) and had sufficient safety evaluations (patients who did not experience a DLT during the first cycle if they had been observed for ≥28 days following the first dose and were considered by the Sponsor and Investigators to have enough safety data to conclude that a DLT did not occur) or who experienced a DLT during cycle 1. ORRs and DCRs were summarized using 95% CIs based on the exact binomial distribution.
Results
Patient and Disease Characteristics
Between March 2015 and April 2016, 20 patients were enrolled (data cutoff: August 25, 2017): 4 patients in COMBO10, 6 patients in COMBO7.5, and 10 patients in COMBO5. Patient and disease characteristics at baseline are shown in Table 1. Median patient age was 61 years and median number of previous treatment regimens was 2 (range: 1-6). Most patients were Caucasian (90%) and had an ECOG performance status of 0 (45%) or 1 (50%). Patients discontinued treatment due to disease progression (n = 15 [75%]), AEs (n = 3 [15%]), death (n = 1 [5%]), or physician decision (n = 1 [5%]) (Table 2). The median (range) duration of exposure to WNT974 + encorafenib + cetuximab for all patients was 22 weeks (0-80 weeks [Table 2]). The median (range) relative dose intensities for WNT974 and encorafenib were 86.3% (60%-100%) and 86.0% (60%-100%), respectively, for all patients. Because enrollment was discontinued and the phase II study was not initiated, MTD and RP2D were not determined.
Table 1.
Patient and disease characteristics at baseline.
COMBO5 (n = 10) |
COMBO7.5 (n = 6) |
COMBO10 (n = 4) |
All patients (N = 20) |
|
---|---|---|---|---|
Median age, years (range) | 63.5 (50-70) | 60.5 (51-75) | 60.5 (60-61) | 61.0 (50-75) |
Female sex | 6 (60.0) | 4 (66.7) | 1 (25.0) | 11 (55.0) |
Race | ||||
Caucasian | 9 (90.0) | 6 (100.0) | 3 (75.0) | 18 (90.0) |
Asian | 1 (10.0) | 0 | 1 (25.0) | 2 (10.0) |
ECOG performance status | ||||
0 | 3 (30.0) | 4 (66.7) | 2 (50.0) | 9 (45.0) |
1 | 6 (60.0) | 2 (33.3) | 2 (50.0) | 10 (50.0) |
2 | 1 (10.0) | 0 | 0 | 1 (5.0) |
Prior surgery | 8 (80.0) | 6 (100.0) | 4 (100.0) | 18 (90.0) |
Number of prior regimens, median (range) | 2.5 (1-6) | 2.0 (2-3) | 3.0 (2-6) | 2.0 (1-6) |
Primary site of cancer | ||||
Colon | 10 (100.0) | 6 (100.0) | 3 (75.0) | 19 (95.0) |
Rectum | 0 | 0 | 1 (25.0) | 1 (5.0) |
Stage at initial diagnosis | ||||
IIB | 1 (10.0) | 0 | 0 | 1 (5.0) |
III | 1 (10.0) | 1 (16.7) | 0 | 2 (10.0) |
IIIB | 1 (10.0) | 1 (16.7) | 0 | 2 (10.0) |
IIIC | 1 (10.0) | 3 (50.0) | 1 (25.0) | 5 (25.0) |
IV | 4 (40.0) | 1 (16.7) | 1 (25.0) | 6 (30.0) |
IVA | 1 (10.0) | 0 | 0 | 1 (5.0) |
IVB | 1 (10.0) | 0 | 1 (25.0) | 2 (10.0) |
Unknown | 0 | 0 | 1 (25.0) | 1 (5.0) |
Stage at study entry | ||||
IV | 5 (50.0) | 3 (50.0) | 3 (75.0) | 11 (55.0) |
IVA | 0 | 1 (16.7) | 0 | 2 (10.0) |
IVB | 5 (50.0) | 2 (33.3) | 1 (25.0) | 8 (40.0) |
Time from initial diagnosis of primary site to start of study treatment (months) | ||||
Mean (SD) | 32.22 (27.1) | 27.16 (15.3) | 27.19 (14.5) | 29.70 (21.2) |
Median (min-max) | 25.49 (3.3-92.0) | 23.01 (14.9-55.4) | 26.53 (12.2-43.5) | 24.97 (3.3-92.0) |
Time from initial diagnosis of primary site to first recurrence/progression (months) | ||||
Mean (SD) | 15.13 (13.7) | 9.63 (5.0) | 16.16 (16.3) | 13.68 (12.0) |
Median (min-max) | 13.14 (2.3-49.6) | 11.78 (0.0-13.2) | 11.53 (1.9-39.7) | 11.86 (0.0-49.6) |
Time from most recent relapse/progression to start of study treatment (months) | ||||
Mean (SD) | 3.19 (3.3) | 9.06 (16.9) | 2.06 (0.9) | 4.73 (9.4) |
Median (min-max) | 2.00 (0.9-12.0) | 2.45 (0.9-43.4) | 1.91 (1.1-3.3) | 2.05 (0.9-43.4) |
Types of lesions at baseline | ||||
Target only | 0 | 0 | 1 (25.0) | 1 (5.0) |
Both target and non-target | 10 (100.0) | 6 (100.0) | 3 (75.0) | 19 (95.0) |
Data are n (%) unless indicated otherwise.
