Purpose: This phase I dose-escalation study investigated the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), safety, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary clinical activity of CH5132799.

Experimental Design: Patients with metastatic solid tumors were eligible for the study. CH5132799 was administered orally once daily or twice daily in 28-day cycles.

Results: Thirty-eight patients with solid tumors received CH5132799 at 2 to 96 mg once daily or 48 to 72 mg twice daily. The MTD was 48 mg on the twice-daily schedule but was not reached on the once daily schedule. DLTs were grade 3 elevated liver function tests (LFT), grade 3 fatigue, grade 3 encephalopathy, grade 3 diarrhea, and grade 3 diarrhea with grade 3 stomatitis; all DLTs were reversible. Most drug-related adverse events were grade 1/2. Diarrhea (34%) and nausea (32%) were the most common events. Mean Cmax and AUC0-24 in steady state at MTD were 175 ng/mL and 1,550 ng·h/mL, respectively, consistent with efficacious exposure based on preclinical modeling. Reduction in SUVmax with [18F] fluorodeoxyglucose positron emission tomography (FDG-PET) was observed in 5 of 7 patients at MTD. A patient with PIK3CA-mutated clear cell carcinoma of the ovary achieved a partial response by GCIG CA125 criteria and further, a heavily pretreated patient with triple-negative breast cancer had marked improvement in her cutaneous skin lesions lasting six cycles.

Conclusion: CH5132799 is well tolerated at the MTD dose of 48 mg twice daily. At this dose, the drug had a favorable PK and PD profile and preliminary evidence of clinical activity. Clin Cancer Res; 20(23); 5908–17. ©2014 AACR.

Translational Relevance

CH5132799 is an oral pan-PI3 kinase inhibitor designed to target both alpha and beta isoforms of PI3K, which are frequently deregulated in cancer due to activating mutations in the oncogene PIK3CA or loss of function of the tumor-suppressor gene, PTEN. The dosing schedule was to be determined by toxicity, PK, and PD parameters and started with a once daily schedule followed if necessary by a twice-daily schedule. The trial started with CH5132799 administered orally once a day but a formal MTD was not reached at 96 mg, however, as there were no significant increments in AUC above a dose of 56 mg once daily and p-AKT showed recovery at 24 hours. Therefore, the dosing schedule was modified to include twice a day dosing. A dose of 48 mg twice daily was declared the recommended phase II dose, at which dose a patient with PIK3CA-mutant clear cell ovarian cancer responded.

The intracellular phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway regulates cellular functions such as cell survival, proliferation, growth, apoptosis, protein synthesis, and glucose metabolism (1–6). Of the three classes of PI3K (I to III), class IA is implicated most in cancer (5). Class IA PI3K heterodimers comprise a p85-regulatory and a p110-catalytic subunit with several isoforms (5). Mutation or amplification of PIK3CA, the gene encoding the catalytic subunit of PI3K (p110α), promotes oncogenic activation of the PI3K pathway and occurs frequently in human cancers such as ovarian cancer and breast cancer (1, 6–12). Phosphatase and tensin homolog (PTEN), a key negative regulator of AKT, can be inactivated via mutations, downregulation, or loss of protein expression, and is associated with tumorigenesis in prostate, gastric, and other cancers (1, 13–19). Moreover, it is likely that PI3K pathway activation is associated with resistance to both chemotherapy (20–22) and targeted agents (23–26). Selective inhibition of the PI3K–AKT–mTOR pathway in cancer represents a promising therapeutic approach and has been the focus of significant research efforts, including the clinical development of novel agents targeting this pathway (1, 19, 27–37).

CH5132799 (Chugai Pharmaceutical Co Ltd.) is an oral PI3K inhibitor with specific and potent activity against class I PI3Ks, especially demonstrated in wild-type and mutant PI3Kα isoforms, and PI3Kγ, at nanomolar concentrations (38). CH5132799 has no inhibitory activity against class III PI3K, or mTOR (38). In vitro experiments demonstrated a strong antiproliferative effect of CH5132799 on human cancer cell lines with alterations in the PI3K pathway (38, 39). In vivo, CH5132799 demonstrated significant antitumor activity in human tumor xenograft models with PIK3CA mutations, with good correlation between CH5132799 exposure and inhibition of PI3K signaling (39).

The primary objective of this first-in-human, phase I, dose-escalation study was to determine the MTD of CH5132799 using a continuous oral schedule in patients with advanced solid tumors. Secondary objectives included the characterization of CH5132799 PK, and the PD profile of PI3K inhibition in tumor and in surrogate tissues such as peripheral blood samples, and by [18F] fluorodeoxyglucose positron emission tomography (FDG-PET).

Study population

Patients had been diagnosed with advanced solid tumors that were not amenable or were refractory to standard therapy. Patients of ages ≥18 years with an Eastern Cooperative Oncology Group (ECOG) performance status of 0 to 2 and adequate bone marrow, renal, hepatic, and cardiac function (see Supplementary data for detailed inclusion and exclusion criteria) and a life expectancy of ≥12 weeks were enrolled.

This study was approved by an independent ethics committee (The Royal Marsden Research Ethics Committee, London, United Kingdom) and conducted in accordance with the Declaration of Helsinki and Good Clinical Practice (GCP). Written informed consent was obtained from all patients before carrying out any study-related procedures.

Study design and CH5132799 dose escalation

This open-label dose-escalation study was conducted at four centers. Before the first treatment cycle, CH5132799 was administered as a single oral dose followed by a 5- to 7-day washout (run-in period). A classic “3+3” design was used for dose escalation with once daily to the early patient cohorts, and then twice daily to others, continuously in 4-week cycles. Dose escalation was determined by the nature and grade/severity of toxicities.

The primary objective was to determine the MTD of CH5132799 based on DLTs observed during the run-in period and first 4-week cycle. The MTD was defined as the highest dose level at which no more than 1 of 6 patients experienced a DLT. A starting dose of 2 mg was chosen on the basis of the highest nonsevere toxic dose in a non-rodent species and the severely toxic dose (10% lethal dose) in rat, which means 7.8-fold and 30-fold safety margins were applied to the two metabolically/kinetically relevant animal species, respectively.

