Skip to main content
Erschienen in: Investigational New Drugs 1/2013

Open Access 01.02.2013 | PHASE I STUDIES

A phase I/II, non-randomized, feasibility/safety and efficacy study of the combination of everolimus, cetuximab and capecitabine in patients with advanced pancreatic cancer

verfasst von: Sil Kordes, Dick J. Richel, Heinz-Josef Klümpen, Mariëtte J. Weterman, Arnoldus J. W. M. Stevens, Johanna W. Wilmink

Erschienen in: Investigational New Drugs | Ausgabe 1/2013

Summary

Background Improvements in knowledge of molecular mechanisms in cancer are the basis for new studies combining chemotherapy with targeted drugs. Inhibition of the epidermal growth factor receptor (EGFR) by erlotinib or cetuximab has limited or no activity, respectively, in pancreatic cancer. The crosstalk between EGFR and mammalian target of rapamycin (mTOR) pathways is a potential mechanism of resistance; therefore we conducted a study to explore safety and efficacy of multiple pathway inhibition by cetuximab and everolimus in combination with capecitabine. Methods Safety and efficacy of fixed standard dose cetuximab in combination with various dose levels of everolimus (5–10 mg/day) and capecitabine (600–800 mg/m2 bid, 2 weeks every 3 weeks) were investigated in a phase I/II study in patients with advanced pancreatic cancer. The primary endpoint was objective response. Results Sixteen patients were treated in the phase I part at two dose levels. Mucositis, rash and hand-foot syndrome were dose-limiting toxicities. Dose level 1 (everolimus 5 mg/day, capecitabine 600 mg/m2 bid for 2 weeks every 3 weeks and cetuximab 250 mg/m2 weekly) was considered the maximum tolerated dose (MTD). Of 31 patients in the phase II part, partial response was documented in two patients (6.5%) and five (16.1%) had stable disease. Median overall survival was 5.0 months (CI 3.1–6.8). Conclusion The schedule of capecitabine, everolimus and cetuximab resulted in considerable epidermal and mucosal toxicities and prevented escalation to optimal dose levels. Because of toxicity and low efficacy this treatment combination cannot be recommended for treatment in pancreatic cancer patients.

Introduction

Pancreatic cancer patients have one of the worst prognoses among all cancer types with an overall survival rate of less than 5% [1]. Since the publication of Burris in 1997 gemcitabine is still considered as the standard treatment for most patients with pancreatic cancer [2]. Several randomized clinical trials comparing gemcitabine in combination with other chemotherapeutic drugs have not resulted in survival improvement [3].
Moving away from the paradigm that combination therapy must be gemcitabine based, a strategy that has invariably failed, Conroy et al. recently demonstrated in a randomized clinical trial a significant survival advantage with the gemcitabine free combination chemotherapy regimen FOLFIRINOX as compared with gemcitabine [4]. Although patients were highly selected and toxicity was considerable, this trial opens the way to new treatment strategies in advanced pancreatic cancer.
Significant improvements in our knowledge of the molecular mechanisms involved in cancer development and progression, and the availability of drugs interfering with aberrant activity in various signaling pathways, have subsequently resulted in numerous clinical trials combining conventional chemotherapy with various targeted drugs. The EGFR/MAPK and PI3K/Akt/mTOR pathways are often dysregulated and considerable evidence supports the important role of these pathways in the biology of pancreatic cancer [5, 6]. Several targets in these pathways are potential candidates to achieve inhibition of aberrant signaling. Erlotinib, a tyrosine kinase EGFR inhibitor, was one of the first FDA approved tyrosine kinase inhibitors. In a randomized clinical trial in pancreatic cancer patients, erlotinib in combination with gemcitabine induced a statistically significant improvement in survival, although the two weeks survival benefit was considered clinically not meaningful [7]. Targeting EGFR with the monoclonal antibody cetuximab in combination with gemcitabine failed to demonstrate a survival advantage[8]. mTOR is an important signaling molecule in the PI3K pathway and inhibition of mTOR could inhibit tumor growth in pancreatic cancer xenograft models [9]. However, in a clinical study no benefit was demonstrated using the mTOR inhibitor everolimus as a single agent in second line [10]. Possible explanations for the relative insensitivity to drugs targeting only one aberrant molecule is the heterogeneous molecular pathogenesis of pancreatic cancer leading to deviant activation of multiple signaling pathways and the intensive crosstalk between these pathways [11, 12]. Although some tumors with specific crucial mutations are sensitive to mono-targeted therapies, such as gastrointestinal stromal tumor and imatinib, for most cancer types including pancreatic cancer this is not the case [13, 14]. Exploration of drug combinations targeting multiple pathways is therefore an interesting strategy to overcome drug resistance. Rational targets for this combined approach in pancreatic cancer are EGFR and mTOR, leading to synergistic anticancer activity as has been demonstrated in pre-clinical models [1520].
Therefore we explored in the present study the feasibility and efficacy of a triple drug combination consisting of cetuximab, everolimus and capecitabine in patients with advanced pancreatic cancer. In an earlier phase I study we demonstrated that everolimus (10 mg daily) in combination with capecitabine (1000 mg/m2 BID) was a safe and tolerable regimen [21]. In the present study gemcitabine was replaced by capecitabine because gemcitabine in combination with everolimus induced severe bone-marrow toxicity already at the gemcitabine dose level of 600 mg/m [22]. The failure of gemcitabine based combination regimens was also taken in to account. The monoclonal antibody cetuximab instead of erlotinib was chosen because of potential pharmacokinetic interactions between erlotinib and mTOR inhibitors (at the level of cytochrome P (CYP) metabolizing enzymes) [23, 24].