Abbreviation: ECOG, Eastern Cooperative Oncology Group.
Table 2.
Treatment discontinuation and duration of exposure.
COMBO5 (n = 10) |
COMBO7.5 (n = 6) |
COMBO10 (n = 4) |
All patients (N = 20) |
|
---|---|---|---|---|
Treatment discontinued, n (%) | 10 (100) | 6 (100) | 4 (100) | 20 (100) |
Primary reason for treatment discontinuation, n (%) | ||||
Adverse event | 1 (10.0) | 1 (16.7) | 1 (25.0) | 3 (15.0) |
Death | 0 | 0 | 1 (25.0) | 1 (5.0) |
Physician decision | 1 (10.0) | 0 | 0 | 1 (5.0) |
Progressive disease | 8 (80.0) | 5 (83.3) | 2 (50.0) | 15 (75.0) |
Duration of exposure, weeksa | ||||
Mean (SD) | 29 (21) | 21 (12) | 26 (16) | 26 (17) |
Median (min, max) | 25 (11, 80) |
21 (0, 35) |
22 (14, 48) |
22 (0, 80) |
aDuration of exposure was defined as (date of last exposure to study treatment - date of first administration of study treatment +1)/7.
Abbreviations: max, maximum; min, minimum.
Dose-Limiting Toxicities
Three DLTs were reported: 1 patient in COMBO10 had grade 2 dysgeusia, 1 patient in COMBO10 had grade 4 increased lipase, and 1 patient in COMBO7.5 had grade 3 hypercalcemia (Table 3). All DLTs resolved following dose adjustment or temporary dose interruption.
Table 3.
Dose-limiting toxicities by preferred term.
COMBO5 (n= 10) |
COMBO7.5 (n = 6) |
COMBO10 (n = 4) |
All patients (N = 20) |
|
---|---|---|---|---|
Any preferred term, n (%) | 1 (16.7) | 2 (50.0) | 3 (20.0) | |
Increased alanine aminotransferase | a | 0 | 0 | 1 (5.0) |
Increased aspartate aminotransferase | a | 0 | 0 | 1 (5.0) |
Increased lipase | 0 | 0 | 1 (25.0) | 1 (5.0) |
Hypercalcemia | 0 | 1 (16.7) | 0 | 1 (5.0) |
Dysgeusia | 0 | 0 | 1 (25.0) | 1 (5.0) |
aOne patient with events of increased alanine aminotransferase and aspartate aminotransferase was originally included as having DLT events; upon investigation, the investigator categorized both events as not suspected to be study treatment-related and it was deemed they were misclassified as DLTs.
Abbreviations: DLT, dose-limiting toxicity.
Safety
The most common AEs (any grade) were hypercalcemia, arthralgia, fatigue, and anemia (Table 4). Grade 3 or 4 AEs occurred in 16 patients, of whom ≥20% experienced hypercalcemia, hypophosphatemia, and increased aspartate aminotransferase. Treatment-related bone fractures occurred in 9 patients; 1 of these patients had metastatic bone lesions. SAEs occurred in 15 patients; the most common (occurring in ≥10% of patients) were bone fractures, hypercalcemia, and pleural effusion. One patient had serious bone pain. Four patients discontinued treatment due to an AE: bone fracture (n = 2), abdominal infection (n = 1), and infusion-related reaction (n = 1). Two patients died during the study (ie, within 30 days of the last treatment dose) due to disease progression.
Table 4.
AEs occurring in ≥20% of all patients.