Assessments

Medical history and demographic data were collected at baseline. Physical examination, monitoring of vital signs, and other safety assessments were performed throughout the study. All toxicities were documented using Common Terminology Criteria for Adverse Events (CTCAE) V4.03 (40). DLTs were defined as grade ≥3, nonhematologic toxicity despite adequate treatment; grade 4, neutropenia lasting ≥7 days; and febrile neutropenia, grade 4 thrombocytopenia lasting ≥7 days or requiring a platelet transfusion.

Tumor response was assessed according to Response Evaluation Criteria In Solid Tumors (RECIST; version 1.1) with imaging at baseline and every two cycles (41).

PK and PD

Plasma PK samples were collected on cycle 0, day 1, followed by cycle 1, days 1, 8, 15, and 22. Plasma concentrations of CH5132799 were measured by a validated LC-MS/MS assay method (Chugai Pharmaceutical Co. Ltd.; data on file) and PK parameters calculated by noncompartmental analysis with first-order oral absorption (WinNonlin Version 5.3 and Phoenix WinNonlin Version 6.1; Pharsight Corporation). The CH5132799-related inhibition of AKT phosphorylation (pAKT) was studied in platelet-rich plasma (PRP). Blood for PRP samples was collected at 0 (predose), 1, 2, 6, 24, 48, and 72 hours postdose on day 1 of the run-in period (cycle 0, day 1) and at 0 (predose) on cycle 1, day 15. Blood samples were collected into BD Vacutainer sodium citrate coagulation tubes and centrifuged at 200 ×g at 4°C for 15 minutes. The isolated PRP layer was incubated with PhosSTOP (Roche) to stabilize the phosphorylation signals and then lysed using the Cell Lysis Buffer (Cell Signaling Technology) containing phenylmethane sulfonyl fluoride (Sigma-Aldrich) and then snap-frozen on dry ice. PD analysis with PRP was performed by The Institute of Cancer Research. All PRP samples were analyzed for the levels of phosphorylated and total forms of PD biomarker AKT by a Meso Scale Discovery electrochemiluminescent (ECL) assay. The assay was modified into a GCP-compliant quantitative assay for AKT by inclusion of a standard curve of recombinant active AKT protein on every plate. The quality, accuracy, and precision of the assays were monitored using quality control (QC) samples created by spiking three known quantities of recombinant AKT into 10% human plasma.

Biopsies were performed at screening and at cycle 1, day 15. Flash frozen tumor biopsies were homogenized using a micro tissue grinder in the Cell Lysis Buffer (w/v) containing PhosSTOP. The tumor lysates were centrifuged to remove debris and the protein concentration measured using the BCA assay (Pierce). Tumor biopsy samples were analyzed for the levels of phosphorylated and total forms of PD biomarker AKT by a quantitative ICR-modified GCP compliant Meso Scale Discovery ECL assay. FDG-PET imaging was performed within 28 days before the first dosing of CH5132799, cycle 1, day 8, and cycle 3, day 1. Lesions with the highest degree of FDG uptake were selected for quantitative analysis and a circular/spherical region of interest drawn. A SUVmax was measured for each selected lesion and the delta change in SUVmax was calculated. Tumor biopsies were taken after FDG-PET scanning to avoid interference of biopsy on FDG uptake.

Detection of mutations

PIK3CA/KRAS/BRAF mutations were studied with the OncoCarta panel v1.0 and detected by mass ARRAY System.

Statistical analyses

Descriptive statistics were used for the analysis of PK, PD, safety, and tumor response data.

Thirty-eight patients received at least one dose of CH5132799 (Table 1), of whom 23 received CH5132799 once daily (2–96 mg/day). The decision to dose twice daily was made as there had been no DLTs and no significant increase in drug concentrations at doses above 56 mg once daily along with PD measures, i.e., recovery of p-AKT after 12 hours. Subsequently, 15 further patients then received CH5132799 twice daily (96–144 mg/day). Overall, patients received a median of 2 cycles (range, 0–6), with a median duration of 52 days of treatment (range, 1–164 days). Archival tumor samples were available in 30 patients. PIK3CA mutations were found in 10% (3 of 30) of the samples studied (Table 1).

Table 1.

Patient demographics and clinical characteristics

CharacteristicsTotal patients (N = 38)
Sex 
 Male 10 (26%) 
 Female 28 (74%) 
Age, y 
 Median 58.5 
 Range 41–76 
Race 
 Asian 3 (8%) 
 Black 3 (8%) 
 White 32 (84%) 
Baseline ECOG performance status 
 0 12 (32%) 
 1 25 (66%) 
 2 1 (3%) 
Prior anticancer therapies, median (range) 3 (1–8) 
Primary tumor site and mutational status 
 Breast 10 (26%) 
 No mutation 
 Unknown mutational status 
Ovarian 6 (16%) 
KRAS G12D 
PIK3CA H1047R 
  No mutation 
  Unknown mutational status 
Esophageal and esophageal-gastric adenocarcinoma 5 (13%) 
KRAS G12D 
EGFR S768I 
  No mutation 
Duodenal and large intestine 4 (11%) 
KRAS G12A 
KRAS G12D and PIK3CA E545K 
 No mutation 
Gastrica 2 (5%) 
Lunga 2 (5%) 
Endometriala 1 (3%) 
Vaginaa 1 (3%) 
Myometriuma 1 (3%) 
Vulvaa 1 (3%) 
Bladdera 1 (3%) 
Pancreasa 1 (3%) 
Prostatea 1 (3%) 
Unknown primary origin 1 (3%) 
PIK3CA E545K 
CharacteristicsTotal patients (N = 38)
Sex 
 Male 10 (26%) 
 Female 28 (74%) 
Age, y 
 Median 58.5 
 Range 41–76 
Race 
 Asian 3 (8%) 
 Black 3 (8%) 
 White 32 (84%) 
Baseline ECOG performance status 
 0 12 (32%) 
 1 25 (66%) 
 2 1 (3%) 
Prior anticancer therapies, median (range) 3 (1–8) 
Primary tumor site and mutational status 
 Breast 10 (26%) 
 No mutation 
 Unknown mutational status 
Ovarian 6 (16%) 
KRAS G12D 
PIK3CA H1047R 
  No mutation 
  Unknown mutational status 
Esophageal and esophageal-gastric adenocarcinoma 5 (13%) 
KRAS G12D 
EGFR S768I 
  No mutation 
Duodenal and large intestine 4 (11%) 
KRAS G12A 
KRAS G12D and PIK3CA E545K 
 No mutation 
Gastrica 2 (5%) 
Lunga 2 (5%) 
Endometriala 1 (3%) 
Vaginaa 1 (3%) 
Myometriuma 1 (3%) 
Vulvaa 1 (3%) 
Bladdera 1 (3%) 
Pancreasa 1 (3%) 
Prostatea 1 (3%) 
Unknown primary origin 1 (3%) 
PIK3CA E545K 

aNo mutation or mutational status not known.