Patients and methods

Study design and statistics

This multicenter open-label phase I/II trial consisted of two parts: the phase I part was traditionally designed with interpatient dose escalation in cohorts of three to six patients with the primary end point of protocol-defined dose limiting toxicity (DLT) and Maximum Tolerated Dose (MTD) [25]. The phase II part was designed to determinate the efficacy and feasibility of the combination of everolimus, capecitabine and cetuximab. Primary endpoint of this part of the study was response rate. Patients were defined as responders when a complete response (CR) or partial response (PR) by response evaluation criteria in solid tumors (RECIST) 1.0 was seen. Secondary endpoints were time to treatment failure (TTF), overall survival (OS), one-year survival rate and the toxicity profile according to NCI–CTC v3.0. TTF and OS were calculated by the Kaplan-Meier method, measured from the date of treatment initiation to the date of documented progression and death of any cause, respectively. All analyses were conducted on an intention-to-treat basis and were performed using SPSS version 18.0.2.
The phase II part was designed in two stages (Simon two-stage optimal design) with an early stopping rule for efficacy: if no objective responses were to be observed within the first 14 patients treated at the MTD, the trial was to be halted, because this event (0/14) has a probability of <0.05 if the true response rate is 0.20.
The study was conducted according to the ethical principles of the Declaration of Helsinki and Good Clinical Practice and was approved by health authorities and the independent ethics committee of the Academic Medical Center Amsterdam. The trial is registered on the USA NCI Web site www.​ClinicalTrials.​gov (NCT01077986)

Patients

Patients with cytological or histological confirmed locally advanced or metastatic adenocarcinoma of the pancreas were eligible. Further inclusion criteria comprised an Eastern cooperative oncology group/World health organization (ECOG/WHO) performance status of 0, 1 or 2, measurable lesions on CT according to RECIST 1.0 criteria (for the phase II part of the study), age eighteen years of age or older and a life-expectancy of at least three months. Patients had to be mentally, physically and geographically able to undergo treatment and follow-up. Adequate renal, liver and bone marrow function was necessary. Laboratory values accompanied hereby were serum creatinine <150 μmol/L, bilirubin <1.5x upper limit of laboratory normal (ULN), aspartate aminotransferase and alanine aminotransferase <2.5x ULN or <5.0x in case of liver metastasis, white blood cell count >3.0x109, platelets >100x109, respectively.
Patients were not eligible if they had previous treatment with an mTOR inhibitor. Other exclusion criteria included pregnancy and lactation, clinical or radiological evidence of central nervous system metastasis at time of enrollment, known hypersensitivity to everolimus or other rapamycins or to its excipients, any severe and/or uncontrolled medical conditions, such as clinically significant heart conditions or myocardial infarction in 6 months prior to randomization, uncontrolled diabetes as defined by fasting glucose above 1.5x ULN, active or uncontrolled infection, serious liver disease and severely impaired lung function, or a serious concomitant systemic disorder that would compromise the safety of the patient, at the discretion of the investigator. Written informed consent was obtained from each patient.