COMBO5 (n = 10) |
COMBO7.5 (n = 6) |
COMBO10 (n = 4) |
All patients (N = 20) |
|||||
---|---|---|---|---|---|---|---|---|
All grades | Grade 3/4 | All grades | Grade 3/4 | All grades | Grade 3/4 | All grades | Grade 3/4 | |
Any AE | 10 (100.0) | 7 (70.0) | 6 (100.0) | 5 (83.3) | 4 (100.0) | 4 (100.0) | 20 (100.0) | 16 (80.0) |
Any SAE | 8 (80.0) | 6 (60.0) | 4 (66.7) | 3 (50.0) | 3 (75.0) | 3 (75.0) | 15 (75.0) | 12 (60.0) |
Hypercalcemia | 5 (50.0) | 1 (10.0) | 5 (83.3) | 2 (33.3) | 4 (100.0) | 4 (100.0) | 14 (70.0) | 7 (35.0) |
Arthralgia | 5 (50.0) | 0 | 4 (66.7) | 0 | 4 (100.0) | 0 | 13 (65.0) | 0 |
Fatigue | 6 (60.0) | 0 | 2 (33.3) | 0 | 3 (75.0) | 0 | 11 (55.0) | 0 |
Anemia | 5 (50.0) | 1 (10.0) | 2 (33.3) | 0 | 3 (75.0) | 2 (50.0) | 10 (50.0) | 3 (15.0) |
Bone fracture | 4 (40.0) | 1 (10.0) | 4 (66.7) | 3 (50.0) | 1 (25.0) | 1 (25.0) | 9 (45.0) | 5 (25.0) |
Constipation | 5 (50.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 0 | 9 (45.0) | 0 |
Nausea | 4 (40.0) | 0 | 4 (66.7) | 0 | 1 (25.0) | 0 | 9 (45.0) | 0 |
Back pain | 3 (30.0) | 1 (10.0) | 3 (50.0) | 1 (16.7) | 2 (50.0) | 0 | 8 (40.0) | 2 (10.0) |
Hypophosphatemia | 4 (40.0) | 3 (30.0) | 2 (33.3) | 1 (16.7) | 2 (50.0) | 1 (25.0) | 8 (40.0) | 5 (25.0) |
Pyrexia | 5 (50.0) | 0 | 2 (33.3) | 0 | 1 (25.0) | 0 | 8 (40.0) | 0 |
Vomiting | 4 (40.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 0 | 8 (40.0) | 0 |
Decreased appetite | 3 (30.0) | 0 | 2 (33.3) | 1 (16.7) | 2 (50.0) | 0 | 7 (35.0) | 1 (5.0) |
Dysgeusia | 2 (20.0) | 0 | 2 (33.3) | 0 | 3 (75.0) | 0 | 7 (35.0) | 0 |
Hypokalemia | 3 (30.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 1 (25.0) | 7 (35.0) | 1 (5.0) |
Hypomagnesemia | 3 (30.0) | 0 | 1 (16.7) | 0 | 3 (75.0) | 1 (25.0) | 7 (35.0) | 1 (5.0) |
Myalgia | 3 (30.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 0 | 7 (35.0) | 0 |
Peripheral edema | 3 (30.0) | 0 | 0 | 0 | 4 (100.0) | 0 | 7 (35.0) | 0 |
Abdominal pain | 2 (20.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 0 | 6 (30.0) | 0 |
Diarrhea | 3 (30.0) | 0 | 1 (16.7) | 0 | 1 (25.0) | 0 | 5 (25.0) | 0 |
Hypocalcemia | 1 (10.0) | 0 | 1 (16.7) | 0 | 3 (75.0) | 1 (25.0) | 5 (25.0) | 1 (5.0) |
Osteoporosis | 2 (20.0) | 0 | 2 (33.3) | 0 | 1 (25.0) | 0 | 5 (25.0) | 0 |
Pain in extremity | 2 (20.0) | 0 | 1 (16.7) | 0 | 2 (50.0) | 1 (25.0) | 5 (25.0) | 1 (5.0) |
Weight decreased | 1 (10.0) | 0 | 2 (33.3) | 0 | 2 (50.0) | 0 | 5 (25.0) | 0 |
Increased aspartate aminotransferase | 2 (20.0) | 2 (20.0) | 1 (16.7) | 1 (16.7) | 1 (25.0) | 1 (25.0) | 4 (20.0) | 4 (20.0) |
Increased blood bilirubin | 1 (10.0) | 0 | 2 (33.3) | 1 (16.7) | 1 (25.0) | 1 (25.0) | 4 (20.0) | 2 (10.0) |
Increased blood creatinine | 0 | 0 | 1 (16.7) | 0 | 3 (75.0) | 1 (25.0) | 4 (20.0) | 1 (5.0) |
Chills | 2 (20.0) | 0 | 1 (16.7) | 0 | 1 (25.0) | 0 | 4 (20.0) | 0 |
Dyspnea | 2 (20.0) | 0 | 2 (33.3) | 1 (16.7) | 0 | 0 | 4 (20.0) | 1 (5.0) |
Headache | 2 (20.0) | 1 (10.0) | 2 (33.3) | 0 | 0 | 0 | 4 (20.0) | 1 (5.0) |
Infusion-related reaction | 1 (10.0) | 0 | 2 (33.3) | 1 (16.7) | 1 (25.0) | 0 | 4 (20.0) | 1 (5.0) |
Musculoskeletal pain | 4 (40.0) | 0 | 0 | 0 | 0 | 0 | 4 (20.0) | 0 |
Peripheral sensory neuropathy | 1 (10.0) | 0 | 2 (33.3) | 0 | 1 (25.0) | 0 | 4 (20.0) | 0 |
Data are n (%).