DLTs

DLTs were observed in 5 of 38 evaluable patients who received CH5132799 twice daily and completed the first cycle (Table 2). Of 7 patients at 48 mg twice daily, 1 patient with hepatocellular carcinoma experienced grade 3 elevated transaminases. Although the patient had liver metastases with elevated LFTs (grade 2) at baseline, a causal link to CH5132799 could not be excluded because clear evidence of disease progression in liver metastasis was not observed. At the next dose level, 2 of 3 patients receiving 72 mg twice daily had DLTs. One had grade 3 fatigue that was resolved following cessation of CH5132799 and she restarted CH5132799 with reduced dose when fatigue had been resolved and she did not experience the event later on, whereas the other developed grade 3 posterior reversible encephalopathy syndrome (PRES) that was presented with seizures. The diagnosis was made on characteristic MRI findings and the symptoms resolved on cessation of CH5132799 as well as instigation of supportive care without restarting CH5132799. Two of 5 patients receiving the intermediate dose of 56 mg twice daily had grade 3 diarrhea, the other grade 3 diarrhea with grade 3 stomatitis, which resolved following cessation of CH5132799 and instigation of supportive treatment; in both cases, study treatment was then restarted at a reduced dose without further DLTs. Therefore, the MTD and recommended phase II dose (RP2D) of CH5132799 administered orally was established at 48 mg twice daily.

Table 2.

DLTs seen on the study

CohortDoseNDLT
7b 48 mg BID Grade 3 elevated LFT at cycle 1 day 8 
72 mg BID Grade 3 cerebral encephalopathy at cycle 1, day 14 
   Grade 3 fatigue at cycle 1 day 8 
56 mg BID Grade 3 diarrhea 
   Grade 3 diarrhea and stomatitis (<75% of the total scheduled dose) 
CohortDoseNDLT
7b 48 mg BID Grade 3 elevated LFT at cycle 1 day 8 
72 mg BID Grade 3 cerebral encephalopathy at cycle 1, day 14 
   Grade 3 fatigue at cycle 1 day 8 
56 mg BID Grade 3 diarrhea 
   Grade 3 diarrhea and stomatitis (<75% of the total scheduled dose) 

Abbreviation: BID, twice daily.

Safety

The most common AEs were diarrhea, nausea, stomatitis, fatigue, and rash, which occurred in 34%, 32%, 29%, 29%, and 24% of patients, respectively (Table 3). Diarrhea was predominantly grade 1 or 2, with no grade 4 diarrhea reported. Four patients experienced grade 3 diarrhea (96 mg once daily, n = 2; 56 mg twice daily, n = 2). One patient experienced grade 3 nausea (96 mg once daily). No grade ≥3 gastrointestinal AEs occurred in the cohort who received CH5132799 at the R2PD (48 mg twice daily). Hyperglycemia was predicted because of the mechanism of action of CH5132799. Five patients experienced grade ≥3 hyperglycemia in the twice-daily dosing schedule, suggesting a dose-dependent effect (48 mg twice daily, n = 1; 56 mg twice daily, n = 2; 72 mg twice daily; n = 2 including one grade 4). No episodes of diabetic ketoacidosis occurred and hyperglycemia was controlled with oral metformin as required. No drug-related deaths were reported.

Table 3.

Treatment-related toxicities occurring in ≥10% patients by CH5132799 dose level

QDBID
AE2 mg (n = 3)4 mg (n = 3)8 mg (n = 3)16 mg (n = 3)32 mg (n = 4)56 mg (n = 3)96 mg (n = 4)48 mg (n = 7)56 mg (n = 5)72 mg (n = 3)
Grade1–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–4Total (n = 38)
Diarrhea              13 (34%) 
Nausea            12 (32%) 
Stomatitis               11 (29%) 
Fatigue            11 (29%) 
Rash               9 (24%) 
Decreased appetite                8 (21%) 
Vomiting                  6 (16%) 
Dry skin                 6 (16%) 
Hyperglycemia                  5 (13%) 
Anemia                5 (13%) 
Abdominal pain                 4 (11%) 
QDBID
AE2 mg (n = 3)4 mg (n = 3)8 mg (n = 3)16 mg (n = 3)32 mg (n = 4)56 mg (n = 3)96 mg (n = 4)48 mg (n = 7)56 mg (n = 5)72 mg (n = 3)
Grade1–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–41–23–4Total (n = 38)
Diarrhea              13 (34%) 
Nausea            12 (32%) 
Stomatitis               11 (29%) 
Fatigue            11 (29%) 
Rash               9 (24%) 
Decreased appetite                8 (21%) 
Vomiting                  6 (16%) 
Dry skin                 6 (16%) 
Hyperglycemia                  5 (13%) 
Anemia                5 (13%) 
Abdominal pain                 4 (11%) 

PK

Following oral administration of a single dose of CH5132799, plasma concentrations peaked with a Tmax at 2.60 hours and t1/2 of 10.2 hours at the MTD (Fig. 1A). Dose proportionality of AUC was shown in the dose range of 2 to 56 mg (Fig. 2B). Steady state was reached by cycle 1, day 8 at the latest with little subsequent accumulation; there was no further increase in AUC at higher doses. At the MTD of 48 mg twice daily, mean Cmax and AUClast were 172 ng/mL and 1,270 ng·h/mL after a single dose, respectively. Mean Cmax and AUC0–24h were 175 ng/mL and 1,550 ng·h/mL at steady state, respectively (Table 4).