Treatment

Treatment was administered in a 3-week regimen consisting of continuous daily oral everolimus, weekly cetuximab, and capecitabine for 14 days followed by 7 days rest. For the phase I part, dose escalations were performed for everolimus and capecitabine, according to Table 1. Cetuximab was given at a fixed dose of 250 mg/m2, with a start-up dose of 400 mg/m2.
Table 1
Dose escalation levels phase I
Dose level
Everolimus (mg)
Capecitabine (bid mg/m2)
Cetuximab (mg)
−1
5
400
250
1
5
600
250
2
10
600
250
3
10
800
250
bid twice daily
If one of three patients experienced dose-limiting toxicity (DLT), three more patients were included at the same dose level. If two or more patients experienced DLT, the previous dose level was considered the MTD. All patients of the phase II part of the study were treated at the MTD.
DLTs were defined as any of the following adverse events as defined by the common terminology criteria for adverse events version 3.0 (CTCAE) in the first two cycles: grade 4 neutropenia lasting > 5 days or febrile neutropenia grade 3; grade 4 thrombocytopenia and grade ≥ 3 red cell count; grade ≥ 2 vomiting and grade ≥ 3 of any other toxicity despite supportive treatment, except rash, which was defined as DLT at grade 4.
In the phase II part, dose modifications were predefined for each drug. Everolimus dose was reduced in case of grade 3 toxicity or recurrence of grade 2 non-hematological toxicity or thrombocytopenia after interruption. Everolimus was discontinued in case of grade 4 toxicity or recurrence of grade 3 hematological toxicity after dose reduction. Capecitabine had to be withheld in case of toxicity grade ≥ 2 until recovery to grade ≤ 1. Dose modifications were dependent on severity and frequency of toxicity, as defined in the protocol. Cetuximab had to be delayed for up to two consecutive infusions in case of grade ≥3 skin toxicity whereas doxycyclin 100 mg daily and local metronidazole treatment was initiated. The same dose level was restarted if toxicity resolved to grade ≤ 2, with continuation of doxycyclin treatment. At second or third recurrence of grade 3 toxicity, dose was reduced to 200 mg/m2 and 150 mg/m2, respectively. Cetuximab was discontinued in case of withholding more than 2 infusions, fourth recurrence of skin toxicity grade ≥ 3, or an allergic/hypersensitivity reaction grade ≥ 3. Treatment was continued until unacceptable toxicity, disease progression, withdrawal of informed consent by the patient or any other reason why continuation was not in the best interest of the patient. Response assessment by CT-scan (RECIST 1.0) was done at baseline and every 9 weeks during active treatment.

Results

Patients

In total 43 patients were enrolled between February 2009 and June 2010. Three patients were excluded from analysis because of major violation of the inclusion criteria; one patient in the phase I part of the study received eight cycles of treatment, while in retrospect no pancreatic cancer cells were seen in pathology. Two patients in the phase II part experienced rapid deterioration between signing informed consent and start of treatment.
Table 2 summarizes the baseline characteristics of all 40 eligible patients, separately for each dose level.
Table 2
Patient demographics and disease characteristics
Characteristic
Phase IIa
DL2
N = 31 (%)
N = 9 (%)
Median Age
57.9
61
 Range
39–78
45–69
Gender
 Male
13 (42)
6 (67)
 Female
18 (58)
3 (33)
WHO performance status
 0
16 (52)
5 (56)
 1
10 (32)
3 (33)
 2
5 (16)
1 (11)
Stage of disease
 Locally advanced
4 (13)
1 (11)
 Metastatic
27 (87)
8 (89)
Localization of primary
 Head
23 (74)
8 (89)
 Tail/corpus
8 (26)
1 (11)
Line of therapy
 First line
22 (71)
6 (67)
 ≥ Second line
9 (29)
3 (33)
WHO World Health Organization DL Dose Level
aPhase II included 7 patients of the DL1 cohort and 24 patients of the phase II cohort

Phase I

Sixteen patients were enrolled in the phase I part of this study. Dose level I was expanded to six patients because 1 patient developed grade 3 mucositis as DLT. The same patient discontinued treatment because of cerebral infarction 6 weeks after start of treatment. Because of vascular problems in the medical history of the patient, this complication was considered not to be related to study medication. Nonetheless, we decided to include an additional patient in this dose level. In the first cohort of dose level II one patient developed grade 3 hand-foot syndrome as DLT. Therefore this dose level was expanded to six patients. One of those patients developed grade 3 hand-foot syndrome and mucositis. These were not considered to be DLTs, because by mistake one additional week of capecitabine was taken by the patient in the first cycle and symptoms resolved after 3 weeks of interruption and the original dose could be restarted. Because two other patients with progressive disease within the first cycle, were not assessable for toxicity, three additional patients were included in DL2. Subsequently one patient developed grade 4 rash and three patients had grade 3 mucositis as DLTs. Due to the five DLTs in DL2, DL1 was considered as the MTD and the recommended dose for the phase II part.
In Table 4 all toxicity for the nine patients in dose level 2 is depicted. In these nine patients, a total of 25 complete cycles were given, with a median of 2 (range 0–10) cycles per patient. Dose reductions and interruptions are depicted in Table 3.
Table 3
Dose interruptions and reductions
 