Abbreviations: AE, adverse event; SAE, serious adverse event.
A phase II study was planned to assess the clinical efficacy and further safety of the treatment combination. However, enrollment was halted in April 2016 prior to initiation of phase II due to the emergence of 6 reported serious AEs of bone fracture across multiple studies involving WNT974. The preliminary efficacy data of the triplet combination (WNT974 + encorafenib + cetuximab) demonstrated limited improvement in clinical activity relative to the efficacy observed with doublet treatment (encorafenib + cetuximab) in patients with BRAF V600E-mutant mCRC in a separate ongoing clinical trial.24 This, in combination with the safety profile observed with this triplet combination (WNT974 + encorafenib + cetuximab), suggested that more restrictive patient selection, additional on-treatment monitoring, and prophylactic treatment to prevent bone resorption may be required, which would have hindered further development of the combination. Therefore, the decision was made to discontinue enrollment and to not initiate phase II of this study.
Pharmacokinetics
Table 5 shows PK data for WNT974, its active metabolite LHA333, and encorafenib on day 15 of cycle 1. The mean areas under the plasma concentration-time curve (AUCs) of WNT974 and LHA333 increased in a dose-proportional manner. Median time to maximum concentration (Tmax) for WNT974, LHA333, and encorafenib ranged from 1 to 4 h across all treatment combinations. Mean half-life (t1/2) values for WNT974 were similar between the COMBO5 and COMBO7.5 cohorts (4.7 and 5.4 h, respectively) and were higher for the COMBO10 cohort (11.0 h). Similar trends occurred with the mean t1/2 of LHA333 (8.0, 7.4, and 15.0 h for COMBO5, COMBO7.5, and COMBO10, respectively). For encorafenib, mean AUC and maximum plasma drug concentration were generally comparable across all treatment combinations. Median Tmax was observed at approximately 2 h for all cohorts, and mean t1/2 was similar across increasing doses of WNT974 at 4.3 h on day 15.
Table 5.
Steady-state pharmacokinetics (day 15 of cycle 1) of WNT974, its active metabolite LHA333, and encorafenib.
WNT974 | LHA333 | Encorafenib | |||||
---|---|---|---|---|---|---|---|
COMBO5 (n = 9) |
COMBO7.5 (n = 5) |
COMBO10 (n = 3) |
COMBO5 (n = 9) |
COMBO7.5 (n = 5) |
COMBO10 (n = 3) |
All cohorts (N = 17) |
|
AUC0−t, h·ng/mL | 220 (86.4) | 384 (109) | 460 (451) | 117 (105) | 241 (63.6) | 239 (206) | 13,700 (7130) |
AUCΤ, h·ng/mL | 227 (83.1) | 384 (109) | 542 (101) | 141 (96.7)† | 241 (63.7) | 291 (101)a | 13,800 (4940)b |
C max, ng/mL | 51.0 (23.2) | 93.4 (55.7) | 42.9 (37.1) | 13.3 (7.48) | 25.5 (8.03) | 14.0 (12.9) | 2840 (1430) |
T max, h | 1.00 (0.48-5.53) |
1.02 (0.47-3.00) |
4.00 (1.00-7.17) |
1.98 (0.92-6.50) |
1.95 (0.47-4.00) |
4.00 (2.00-7.17) |
1.95 (0.48-7.17) |
t 1/2, h | 4.74 (2.17) | 5.37 (0.697) | 11.0 (9.12)a | 7.95 (2.53)c | 7.43 (1.07) | 15.0 (12.5)a | 4.29 (2.42)b |
Data are mean (SD) for AUC0−t, AUCΤ, Cmax, and t1/2 and median (range) for Tmax.
a n = 2; bn = 16; cn = 8.
Abbreviations: AUC0−t, area under the plasma concentration-time curve from time zero to infinity; AUCΤ, area under the plasma concentration-time curve during a dosing interval; Cmax, maximum plasma drug concentration; t1/2, terminal half-life; Tmax, time to Cmax.
Anti-Tumor Activity
Overall, the confirmed ORR and DCR were 10% (95% CI, 1.2-31.7) and 85% (95% CI, 62.1-96.8), respectively. Most patients (n = 15, 75%) achieved stable disease as their best response to treatment. Two patients (both on the COMBO5 dose) had a partial response. No complete responses were observed. A summary of response outcomes is shown in Table 6. There were 18 patients evaluable for RNF43 mutation status, 8 of whom had RNF43 mutant. There did not appear to be any correlation between the best percentage change from baseline in sum of longest diameters and RNF43 mutation status (based on local testing; data not shown).