Figure 1.

Pharmacokinetic parameters of CH5132799. A, plasma concentration of CH5132799 following a single administration across the dose range of 2 to 96 mg. B, the correlation of the AUC of CH5132799 and dose. Horizontal lines, effective AUC calculated to cause TGI (tumor grown inhibition) of 50% and 90% of the KPL4 and HCT116 xenograft models.

Figure 1.

Pharmacokinetic parameters of CH5132799. A, plasma concentration of CH5132799 following a single administration across the dose range of 2 to 96 mg. B, the correlation of the AUC of CH5132799 and dose. Horizontal lines, effective AUC calculated to cause TGI (tumor grown inhibition) of 50% and 90% of the KPL4 and HCT116 xenograft models.

Close modal
Figure 2.

PD parameters of CH5132799. A, mean percentage change of phosphorylated AKT normalized to total AKT compared with pretreatment control values, 2 hours following a single dose of CH5132799. B, the temporal course of plasma concentration of CH5132799 and the percentage change of p-AKT normalized to total AKT compared with pretreatment control values following a single dose of the recommended phase II dose of CH5132799 (48 mg). C, percentage change of p-AKT normalized to total AKT in tumor biopsies when compared with pretreatment samples. D, SUVmax changes on FDG-PET scans in patients who had evaluable pre- and posttreatment FDG-PET scans.

Figure 2.

PD parameters of CH5132799. A, mean percentage change of phosphorylated AKT normalized to total AKT compared with pretreatment control values, 2 hours following a single dose of CH5132799. B, the temporal course of plasma concentration of CH5132799 and the percentage change of p-AKT normalized to total AKT compared with pretreatment control values following a single dose of the recommended phase II dose of CH5132799 (48 mg). C, percentage change of p-AKT normalized to total AKT in tumor biopsies when compared with pretreatment samples. D, SUVmax changes on FDG-PET scans in patients who had evaluable pre- and posttreatment FDG-PET scans.

Close modal
Table 4.

Summary of PK of CH5132799 in patients following oral administration

Single doseRepeat dose
RegimenAUClasta (ng•h/mL)Cmax (ng/mL)t1/2 (h)AUC0–24 hb (ng•h/mL)Cmax (ng/mL)Accumulation ratioc
2 mg (QD) 53.0 (3) 10.0 (3) 7.52 (3) 48.8 (3) 9.92 (3) 0.912 (3) 
4 mg (QD) 112 (3) 17.2 (3) 8.30 (3) 167 (3) 22.5 (3) 1.54 (3) 
8 mg (QD) 133 (3) 41.1 (3) 23.7 (3) 159 (2) 46.3 (2) 1.22 (2) 
16 mg (QD) 883 (1) 113 (1) 13.7 (1) 621 (2) 100 (3) 0.972 (1) 
32 mg (QD) 859 (4) 165 (4) 12.2 (4) 1,740 (2) 345 (3) 1.57 (2) 
56 mg (QD) 969 (3) 163 (3) 14.5 (3) 748 (3) 184 (3) 1.07 (3) 
96 mg (QD) 1,650 (4) 206 (4) 23.2 (3) 691 (3) 103 (3) 0.640 (3) 
48 mg (BID) 1,270 (4) 172 (5) 10.2 (4) 1,550 (5)b 175 (5) 1.10 (2) 
56 mg (BID) 3,030 (5) 428 (5) 16.3 (5) 5,940 (2)b 331 (3) 1.72 (2) 
72 mg (BID) 2,480 (3) 265 (3) 18.3 (3) 5,580 (1)b 631(1) 2.25 (1) 
Single doseRepeat dose
RegimenAUClasta (ng•h/mL)Cmax (ng/mL)t1/2 (h)AUC0–24 hb (ng•h/mL)Cmax (ng/mL)Accumulation ratioc
2 mg (QD) 53.0 (3) 10.0 (3) 7.52 (3) 48.8 (3) 9.92 (3) 0.912 (3) 
4 mg (QD) 112 (3) 17.2 (3) 8.30 (3) 167 (3) 22.5 (3) 1.54 (3) 
8 mg (QD) 133 (3) 41.1 (3) 23.7 (3) 159 (2) 46.3 (2) 1.22 (2) 
16 mg (QD) 883 (1) 113 (1) 13.7 (1) 621 (2) 100 (3) 0.972 (1) 
32 mg (QD) 859 (4) 165 (4) 12.2 (4) 1,740 (2) 345 (3) 1.57 (2) 
56 mg (QD) 969 (3) 163 (3) 14.5 (3) 748 (3) 184 (3) 1.07 (3) 
96 mg (QD) 1,650 (4) 206 (4) 23.2 (3) 691 (3) 103 (3) 0.640 (3) 
48 mg (BID) 1,270 (4) 172 (5) 10.2 (4) 1,550 (5)b 175 (5) 1.10 (2) 
56 mg (BID) 3,030 (5) 428 (5) 16.3 (5) 5,940 (2)b 331 (3) 1.72 (2) 
72 mg (BID) 2,480 (3) 265 (3) 18.3 (3) 5,580 (1)b 631(1) 2.25 (1) 

NOTE: Mean value and number (noted in parenthesis) are represented for each pharmacokinetic parameter.

Abbreviations: BID, twice daily; Cmax, maximum concentration measured; QD, once daily; t1/2: elimination half-life.

aAUClast in single dose represented AUC0-72h;

bAUC0-24h in BID regimen of repeat dose were calculated on the basis of the data from 0 to 12 hours (AUC0-12h);

cAccumulation ratio represents AUC0-24h/0-12h in repeat dose/AUC0-24h/0-12h in single dose.

PD

Significant inhibition of PI3K signaling was observed in PRP, with reduction of AKT phosphorylation at doses of 32 mg and above (Fig. 2A). The temporal relationship between reduction in phosphorylation of AKT and dosing administration with CH5132799 suggested target engagement for less than 24 hours (Fig. 2B). This observation contributed to the decision to increase dosing frequency to twice daily.