Interruptions
Reductions
Cycles (%)
Patients (%)
Cycles (%)
Patients (%)
Total DL2
10 (40)
5 (56)
5 (20)
5 (56)
Capecitabine
5 (20)
4 (44)
1 (4)
1 (11)
Everolimus
4 (16)
4 (44)
4 (16)
4 (44)
Cetuximab
1 (4)
1 (11)
0 (0)
0 (0)
Total phase 2
22 (20)
8 (26)
6 (5)
6 (19)
Capecitabine
6 (5)
3 (10)
5 (4)
5 (16)
Everolimus
5 (4)
5 (16)
1 (1)
1 (3)
Cetuximab
11 (10)
5 (16)
0 (0)
0 (0)
Total cycles: 25 in dose level 2 (DL2), 113 in phase 2. Total patients: 9 in DL2 and 31 in phase 2

Phase II

Safety and toxicity

In the phase II part of this study, 24 eligible patients were enrolled. When combined with the seven patients treated at MTD in the phase I part, a total of 31 patients could be evaluated according to the protocol. In these 31 patients, a total of 113 complete cycles were given, with a median (range) of 3.0 (0–20) cycles per patient. Dose reductions and interruptions are depicted in Table 3. Table 4 represents the treatment related toxicity. The most common grade 3/4 toxicities of the phase II part included hyperglycemia (26%), rash (19%), mucositis (13%) and fatigue (13%).
Table 4
Treatment related toxicity
 
31 patients in phase II analysisa
9 patients in DL2
 
Any grade (%)
Grade 3–4 (%)
Any grade (%)
Grade 3–4 (%)
Hematologic
Anemia
14 (45)
1 (3)
6 (67)
0 (0)
Neutropenia
7 (23)
0 (0)
5 (56)
0 (0)
Thrombocytopenia
8 (26)
0 (0)
3 (33)
0 (0)
Non hematological
Rash
24 (77)
6 (19)
6 (67)
3 (27)
Mucositis
14 (45)
4 (13)
6 (67)
4 (36)
Fatigue
13 (42)
4 (13)
4 (44)
1 (9)
Diarrhea
10 (32)
4 (13)
3 (33)
0 (0)
Hand-foot syndrome
10 (32)
2 (6)
1 (11)
2 (18)
Infection
5 (16)
1 (3)
2 (22)
2 (18)
Nausea/vomit
16 (52)
1 (3)
6 (67)
0 (0)
Hypokalemia
17 (55)
4 (13)
1 (11)
0 (0)
Hyperglycaemia
19 (61)
8 (26)
5 (56)
1 (18)
Total unique patients
31 (100)
23 (74)
9 (100)
8 (89)
DL Dose Level a Phase II includes 7 patients from DL1 and 24 from phase II

Efficacy and survival

Table 5 summarizes the objective response observed in the 31 treated patients (71% first line, 29% second line) at the MTD level. Out of 31 patients, 24 patients were evaluable for response. Seven patients were not evaluable for response due to events before the first planned CT-evaluation: two patients were clinically progressive, one patient died (as described in the phase I part) and three patients stopped treatment because of treatment related toxicity (grade 3 diarrhea, intolerable rash). One patient refused further treatment. These seven patients were considered as non-responders.
Table 5
Objective treatment response
Response
N = 31(%)
CI
PR
2 (6.5)
1.1–18.8
SD
5 (16.1)
6.2–31.2
PD
17 (54.8)
37.9–71.1
Not assessed
7 (22.6)
10.6–38.8
Objective Response ratea
2 (6.5)
1.1–18.8
Disease Control rateb
7 (22.6)
10.6–38.8
PR partial response, SD stable disease PD progressive disease, CI confidence interval a Objective response rate = PR b Disease Control rate = PR + SD
We observed two (6.5% CI: 1.1–18.8%) partial responses according to RECIST 1.1. Five (16.1% CI: 6.2–31.2%) patients had stable disease, giving a disease control rate of 23.8%. Seventeen (54.8% CI: 37.9–71.1%) patients had progressive disease.
Overall survival of the 31 patients treated at MTD was 5.0 months (CI 3.1–6.8 months) (Fig. 1). The one-year survival rate was 12,9%. Time to treatment failure was 2.3 months (CI 1.7–2.8 months).
Table 6 summarizes overall survival differences between groups based on differences in baseline characteristics, measured by KM-method and a log-rank test. Overall survival for first line patients was 5.0 (CI 4.1–5.9) months, and for second line patients 3.6 months (CI 0.0–7.7) months, which was not a significant difference.
Table 6
Kaplan-Meyer analysis for overall survival, effect of baseline characteristics
 
Total
Censored
Median survival (Months, CI)
1 year survival (%)
Logrank_p
All Patients
31
4
5.0 (3.1–6.8)
12.9
 