Table 6.
Response outcomes.
COMBO5 (n = 10) |
COMBO7.5 (n = 6) |
COMBO10 (n = 4) |
All patients (N = 20) |
|
---|---|---|---|---|
Best overall response | ||||
Complete response | 0 | 0 | 0 | 0 |
Partial response | 2 (20.0) | 0 | 0 | 2 (10.0) |
Stable disease | 7 (70.0) | 4 (66.7) | 4 (100.0) | 15 (75.0) |
Progressive disease | 1 (10.0) | 0 | 0 | 1 (5.0) |
Unknown | 0 | 2 (33.3) | 0 | 2 (10.0) |
Overall response rate | 2 (20.0) | 0 | 0 | 2 (10.0) |
95% CI | 2.5-55.6 | — | — | 1.2-31.7 |
Disease control rate | 9 (90.0) | 4 (66.7) | 4 (100.0) | 17 (85.0) |
95% CI | 55.5-99.7 | 22.3-95.7 | 39.8-100.0 | 62.1-96.8 |
Data are n (%) unless indicated otherwise.
Discussion
This phase Ib trial of the triplet combination of WNT974 + encorafenib + cetuximab showed the safety of the combination was not established in patients with BRAF V600E-mutant KRAS WT mCRC harboring Wnt pathway mutations. Taken together, these data suggest further development of the triplet combination would require additional on-treatment monitoring to address safety concerns, including prophylactic treatments to prevent bone resorption, potentially hindering further development of the combination. Hence, enrollment was discontinued and after a safety and efficacy evaluation, following the consensus agreement of the study investigators, the phase II study was not initiated.
In this single-arm trial of encorafenib + cetuximab + WNT974 in the same type of patient population, bone-related complications were among the most common and the most serious treatment-related toxicities observed. The combination treatment was associated with a higher incidence of bone-related toxicities, including increased susceptibility to fractures, bone pain, and hypercalcemia, compared with doublet treatment (encorafenib + cetuximab) or WNT974 monotherapy. In the phase III BEACON trial for BRAF V600E-mutant mCRC, there were no reports of osteopenia/osteoporosis, hypercalcemia, or bone fracture (of any grade) among the 218 patients treated with encorafenib + cetuximab or in patients treated with encorafenib + binimetinib + cetuximab.25 In another phase Ib trial evaluating encorafenib + cetuximab with or without alpelisib, hypophosphatemia was very common in the encorafenib + cetuximab arm, with 19% of patients experiencing grade 3/4.26 In a previous phase I trial of WNT974, 6 patients experienced 7 bone-related toxicities, of which 5 were considered related to the study treatment. Suspected treatment-related bone-associated toxicities included, osteoporosis, pathological fracture, osteopenia, and spinal fracture.23 Hypophosphatemia can be a cause of bone toxicity also due to decreased bone mineralization (prolonged hypophospatemia over months to years leads to rickets and osteomalacia), and this may be what was contributing to the additive effect on bone toxicity observed, even if on their own encorafenib and cetuximab did not lead to clinically detectable bone fractures.
The role of the Wnt pathway in bone homeostasis, repair, and regeneration is well known.27 A study in mice showed that bisphosphonates can mitigate acute bone loss due to PORCN inhibition.28 However, in clinical practice, phase I trials of Wnt pathway inhibitors in which patients received prophylactic bisphosphonates have shown mixed results; in patients with advanced solid tumors, ipafricept (a Wnt pathway inhibitor) was well tolerated and alterations in bone remodeling were mostly reversible with bisphosphonate therapy. Alternatively, trials of vantictumab (an anti-Frizzled antibody) + chemotherapy for the treatment of patients with HER2-negative breast cancer or pancreatic cancer closed early due to high rates of bone fracture.29-31
The PK data for WNT974 and LHA333 should be interpreted with caution. Ninety-one of 446 WNT974 and LHA333 samples included in the calculation of the WNT974 and LHA333 PK parameters were analyzed after the validated storage stability time of 345 days. None of the samples analyzed for encorafenib were affected by stability issues. The study was not adequately powered to assess specific biomarker-related hypotheses. However, the molecular landscape should be evaluated in this patient population in future trials to better understand how best to treat these patients.
The 2 patients who achieved a response had a duration of response of 106 and 291 days, respectively. Neither patient had any specific characteristics that appeared to enhance their chance of response. The patient with the longest duration of response experienced an SAE of grade 2 hypercalcemia, which resolved, followed by a grade 3 AE that was not suspected to be treatment-related but resulted in permanent discontinuation of the study treatment.