Pre- and posttreatment tumor biopsies were optional and the minimum number to be conducted during the study was not based on statistical assumptions. Tumor biopsy samples were collected before and after CH5132799 from 3 patients (one patient each on 56 mg once daily, 96 mg once daily, and 56 mg twice daily). There was a more than 50% reduction in normalized p-AKT levels in 2 of the 3 patients (Fig. 2C).

A decrease in FDG avidity between baseline and cycle 1, day 8 was observed in 74% of patients (17 of 23) who underwent serial PET imaging (Fig. 2D). It was interesting that 5 of 7 patients at the RP2D who had pre-and post-(cycle1, day 8) treatment PET scans showed a reduction in maximum standardized uptake value (SUVmax); 2 had a reduction in SUVmax of 55% and 44%.

There was no correlation between the PD changes in the limited number of biopsies, PET responses, or PRP inhibition.

Efficacy

There were no RECIST partial or complete responses, but 1 patient with PIK3CA H1047R-mutated clear cell ovarian cancer had a GCIG CA125 response having been treated at 48 mg twice daily and remained on study for 6 cycles (Fig. 3A). A second patient with heavily pretreated triple-negative breast cancer bearing no PIK3CA or AKT mutations who was treated at the 72 mg twice daily dose level had marked symptomatic and objective improvement in her cutaneous skin lesions and remained on treatment for 6 cycles (Fig. 3B). Disease stabilization was seen in 8 patients up to week 16 (approximately 4 cycles), including 2 patients with PIK3CA mutation.

Figure 3.

Clinical efficacy of CH5132799. A, FDG-PET-CT showing reduction in uptake of tracer in pelvic masses and a reduction of CA125 following treatment with CH5132799. The patient had a clear cell ovarian cancer that harbored a PIK3CA mutation. B, representative skin lesions, before and after treatment with CH5132799, of a patient with triple-negative breast cancer.

Figure 3.

Clinical efficacy of CH5132799. A, FDG-PET-CT showing reduction in uptake of tracer in pelvic masses and a reduction of CA125 following treatment with CH5132799. The patient had a clear cell ovarian cancer that harbored a PIK3CA mutation. B, representative skin lesions, before and after treatment with CH5132799, of a patient with triple-negative breast cancer.

Close modal

We report the first-in-human study of an oral PI3K class I inhibitor CH5132799. No DLTs occurred at the once daily schedule, but the twice-daily schedule was selected on the basis of the duration of target inhibition observed in platelet-rich plasma and the lack of dose-dependent incremental elevation in concentrations of drug above 56 mg once daily. The twice-daily schedule of CH5132799 was well tolerated and showed evidence of clinical and PD activity with a RP2D of 48 mg twice daily.

The most frequently observed toxicities were gastrointestinal, including diarrhea, nausea, and stomatitis. These have been described previously in studies of PI3K, AKT, and m-TOR inhibitors (28, 30–38). Reversible grade 3 LFT elevation was observed in 1 of 7 patients treated at the MTD of 48 mg twice daily. This patient with hepatocellular carcinoma had liver metastases and had previously undergone hepatic resection; therefore, the event was possibly caused by disease progression and/or overload of a liver with insufficient metabolic capability. No other cases of drug-related LFT abnormalities were noted, although they have been described in trials of other PI3K inhibitors (30–32, 34, 42). Of note, we observed one case of PRES at a dose above the MTD. This has not been reported previously in the trials of other drugs in this class; nevertheless, it raised the possibility that CH5132799 crosses the blood–brain barrier, which may be of future relevance in the treatment of patients with intracerebral malignancies. Mood alterations have been described as AEs in with other PI3 kinase inhibitors (30, 42), however, was not seen in this trial. Although skin-related toxicities have been frequently observed in the trials of other PI3K inhibitors (28–30, 32, 33, 35–37, 42–44), CH5132799 was associated with only mild-to-moderate skin toxicities at a lower frequency. On-target toxicity such as hyperglycemia was observed with CH5132799, as with other PI3K, AKT, and m-TOR inhibitors (28–30, 35–37, 43, 44) and was not associated with ketoacidosis. The hyperglycemia was well controlled by metformin. The favorable toxicity profile of CH5132799 may make it suitable to combine with other targeted agents such as epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) or MAPK/ERK kinase (MEK) inhibitors.

The PK data show that it was possible to attain CH5132799 concentrations in patients that had been efficacious in xenograft models (39). The AUC increased proportionally with dose from 2 to 56 mg. The PD data indicated target modulation using phosphorylation of AKT at Sr473 as a proof-of-mechanism biomarker. Levels of p-AKT were reduced in PRP consistently at dose levels 8 to 96 mg. The maximal inhibition was approximately 50% compared with baseline and this was achieved at dose levels of 32 mg. p-AKT levels in PRP frequently recovered by 24 hours following the treatment. The duration of PD biomarker changes in normal tissue and the fact that there was no significant further reduction in p-AKT levels after the 32 mg cohort, along with the fact that the AUC of CH5132799 did not rise significantly above 56 mg, led us to explore a twice-weekly schedule, despite not reaching an conventional MTD at 96 mg on the once-daily dosing cohorts. At the recommended phase II dose of 48 mg twice daily, levels of p-AKT were reduced in PRP and in a limited number of posttreatment tumor biopsies. There was a reduction in levels of p-AKT (Ser473) in 2 of 3 post-treatment tumor biopsies at the higher dose levels, but there was considerable variation in reduction between patients (Fig. 2C). The degree of p-AKT reduction in the tumor from patients was less than what was seen in xenograft models (39), however, this could reflect different platforms to assess AKT phosphorylation and other factors such as intratumoral heterogeneity.