Sex
 Female
18
0
5.0 (4.2–5.7)
5.6
0.203
 Male
13
4
4.5 (1.7–7.3)
23.1
 
 Age
 <65
24
2
4.5 (1.2–7.7)
12.5
0.112
 ≥65
7
2
10.2 (0.0–20.5)
14.3
 
WHO performance status
 0
16
4
5.2 (4.1–6.4)
25.0
0.093
 1
10
0
4.5 (0.1–8.8)
0.0
 
 2
5
0
2.3 (1.9–2.7)
0.0
 
Metastasis
 Locally advanced
4
1
5.2 (2.0–8.4)
25.0
0.547
 Metastatic
27
3
4.9 (2.5–7.3)
11.1
 
Line of therapy
 First line
22
3
5.0 (4.1–5.9)
18.2
0.603
 ≥Second line
9
1
3.6 (0.0–7.3)
0.0
 
WHO World Health Organization CI Confidence Interval

Discussion

The MTD for everolimus and capecitabine in combination with cetuximab (cetuximab 250 mg/m2 weekly) was already reached at the first dose level (everolimus 5 mg daily, and capecitabine 600 mg/m2 BID). The DLTs were mucositis, rash and hand-foot syndrome. In the phase II part of this study the incidence of grade 3–4 hyperglycemia, a well known complication of everolimus, was considerable and seems even to be higher compared to studies with everolimus alone. Despite the relative low dose level of everolimus (5 mg) the incidence of severe mucositis (13% grade 3–4) was still considerable and also seems to be higher compared to trials with everolimus monotherapy (10 mg). In other studies using single agent everolimus the incidence of grade 3–4 mucositis was 1–7% [26, 27]. In our previous phase I study (Deenen et al.) with everolimus 10 mg and capecitabine 500–1000 mg/m2, mucositis was not dose-limiting, however grade 1–2 mucositis was present in 50% of the patients [21]. Thus, the addition of cetuximab resulted in more toxicity and prevented dose escalations of everolimus and capecitabine to more optimal dosages [28]. Although the underlying mechanism is unclear, co- administration of three agents with overlapping toxicities may be an important explanation for the excessive mucosal and/or epidermal toxicities seen in this study. Because the study of Deenen et al. demonstrated no pharmacokinetic interactions between everolimus and capecitabine, and monoclonal antibodies do not interfere at pharmacokinetic level, the increased toxicity is probably caused by pharmacodynamic interaction between the three drugs.
The objective response rate was only 6.5% (CI 1.1–18.8) with an overall survival of 5 months. In the first line cohort the OS was also 5.0 months, which even seems to be slightly inferior in comparison with gemcitabine as first line treatment [2]. In preclinical studies with cell lines of non small cell lung, pancreatic, colon, and breast cancer combined inhibition of mTOR and EGFR resulted in a potentiation of anti cancer activity and resensitization of cell lines resistant to EGFR inhibitors [1518, 29]. Despite these promising preclinical results, exploration of this strategy in pancreatic cancer patients in the present study was disappointing. Possible reasons for clinical resistance to this treatment combination are the following. Toxicity prevented the administration of everolimus at optimal dosages. Everolimus shows most effective mTOR inhibition at a dose of 10 mg, and capecitabine monotherapy showed the optimal efficacy at 1000–1250 mg/m2 (BID) [28, 30, 31]. Secondly, the desmoplastic nature of pancreatic cancer with a high fibrotic component and minimal vascularization might prevent an adequate drug penetration, especially a large molecule like the monoclonal antibody cetuximab. Another, in various cancer types well established, cause of resistance to anti-EGFR treatment is KRAS mutation, predominantly present in pancreatic cancer cells, which accounts for constitutive signaling directly downstream of EGFR. However, inhibitors of EGFR still show efficacy in some KRAS mutated pancreatic cancer cell lines [32].
In conclusion, dose escalation of everolimus and capecitabine in the present triple drug schedule with cetuximab was not possible because of severe, especially epidermal and mucosal, toxicity. At the relative low everolimus and capecitabine dosages in the phase II part of this study the toxicity was still considerable, leading to dose interruptions and adaptations. Considering the toxicity, the response rate of 6.5%, and median survival of 5 months in first line treated patients, this regimen does not deserve further exploration in pancreatic cancer patients.

Funding

Merck provided cetuximab and Novartis provided everolimus.