It is unknown whether RSPO fusions contributed to differential responses. It has been shown that RSPO fusions occur less frequently compared with RNF43 mutations but Wnt expression levels were not evaluated.7 Wnt expression levels were not evaluated, and its role as a true oncogenic driver is not yet fully understood.32
Conclusion
Although the triplet combination in this study was limited by toxicities, the rationale for combining encorafenib and cetuximab with a Wnt pathway inhibitor holds promise. Future clinical trials involving Wnt pathway inhibitors must mitigate the risk of bone-related toxicities by intensively monitoring bone homeostasis and including bone protective measurements in their clinical trial protocols and patient selection. While novel combination approaches beyond inhibition of BRAF and EGFR are warranted for patients with BRAF V600E-mutant mCRC, it appears that the addition of a PORCN inhibitor to this combination resulted in excessive toxicity that did not justify further study.
Supplementary Material
Acknowledgments
We thank the patients and their families, as well as the participating study teams, for making this study possible. Medical writing and editorial assistance were provided by Raya Mahbuba of Caudex and were funded by Pfizer.
Contributor Information
Josep Tabernero, Vall d’Hebron Hospital Campus, Vall d’Hebron Institute of Oncology (VHIO), UVic-UCC, IOB-Quiron, Barcelona, Spain.
Eric Van Cutsem, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium.
Elena Garralda, START Madrid, Hospital Universitario HM Sanchinarro, Madrid, Spain.
David Tai, Division of Medical Oncology, National Cancer Centre Singapore, Singapore.
Filippo De Braud, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
Ravit Geva, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.
Mark T J van Bussel, Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
Katia Fiorella Dotti, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
Elena Elez, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain.
María J de Miguel, START Madrid, Hospital Universitario HM Sanchinarro, Madrid, Spain.
Kevin Litwiler, Pfizer, Boulder, CO, USA.
Danielle Murphy, Pfizer, La Jolla, CA, USA.
Michelle Edwards, Pfizer, New York, NY, USA.
Van Karlyle Morris, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Funding
This study was sponsored by Array BioPharma in collaboration with Novartis. Array BioPharma was acquired by Pfizer in July 2019. For V.K.M., this work was supported by the generous philanthropic contributions to the Khalifa Scholars Programs (from the Khalifa Bin Zayed Al Nahyan Foundation), the Advanced Scholar Program (from the CG Johnson Foundation), and the NIH/NCI (award number K12 CA088084).
Conflict of Interest
Josep Tabernero Array Biopharma, AstraZeneca, Avvinity, Bayer, Boehringer Ingelheim, Chugai, Daiichi Sankyo, F. Hoffmann-La Roche Ltd, Genentech Inc., HalioDX SAS, Hutchison MediPharma International, Ikena Oncology, IQVIA, Lilly, Menarini, Merck Serono, Merus, MSD, Mirati, NeoPhore, Novartis, Ona Therapeutics, Orion Biotechnology, Peptomyc, Pfizer, Pierre Fabre, Samsung Bioepis, Sanofi, Seattle Genetics, Scandion Oncology, Servier, Taiho, Tessa Therapeutics, and TheraMyc (consulting/advisory); Imedex, Medscape Education, MJH Life Sciences, PeerView Institute for Medical Education, and Physicians Education Resource (PER) (educational collaboration); Amgen Inc., Array Biopharma Inc., AstraZeneca Pharmaceuticals LP, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Debiopharm International SA, F. Hoffmann-La Roche Ltd, Genentech Inc., HalioDX SAS, Hutchison MediPharma International, Janssen-Cilag SA, MedImmune, Menarini, Merck Health KGAA, Merck Sharp & Dohme, Merus NV, Mirati, Novartis Farmacéutica SA, Pfizer, Pharma Mar, Sanofi Aventis Recherche & Développement, Servier, Taiho Pharma USA Inc., Spanish Association Against Cancer Scientific Foundation, and Cancer Research UK (research funding). Eric Van Cutsem Array, AstraZeneca, Bayer, Biocartis, Bristol Myers Squibb, Celgene, Eli Lilly, Ipsen, MSD, Merck KGaA, Novartis, Pierre Fabre, Roche, Servier, Sirtex, and Taiho (consulting/advisory); Amgen, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eli Lilly, Ipsen, Merck, Merck KGaA, Novartis, Roche, and Servier (research funding). Elena Garralda Alkermes, Anaveon, Boehringer, Bristol Myers Squibb, F. Hoffman/La Roche, Ingelheim, Janssen Global Services, MabDiscovery, Roche/Genentech, Seattle Genetics, TFS, and Thermo Fisher (consulting/advisory); AstraZeneca, BeiGene, Novartis, Roche, Taiho, and Thermo Fisher (research funding). David Tai F. Hoffman/La Roche. Filippo De Braud AstraZeneca, EMD