Further evidence of tumor target modulation was observed with FDG-PET scans with all patients at recommended dose. All 5 patients scanned at the RP2D had a reduction in SUVmax and two of them achieved a PET response by PERCIST criteria (45). Although the number of patients is too small to draw definitive conclusions, these PD data are highly encouraging. There was no correlation between the PD changes in the limited number of biopsies and PET responses or PRP inhibition. The FDG PET was done pretreatment and on day 8 of treatment, whereas tumor biopsies were performed pretreatment and day 15 of treatment, while the detailed p-AKT studies in PRP were done on day 1 of treatment. The small number of biopsies and the disparities in the days when tests were conducted could have led to the lack of correlation between p-AKT levels in tumor, PRP, and changes in SUVmax on PET.

The 8 study participants (21%) showing a best response of stable disease by RECIST included 2 participants who had clear clinical responses. In reports of other PI3K, AKT, and mTOR inhibitors as single agent in patients with solid tumors, disease stabilization was observed, but only a few cases of radiologic response have been reported (28–32, 34–36, 43). Therefore, there was clinical evidence as well as PD activity for CH5132799. These results indicated that CH5132799 was comparable with other PI3K, AKT, and m-TOR inhibitors in terms of single-agent activity.

Three patients were found to have PIK3CA mutations. Although there were no objective radiologic responses, it was interesting that 1 of these patients (with clear cell ovarian cancer) had a GCIG-defined partial response in her CA125 tumor marker. Another patient with triple-negative breast cancer had a visible response in cutaneous metastases. This patient with breast cancer did not have a PIK3CA mutation, although triple-negative breast cancer was known frequently to diseases associated with other perturbations in the PI3K pathway (46). Preclinical models suggest that PIK3CA mutations predict for sensitivity to PI3K inhibitors as either single agents (47, 48) or in combination (49). This was also seen with preclinical studies of CH5132799 (39). However, the clinical correlation between PIK3CA mutations and PI3K inhibitors to this has not been fully realized (50) in the clinic, and it is possible that other activating mutations (51) or intratumoral heterogeneity (52) may play a role.

We conclude that CH5132799 at RP2D of 48 mg twice daily is well tolerated and attains drug concentrations that show evidence of achieving target engagement in both normal tissues and tumor. CH5132799 has clinical, as well as PD activity, with evidence of proof of mechanism from a clinical response in 1 patient with PIK3CA-mutant ovarian cancer. The favorable toxicity profile and activity observed with single-agent CH5132799 suggests that it is suitable for combination with other targeted therapies and for exploration in subsets of patients with cancers harboring predefined mutations.

U. Banerji reports receiving a commercial research grant from Chugai Pharma Europe Ltd. No potential conflicts of interest were disclosed by the other authors.

Conception and design: S. Blagden, K. Noguchi, R. Shiokawa, M. Inatani, J. Prince, J. Spicer, U. Banerji

Development of methodology: S. Blagden, A. Olmin, S. Decordova, K. Noguchi, R. Shiokawa, M. Inatani, J. Prince, J. Spicer, U. Banerji

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Blagden, A. Olmin, D. Josephs, C. Stavraka, A. Zivi, D.J. Pinato, A. Anthoney, K. Swales, L. Pope, K. Noguchi, R. Shiokawa, M. Inatani, C. Twelves, J. Spicer, U. Banerji

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Olmin, D.J. Pinato, S. Decordova, K. Swales, L. Pope, K. Noguchi, R. Shiokawa, M. Inatani, C. Twelves, J. Spicer, U. Banerji

Writing, review, and/or revision of the manuscript: S. Blagden, A. Olmin, C. Stavraka, A. Zivi, D.J. Pinato, A. Anthoney, K. Swales, R. Riisnaes, L. Pope, K. Noguchi, R. Shiokawa, M. Inatani, J. Prince, K. Jones, C. Twelves, J. Spicer, U. Banerji

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C. Stavraka, K. Swales, R. Riisnaes, K. Noguchi, R. Shiokawa, M. Inatani

Study supervision: S. Blagden, C. Stavraka, J. Prince, C. Twelves, U. Banerji

The authors thank all the patients who took part in this study, together with their families and caretakers.