Conflict of interest

The authors declare that they have no conflict of interest.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Jemal A, Siegel R, Xu J, Ward E (2010) Cancer statistics, 2010. CA Cancer J Clin 60:277–300PubMedCrossRef Jemal A, Siegel R, Xu J, Ward E (2010) Cancer statistics, 2010. CA Cancer J Clin 60:277–300PubMedCrossRef
2.
Zurück zum Zitat Burris HA III, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15:2403–2413PubMed Burris HA III, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15:2403–2413PubMed
3.
Zurück zum Zitat Conroy T, Gavoille C, Adenis A (2011) Metastatic pancreatic cancer: old drugs, new paradigms. Curr Opin Oncol 23:390–395PubMedCrossRef Conroy T, Gavoille C, Adenis A (2011) Metastatic pancreatic cancer: old drugs, new paradigms. Curr Opin Oncol 23:390–395PubMedCrossRef
4.
Zurück zum Zitat Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, Bennouna J, Bachet JB, Khemissa-Akouz F, Pere-Verge D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M (2011) FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364:1817–1825PubMedCrossRef Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, Bennouna J, Bachet JB, Khemissa-Akouz F, Pere-Verge D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M (2011) FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364:1817–1825PubMedCrossRef
5.
Zurück zum Zitat Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SA (2004) The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-kappaB and c-Myc in pancreatic cancer cells. Oncogene 23:8571–8580PubMedCrossRef Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SA (2004) The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-kappaB and c-Myc in pancreatic cancer cells. Oncogene 23:8571–8580PubMedCrossRef
6.
Zurück zum Zitat Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA (2006) Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 20:1218–1249PubMedCrossRef Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA (2006) Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 20:1218–1249PubMedCrossRef
7.
Zurück zum Zitat Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, Campos D, Lim R, Ding K, Clark G, Voskoglou-Nomikos T, Ptasynski M, Parulekar W (2007) Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 25:1960–1966PubMedCrossRef Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, Campos D, Lim R, Ding K, Clark G, Voskoglou-Nomikos T, Ptasynski M, Parulekar W (2007) Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 25:1960–1966PubMedCrossRef
8.
Zurück zum Zitat Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, Khorana AA, Goldman B, Fenoglio-Preiser CM, Abbruzzese JL, Blanke CD (2010) Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol 28:3605–3610PubMedCrossRef Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, Khorana AA, Goldman B, Fenoglio-Preiser CM, Abbruzzese JL, Blanke CD (2010) Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol 28:3605–3610PubMedCrossRef
9.
Zurück zum Zitat Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, Kawaguchi Y, Doi R (2006) In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer 118:2337–2343PubMedCrossRef Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, Kawaguchi Y, Doi R (2006) In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer 118:2337–2343PubMedCrossRef
10.
Zurück zum Zitat Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP, Fuchs CS (2009) Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol 27:193–198PubMedCrossRef Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP, Fuchs CS (2009) Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol 27:193–198PubMedCrossRef
11.
Zurück zum Zitat Camp ER, Summy J, Bauer TW, Liu W, Gallick GE, Ellis LM (2005) Molecular mechanisms of resistance to therapies targeting the epidermal growth factor receptor. Clin Cancer Res 11:397–405PubMed Camp ER, Summy J, Bauer TW, Liu W, Gallick GE, Ellis LM (2005) Molecular mechanisms of resistance to therapies targeting the epidermal growth factor receptor. Clin Cancer Res 11:397–405PubMed
12.
Zurück zum Zitat Sampaio C, Dance M, Montagner A, Edouard T, Malet N, Perret B, Yart A, Salles JP, Raynal P (2008) Signal Strength Dictates Phosphoinositide 3-Kinase Contribution to Ras/Extracellular Signal-Regulated Kinase 1 and 2 Activation via Differential Gab1/Shp2 Recruitment: Consequences for Resistance to Epidermal Growth Factor Receptor Inhibition. Mol Cell Biol 28:587–600PubMedCrossRef Sampaio C, Dance M, Montagner A, Edouard T, Malet N, Perret B, Yart A, Salles JP, Raynal P (2008) Signal Strength Dictates Phosphoinositide 3-Kinase Contribution to Ras/Extracellular Signal-Regulated Kinase 1 and 2 Activation via Differential Gab1/Shp2 Recruitment: Consequences for Resistance to Epidermal Growth Factor Receptor Inhibition. Mol Cell Biol 28:587–600PubMedCrossRef