Serono, Incyte, Menarini, MSD, NMS Nerviano Medical Science, Novartis, Roche, and Sanofi (consulting/advisory). Ravit Geva AstraZeneca, Bayer, Boehringer Ingelheim, BOL Pharma, EISAI, MSD, Novartis, and Roche (consulting/advisory); Bristol Myers Squibb, Janssen, Lilly, Medison, Merck, MSD, Novartis, Pfizer, Roche, and Takeda (honoraria); Novartis (research funding); Bayer, Bristol Myers Squibb, Medison, Merck, BOL Pharma, and Pyxis (travel, accommodations and/or expenses). Mark T.J. van Bussel reported no financial relationships. Katia Fiorella Dotti reported no financial relationships. Elena Elez Amgen, Array BioPharma, Bayer, Merck Serono, MSD, Pierre Fabre, Roche, Sanofi, and Servier (consulting/advisory). María J. de Miguel MSD, PharmaMar, Roche, Novartis, AbbVie, Array, Eisai, Faron, and Sanofi (research funding); MSD, Janssen, Roche (speakers’ bureau). Kevin Litwiler Volestra (consultant/advisory role); Pfizer (employee); OnKure Therapeutics (employee, leadership role, stockholder). Danielle Murphy Pfizer (employee, ownership interests). Michelle Edwards Pfizer (former employee and stockholder); Arvinia (employee and stockholder). Van Karlyle Morris Array, Axiom Healthcare Strategies, Bayer, Bicara Therapeutics, BioMedical Insights, Boehringer Ingelheim, Incyte, Pfizer, and Servier (consulting/advisory); BioNTech AG, Boehringer Ingelheim, Bristol Myers Squibb, EMD Serono, Immatics, Novartis, and Pfizer (research funding).
Author Contributions
Conception/design/methodology: J.T., K.L., D.M., V.K.M. Provision of study material or patients: J.T., M.T.J.vB., E.E. Collection and/or assembly of data, investigation: J.T., D.T., M.T.J.vB., E.E., K.L., V.K.M. Data analysis and interpretation: J.T., E.V.C., K.L., D.M. Manuscript writing: J.T., E.V.C., D.T., M.T.J.vB., K.F.D., D.M., M.E., V.K.M. Final approval of manuscript: all authors.
Data Availability
The data underlying this article are available in the article and in its online supplementary material. Upon request, and subject to review, Pfizer will provide the data supporting the findings of this study. Subject to certain criteria, conditions, and exceptions, Pfizer may also provide access to the related individual de-identified participant data. See https://www.pfizer.com/science/clinical-trials/trial-data-and-results for more information.
References
- 1. World Health Organization. Factsheets: Cancer. 2022. https://www.iarc.who.int/featured-news/colorectal-cancer-awareness-month-2022/
- 2. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487: 330-337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Schatoff EM, Leach BI, Dow LE.. Wnt Signaling and colorectal cancer. Curr Colorectal Cancer Rep. 2017;13:101-110. https://doi.org/ 10.1007/s11888-017-0354-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Bond CE, McKeone DM, Kalimutho M, et al. RNF43 and ZNRF3 are commonly altered in serrated pathway colorectal tumorigenesis. Oncotarget. 2016;7:70589-70600. https://doi.org/ 10.18632/oncotarget.12130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Fischer MM, Yeung VP, Cattaruzza F, et al. RSPO3 antagonism inhibits growth and tumorigenicity in colorectal tumors harboring common Wnt pathway mutations. Sci Rep. 2017;7(1):15270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Kim SY, Kim TW.. Current challenges in the implementation of precision oncology for the management of metastatic colorectal cancer. ESMO Open. 2020;5:e000634. https://doi.org/ 10.1136/esmoopen-2019-000634 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Seshagiri S, Stawiski EW, Durinck S, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488:660-664. https://doi.org/ 10.1038/nature11282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Giannakis M, Hodis E, Jasmine Mu X, et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat Genet. 2014;46(12):1264-1266. https://doi.org/ 10.1038/ng.3127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Hao HX, Jiang X, Cong F.. Control of Wnt receptor turnover by R-spondin-ZNRF3/RNF43 signaling module and its dysregulation in cancer. Cancers. 2016;8:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Djanani A, Eller S, Öfner D, Troppmair J, Maglione M.. The role of BRAF in metastatic colorectal carcinoma-past, present, and future. Int J Mol Sci. 2020;21:9001. https://doi.org/ 10.3390/ijms21239001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949-954. [DOI] [PubMed] [Google Scholar]