This study was funded by Chugai Pharmaceutical Co. Ltd. Infrastructure; support was provided by a Cancer Research UK Joint Phase I Clinical Core Grant (grant number C51/A6883) to The Institute of Cancer Research and The Royal Marsden. The recruiting sites acknowledge infrastructural funding from the Experimental Cancer Medicine Centres and NIHR Biomedical Research Centre initiatives.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Courtney
KD
,
Corcoran
RB
,
Engelman
JA
. 
The PI3K pathway as drug target in human cancer.
J Clin Oncol
2010
;
28
:
1075
83
.
2.
Liu
P
,
Cheng
H
,
Roberts
TM
,
Zhao
JJ
. 
Targeting the phosphoinositide 3-kinase pathway in cancer.
Nat Rev Drug Discov
2009
;
8
:
627
44
.
3.
Katso
R
,
Okkenhaug
K
,
Ahmadi
K
,
White
S
,
Timms
J
,
Waterfield
MD
. 
Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancer.
Annu Rev Cell Dev Biol
2001
;
17
:
615
75
.
4.
Cantley
LC
. 
The phosphoinositide 3-kinase pathway.
Science
2002
;
296
:
1655
7
.
5.
Engelman
JA
,
Luo
J
,
Cantley
LC
. 
The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism.
Nat Rev Genet
2006
;
7
:
606
19
.
6.
Wong
KK
,
Engelman
JA
,
Cantley
LC
. 
Targeting the PI3K signaling pathway in cancer.
Curr Opin Genet Dev
2010
;
20
:
87
90
.
7.
Shayesteh
L
,
Lu
Y
,
Kuo
WL
,
Baldocchi
R
,
Godfrey
T
,
Collins
C
, et al
PIK3CA is implicated as an oncogene in ovarian cancer.
Nat Genet
1999
;
21
:
99
102
.
8.
Samuels
Y
,
Wang
Z
,
Bardelli
A
,
Silliman
N
,
Ptak
J
,
Szabo
S
, et al
High frequency of mutations of the PIK3CA gene in human cancers.
Science
2004
;
304
:
554
.
9.
Bader
AG
,
Kang
S
,
Zhao
L
,
Vogt
PK
. 
Oncogenic PI3K deregulates transcription and translation.
Nat Rev Cancer
2005
;
5
:
921
9
.
10.
Levine
DA
,
Bogomolniy
F
,
Yee
CJ
,
Lash
A
,
Barakat
RR
,
Borgen
PI
, et al
Frequent mutation of the PIK3CA gene in ovarian and breast cancers.
Clin Cancer Res
2005
;
11
:
2875
8
.
11.
Nosho
K
,
Kawasaki
T
,
Ohnishi
M
,
Suemoto
Y
,
Kirkner
GJ
,
Zepf
D
, et al
PIK3CA mutation in colorectal cancer: relationship with genetic and epigenetic alterations.
Neoplasia
2008
;
10
:
534
41
.
12.
Zhao
L
,
Vogt
PK
. 
Helical domain and kinase domain mutations in p110alpha of phosphatidylinositol 3-kinase induce gain of function by different mechanisms.
Proc Natl Acad Sci U S A
2008
;
105
:
2652
7
.
13.
Li
J
,
Yen
C
,
Liaw
D
,
Podsypanina
K
,
Bose
S
,
Wang
SI
, et al
PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science
1997
;
275
:
1943
7
.
14.
Cantley
LC
,
Neel
BG
. 
New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway.
Proc Natl Acad Sci U S A
1999
;
96
:
4240
5
.
15.
Carracedo
A
,
Pandolfi
PP
. 
The PTEN-PI3K pathway: of feedbacks and cross-talks.
Oncogene
2008
;
27
:
5527
41
.
16.
Salvesen
HB
,
MacDonald
N
,
Ryan
A
,
Jacobs
IJ
,
Lynch
ED
,
Akslen
LA
, et al
PTEN methylation is associated with advanced stage and microsatellite instability in endometrial carcinoma.
Int J Cancer
2001
;
91
:
22
6
.
17.
Carver
BS
,
Chapinski
C
,
Wongvipat
J
,
Hieronymus
H
,
Chen
Y
,
Chandarlapaty
S
, et al
Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer.
Cancer Cell
2011
;
19
:
575
86
.
18.
Song
MS
,
Salmena
L
,
Pandolfi
PP
. 
The functions and regulation of the PTEN tumour suppressor.
Nat Rev Mol Cell Biol
2012
;
13
:
283
96
.
19.
Samuels
Y
,
Ericson
K
. 
Oncogenic PI3K and its role in cancer.
Curr Opin Oncol
2006
;
18
:
77
82
.
20.
Brognard
J
,
Clark
AS
,
Ni
Y
,
Dennis
PA
. 
Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation.
Cancer Res
2001
;
61
:
3986
97
.
21.
Hu
L
,
Hofmann
J
,
Lu
Y
,
Mills
GB
,
Jaffe
RB
. 
Inhibition of phosphatidylinositol 3′-kinase increases efficacy of paclitaxel in in vitro and in vivo ovarian cancer models.
Cancer Res
2002
;
62
:
1087
92
.
22.
Carden
CP
,
Stewart
A
,
Thavasu
P
,
Kipps
E
,
Pope
L
,
Crespo
M
, et al
The association of PI3 kinase signaling and chemoresistance in advanced ovarian cancer.
Mol Cancer Ther
2012
;
11
:
1609
17
.
23.
Berns
K
,
Horlings
HM
,
Hennessy
BT
,
Madiredjo
M
,
Hijmans
EM
,
Beelen
K
, et al
A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer.
Cancer Cell
2007
;
12
:
395
402
.
24.
Faber
AC
,
Li
D
,
Song
Y
,
Liang
MC
,
Yeap
BY
,
Bronson
RT
, et al
Differential induction of apoptosis in HER2 and EGFR addicted cancers following PI3K inhibition.
Proc Natl Acad Sci U S A
2009
;
106
:
19503
8
.
25.
Sartore-Bianchi
A
,
Martini
M
,
Molinari
F
,
Veronese
S
,
Nichelatti
M
,
Artale
S
, et al
PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies.
Cancer Res
2009
;
69
:
1851
7
.
26.
O'Brien
NA
,
Browne
BC
,
Chow
L
,
Wang
Y
,
Ginther
C
,
Arboleda
J
, et al
Activated phosphoinositide 3-kinase/AKT signaling confers resistance to trastuzumab but not lapatinib.
Mol Cancer Ther
2010
;
9
:
1489
502
.
27.
Engelman
JA
. 
Targeting PI3K signalling in cancer: opportunities, challenges and limitations.
Nat Rev Cancer
2009
;
9
:
550
62
.
28.
Baird
RD
,
Kristeleit
RS
,
Sarker
D
,
Olmos
D
,
Sandhu
SK
,
Yan
Y
, et al
A phase I study evaluating the pharmacokinetics (PK) and pharmacodynamics (PD) of the oral pan-phosphoinositide-3 kinase (PI3K) inhibitor GDC-0941.
J Clin Oncol
2010
;
28
(
15 suppl
):
2613
.
29.
Wagner
AJ
,
Von Hoff
DH
,
LoRusso
PM
,
Tibes
R
,
Mazina
KE
,
Ware
JA
, et al
A first-in-human phase I study to evaluate the pan-PI3K inhibitor GDC-0941 administered QD or BID in patients with advanced solid tumors.