13.
Zurück zum Zitat Blanke CD, Rankin C, Demetri GD, Ryan CW, von Mehren M, Benjamin RS, Raymond AK, Bramwell VH, Baker LH, Maki RG, Tanaka M, Hecht JR, Heinrich MC, Fletcher CD, Crowley JJ, Borden EC (2008) Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J Clin Oncol 26:626–632PubMedCrossRef Blanke CD, Rankin C, Demetri GD, Ryan CW, von Mehren M, Benjamin RS, Raymond AK, Bramwell VH, Baker LH, Maki RG, Tanaka M, Hecht JR, Heinrich MC, Fletcher CD, Crowley JJ, Borden EC (2008) Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J Clin Oncol 26:626–632PubMedCrossRef
14.
Zurück zum Zitat Shepherd FA, Rodrigues PJ, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins R, van Kooten M, Dediu M, Findlay B, Tu D, Johnston D, Bezjak A, Clark G, Santabarbara P, Seymour L (2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353:123–132PubMedCrossRef Shepherd FA, Rodrigues PJ, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins R, van Kooten M, Dediu M, Findlay B, Tu D, Johnston D, Bezjak A, Clark G, Santabarbara P, Seymour L (2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353:123–132PubMedCrossRef
15.
Zurück zum Zitat Bianco R, Garofalo S, Rosa R, Damiano V, Gelardi T, Daniele G, Marciano R, Ciardiello F, Tortora G (2008) Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs. Br J Cancer 98:923–930PubMedCrossRef Bianco R, Garofalo S, Rosa R, Damiano V, Gelardi T, Daniele G, Marciano R, Ciardiello F, Tortora G (2008) Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs. Br J Cancer 98:923–930PubMedCrossRef
16.
Zurück zum Zitat Buck E, Eyzaguirre A, Brown E, Petti F, McCormack S, Haley JD, Iwata KK, Gibson NW, Griffin G (2006) Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther 5:2676–2684PubMedCrossRef Buck E, Eyzaguirre A, Brown E, Petti F, McCormack S, Haley JD, Iwata KK, Gibson NW, Griffin G (2006) Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther 5:2676–2684PubMedCrossRef
17.
Zurück zum Zitat Goudar RK, Shi Q, Hjelmeland MD, Keir ST, McLendon RE, Wikstrand CJ, Reese ED, Conrad CA, Traxler P, Lane HA, Reardon DA, Cavenee WK, Wang XF, Bigner DD, Friedman HS, Rich JN (2005) Combination therapy of inhibitors of epidermal growth factor receptor/vascular endothelial growth factor receptor 2 (AEE788) and the mammalian target of rapamycin (RAD001) offers improved glioblastoma tumor growth inhibition. Mol Cancer Ther 4:101–112PubMed Goudar RK, Shi Q, Hjelmeland MD, Keir ST, McLendon RE, Wikstrand CJ, Reese ED, Conrad CA, Traxler P, Lane HA, Reardon DA, Cavenee WK, Wang XF, Bigner DD, Friedman HS, Rich JN (2005) Combination therapy of inhibitors of epidermal growth factor receptor/vascular endothelial growth factor receptor 2 (AEE788) and the mammalian target of rapamycin (RAD001) offers improved glioblastoma tumor growth inhibition. Mol Cancer Ther 4:101–112PubMed
18.
Zurück zum Zitat Jimeno A, Kulesza P, Wheelhouse J, Chan A, Zhang X, Kincaid E, Chen R, Clark DP, Forastiere A, Hidalgo M (2007) Dual EGFR and mTOR targeting in squamous cell carcinoma models, and development of early markers of efficacy. Br J Cancer 96:952–959PubMedCrossRef Jimeno A, Kulesza P, Wheelhouse J, Chan A, Zhang X, Kincaid E, Chen R, Clark DP, Forastiere A, Hidalgo M (2007) Dual EGFR and mTOR targeting in squamous cell carcinoma models, and development of early markers of efficacy. Br J Cancer 96:952–959PubMedCrossRef
19.
Zurück zum Zitat La MS, Galetti M, Alfieri RR, Cavazzoni A, Ardizzoni A, Tiseo M, Capelletti M, Goldoni M, Tagliaferri S, Mutti A, Fumarola C, Bonelli M, Generali D, Petronini PG (2009) Everolimus restores gefitinib sensitivity in resistant non-small cell lung cancer cell lines. Biochem Pharmacol 78:460–468CrossRef La MS, Galetti M, Alfieri RR, Cavazzoni A, Ardizzoni A, Tiseo M, Capelletti M, Goldoni M, Tagliaferri S, Mutti A, Fumarola C, Bonelli M, Generali D, Petronini PG (2009) Everolimus restores gefitinib sensitivity in resistant non-small cell lung cancer cell lines. Biochem Pharmacol 78:460–468CrossRef
20.