- 12. Mauri G, Bonazzina E, Amatu A, et al. The evolutionary landscape of treatment for BRAF(V600E) mutant metastatic colorectal cancer. Cancers. 2021;13:137. https://doi.org/ 10.3390/cancers13010137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kopetz S, Desai J, Chan E, et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33:4032-4038. https://doi.org/ 10.1200/jco.2015.63.2497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100-103. https://doi.org/ 10.1038/nature10868 [DOI] [PubMed] [Google Scholar]
- 15. Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2(3):227-235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med. 2019;381(17):1632-1643. [DOI] [PubMed] [Google Scholar]
- 17. Krejci P, Aklian A, Kaucka M, et al. Receptor tyrosine kinases activate canonical WNT/β-catenin signaling via MAP kinase/LRP6 pathway and direct β-catenin phosphorylation. PLoS One. 2012;7:e35826. https://doi.org/ 10.1371/journal.pone.0035826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Punt CJ, Koopman M, Vermeulen L.. From tumour heterogeneity to advances in precision treatment of colorectal cancer. Nat Rev Clin Oncol. 2017;14(4):235-246. [DOI] [PubMed] [Google Scholar]
- 19. Liu J, Pan S, Hsieh MH, et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc Natl Acad Sci USA. 2013;110(50):20224-20229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Bagheri M, Tabatabae Far MA, Mirzaei H, Ghasemi F.. Evaluation of antitumor effects of aspirin and LGK974 drugs on cellular signaling pathways, cell cycle and apoptosis in colorectal cancer cell lines compared to oxaliplatin drug. Fundam Clin Pharmacol. 2020;34(1):51-64. [DOI] [PubMed] [Google Scholar]
- 21. Han T, Schatoff EM, Murphy C, et al. R-Spondin chromosome rearrangements drive Wnt-dependent tumour initiation and maintenance in the intestine. Nat Commun. 2017;8:15945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Jiang X, Hao HX, Growney JD, et al. Inactivating mutations of RNF43 confer Wnt dependency in pancreatic ductal adenocarcinoma. Proc Natl Acad Sci USA. 2013;110(31):12649-12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Rodon J, Argiles G, Connolly RM, et al. Phase 1 study of single-agent WNT974, a first-in-class Porcupine inhibitor, in patients with advanced solid tumours. Br J Cancer. 2021;125:28-37. https://doi.org/ 10.1038/s41416-021-01389-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Tabernero J, Geel RV, Guren TK, et al. Phase 2 results: encorafenib (ENCO) and cetuximab (CETUX) with or without alpelisib (ALP) in patients with advanced BRAF-mutant colorectal cancer (BRAFm CRC). J Clin Oncol. 2016;34:3544-3544. https://doi.org/ 10.1200/jco.2016.34.15_suppl.354427573652 [DOI] [Google Scholar]
- 25. Tabernero J, Grothey A, Van Cutsem E, et al. Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E-mutant metastatic colorectal cancer: updated survival results and subgroup analyses from the BEACON study. J Clin Oncol. 2021;39:273-284. https://doi.org/ 10.1200/jco.20.02088 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. van Geel R, Tabernero J, Elez E, et al. A Phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov. 2017;7(6):610-619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Houschyar KS, Tapking C, Borrelli MR, et al. Wnt pathway in bone repair and regeneration - what do we know so far. Front Cell Dev Biol. 2018;6:170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Madan B, McDonald MJ, Foxa GE, et al. Bone loss from Wnt inhibition mitigated by concurrent alendronate therapy. Bone Res. 2018;6(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Jimeno A, Gordon M, Chugh R, et al. A first-in-human phase I study of the anticancer stem cell agent ipafricept (OMP-54F28), a decoy receptor for Wnt ligands, in patients with advanced solid tumors. Clin Cancer Res. 2017;23(24):7490-7497. [DOI] [PubMed] [Google Scholar]
- 30. Davis SL, Cardin DB, Shahda S, et al. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer. Invest New Drugs. 2020;38(3):821-830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Diamond JR, Becerra C, Richards D, et al. Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer. Breast Cancer Res Treat. 2020;184:53-62. https://doi.org/ 10.1007/s10549-020-05817-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Parsons MJ, Tammela T, Dow LE.. WNT as a driver and dependency in cancer. Cancer Discov. 2021;11(10):2413-2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data underlying this article are available in the article and in its online supplementary material. Upon request, and subject to review, Pfizer will provide the data supporting the findings of this study. Subject to certain criteria, conditions, and exceptions, Pfizer may also provide access to the related individual de-identified participant data. See https://www.pfizer.com/science/clinical-trials/trial-data-and-results for more information.