J Clin Oncol
2009
;
27
(
15 suppl
):
3501
.
30.
Bendell
JC
,
Rodon
J
,
Burris
HA
,
de Jonge
M
,
Verweij
J
,
Birle
D
, et al
Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors.
J Clin Oncol
2012
;
30
:
282
90
.
31.
Hong
DS
,
Bowles
DW
,
Falchook
GS
,
Messersmith
WA
,
George
GC
,
O'Bryant
CL
, et al
A multicenter phase I trial of PX-866, an oral irreversible phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors.
Clin Cancer Res
2012
;
18
:
4173
82
.
32.
Brana
I
,
LoRusso
P
,
Baselga
J
,
Heath
EI
,
Patnaik
A
,
Gendreau
S
, et al
A phase I dose-escalation study of the safety, pharmacokinetics (PK), and pharmacodynamics of XL765 (SAR245409), a PI3K/TORC1/TORC2 inhibitor administered orally to patients (pts) with advanced malignancies.
J Clin Oncol
2010
;
28
(
15 suppl
):
3030
.
33.
Edelman
G
,
Bedell
C
,
Shapiro
G
,
Pandya
SS
,
Kwak
EL
,
Scheffold
C
, et al
A phase I dose-escalation study of XL147 (SAR245408), a PI3K inhibitor administered orally to patients (pts) with advanced malignancies.
J
Clin Oncol
2010
:
28
(
15 suppl
):
3004
.
34.
Furman
RR
,
Byrd
JC
,
Flinn
IW
,
Coutre
SE
,
Benson
DM
,
Brown
JR
, et al
Interim results from a phase I study of CAL-101, a selective oral inhibitor of phosphatidylinositol 3-kinase p110d isoform, in patients with relapsed or refractory hematologic malignancies.
J Clin Oncol
2010
:
29
(
15 suppl
):
3032
.
35.
Moreno Garcia
V
,
Baird
RD
,
Shah
KJ
,
Basu
B
,
Tunariu
N
,
Blanco
M
, et al
A phase I study evaluating GDC-0941, an oral phosphoinositide-3 kinase (PI3K) inhibitor, in patients with advanced solid tumors or multiple myeloma.
J Clin Oncol
2011
;
29
(
15 suppl
):
3021
.
36.
Wagner
AJ
,
Bendell
JC
,
Dolly
S
,
Morgan
JA
,
Ware
JA
,
Fredrickson
J
, et al
A first-in-human phase I study to evaluate GDC-0980, an oral PI3K/mTOR inhibitor, administered QD in patients with advanced solid tumors.
J Clin Oncol
2011
;
29
(
15 suppl
):
3020
.
37.
Yap
TA
,
Yan
L
,
Patnaik
A
,
Fearen
I
,
Olmos
D
,
Papadopoulos
K
, et al
First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors.
J Clin Oncol
2011
;
29
:
4688
95
.
38.
Ohwada
J
,
Ebiike
H
,
Kawada
H
,
Tsukazaki
M
,
Nakamura
M
,
Miyazaki
T
, et al
Discovery and biological activity of a novel class I PI3K inhibitor, CH5132799.
Bioorg Med Chem Lett
2011
;
21
:
1767
72
.
39.
Tanaka
H
,
Yoshida
M
,
Tanimura
H
,
Fujii
T
,
Sakata
K
,
Tachibana
Y
, et al
The selective class I PI3K inhibitor CH5132799 targets human cancers harboring oncogenic PIK3CA mutations.
Clin Cancer Res
2011
;
17
:
3272
81
.
40.
National Cancer Institute Cancer Therapy Evaluation Program Common Terminology Criteria for Adverse Events, version 4.03. 14; June 2010. Available from
: http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5×7.pdf.
41.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).
Eur J Cancer
2009
;
45
:
228
47
.
42.
Rodon
J
,
Bendell
J
,
Abdul
RA
,
Homji
N
,
Trandafir
L
,
Quadt
C
, et al
P3-16-01: Safety Profile and Clinical Activity of Single-Agent BKM120, a Pan-Class I PI3K Inhibitor, for the Treatment of Patients with Metastatic Breast Carcinoma.
Cancer Res
2011
(
24 suppl
):
P3-16-01
.
43.
Hudis
C
,
Swanton
C
,
Janjigian
YY
,
Lee
R
,
Sutherland
S
,
Lehman
R
, et al
A phase 1 study evaluating the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors.
Breast Cancer Res
2013
;
15
:
R110
.
44.
Burris
H
,
Rodon
J
,
Sharma
S
,
Herbst
RS
,
Tabernero
J
,
Infante
JR
, et al
First-in-human phase I study of the oral PI3K inhibitor BEZ235 in patients (pts) with advanced solid tumors.
J Clin Oncol
2010
;
28
(
15 suppl
):
3005
.
45.
Wahl
RL
,
Jacene
H
,
Kasamon
Y
,
Lodge
MA
. 
From RECIST to PERCIST: Evolving Considerations for PET response criteria in solid tumors.
J Nucl Med
2009
;
50
Suppl 1
:
122S
50S
.
46.
Gordon
V
,
Banerji
S
. 
Molecular pathways: PI3K pathway targets in triple-negative breast cancers.
Clin Cancer Res
2013
;
19
:
3738
44
.
47.
O'Brien
C
,
Wallin
JJ
,
Sampath
D
,
GuhaThakurta
D
,
Savage
H
,
Punnoose
EA
, et al
Predictive biomarkers of sensitivity to the phosphatidylinositol 3′ kinase inhibitor GDC-0941 in breast cancer preclinical models.
Clin Cancer Res
2010
;
16
:
3670
83
.
48.
Fritsch
C
,
Huang
A
,
Chatenay-Rivauday
C
,
Schnell
C
,
Reddy
A
,
Liu
M
, et al
Characterization of the novel and specific PI3Kalpha inhibitor NVP-BYL719 and development of the patient stratification strategy for clinical trials.
Mol Cancer Ther
2014
;
13
:
1117
29
.
49.
Spoerke
JM
,
O'Brien
C
,
Huw
L
,
Koeppen
H
,
Fridlyand
J
,
Brachmann
RK
, et al
Phosphoinositide 3-kinase (PI3K) pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models.
Clin Cancer Res
2012
;
18
:
6771
83
.
50.
Janku
F
,
Hong David
S
,
Fu
S
,
Piha-Paul Sarina
A
,
Naing
A
,
Falchook Gerald
S
, et al
Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors.
Cell Rep
2014
;
6
:
377
87
.
51.
Ganesan
P
,
Janku
F
,
Naing
A
,
Hong
DS
,
Tsimberidou
AM
,
Falchook
GS
, et al
Target-based therapeutic matching in early-phase clinical trials in patients with advanced colorectal cancer and PIK3CA mutations.
Mol Cancer Ther
2013
;
12
:
2857
63
.
52.
Gerlinger
M
,
Rowan
AJ
,
Horswell
S
,
Larkin
J
,
Endesfelder
D
,
Gronroos
E
, et al
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing.
N Engl J Med
2012
;
366
:
883
92
.