Zurück zum Zitat Nakachi I, Naoki K, Soejima K, Kawada I, Watanabe H, Yasuda H, Nakayama S, Yoda S, Satomi R, Ikemura S, Terai H, Sato T, Ishizaka A (2010) The combination of multiple receptor tyrosine kinase inhibitor and mammalian target of rapamycin inhibitor overcomes erlotinib resistance in lung cancer cell lines through c-Met inhibition. Mol Canc Res 8:1142–1151CrossRef Nakachi I, Naoki K, Soejima K, Kawada I, Watanabe H, Yasuda H, Nakayama S, Yoda S, Satomi R, Ikemura S, Terai H, Sato T, Ishizaka A (2010) The combination of multiple receptor tyrosine kinase inhibitor and mammalian target of rapamycin inhibitor overcomes erlotinib resistance in lung cancer cell lines through c-Met inhibition. Mol Canc Res 8:1142–1151CrossRef
21.
Zurück zum Zitat Deenen MJ, Klumpen HJ, Richel DJ, Sparidans RW, Weterman MJ, Beijnen JH, Schellens JH, Wilmink JW (2011) Phase I and pharmacokinetic study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced solid malignancies. Investigational New Drugs. doi:10.1007/s10637-011-9723-4 Deenen MJ, Klumpen HJ, Richel DJ, Sparidans RW, Weterman MJ, Beijnen JH, Schellens JH, Wilmink JW (2011) Phase I and pharmacokinetic study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced solid malignancies. Investigational New Drugs. doi:10.​1007/​s10637-011-9723-4
22.
Zurück zum Zitat Pacey S, Rea D, Steven N, Brock C, Knowlton N, Shand N, Hazell K, Zoellner U, O’Donnell A, Judson I (2004) Results of a phase 1 clinical trial investigating a combination of the oral mTOR-inhibitor Everolimus (E, RAD001) and Gemcitabine (GEM) in patients (pts) with advanced cancers. J Clin Oncol, 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition) 22(No 14S (July 15 Supplement)):3120 Pacey S, Rea D, Steven N, Brock C, Knowlton N, Shand N, Hazell K, Zoellner U, O’Donnell A, Judson I (2004) Results of a phase 1 clinical trial investigating a combination of the oral mTOR-inhibitor Everolimus (E, RAD001) and Gemcitabine (GEM) in patients (pts) with advanced cancers. J Clin Oncol, 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition) 22(No 14S (July 15 Supplement)):3120
23.
Zurück zum Zitat Li J, Zhao M, He P, Hidalgo M, Baker SD (2007) Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes. Clin Cancer Res 13:3731–3737PubMedCrossRef Li J, Zhao M, He P, Hidalgo M, Baker SD (2007) Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes. Clin Cancer Res 13:3731–3737PubMedCrossRef
24.
Zurück zum Zitat Bullock KE, Petros WP, Younis I, Uronis HE, Morse MA, Blobe GC, Zafar SY, Gockerman JP, Lager JJ, Truax R, Meadows KL, Howard LA, O’Neill MM, Broadwater G, Hurwitz HI, Bendell JC (2011) A phase I study of bevacizumab (B) in combination with everolimus (E) and erlotinib (E) in advanced cancer (BEE). Cancer Chemother Pharmacol 67:465–474PubMedCrossRef Bullock KE, Petros WP, Younis I, Uronis HE, Morse MA, Blobe GC, Zafar SY, Gockerman JP, Lager JJ, Truax R, Meadows KL, Howard LA, O’Neill MM, Broadwater G, Hurwitz HI, Bendell JC (2011) A phase I study of bevacizumab (B) in combination with everolimus (E) and erlotinib (E) in advanced cancer (BEE). Cancer Chemother Pharmacol 67:465–474PubMedCrossRef
25.
Zurück zum Zitat Le TC, Lee JJ, Siu LL (2009) Dose escalation methods in phase I cancer clinical trials. J Natl Canc Inst 101:708–720CrossRef Le TC, Lee JJ, Siu LL (2009) Dose escalation methods in phase I cancer clinical trials. J Natl Canc Inst 101:708–720CrossRef
26.
Zurück zum Zitat Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grunwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A (2008) Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372:449–456PubMedCrossRef Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grunwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A (2008) Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372:449–456PubMedCrossRef
27.
Zurück zum Zitat Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van CE, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514–523PubMedCrossRef Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van CE, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514–523PubMedCrossRef
28.
Zurück zum Zitat Cartwright TH, Cohn A, Varkey JA, Chen YM, Szatrowski TP, Cox JV, Schulz JJ (2002) Phase II study of oral capecitabine in patients with advanced or metastatic pancreatic cancer. J Clin Oncol 20:160–164PubMedCrossRef Cartwright TH, Cohn A, Varkey JA, Chen YM, Szatrowski TP, Cox JV, Schulz JJ (2002) Phase II study of oral capecitabine in patients with advanced or metastatic pancreatic cancer. J Clin Oncol 20:160–164PubMedCrossRef
29.
Zurück zum Zitat Johnson BE, Jackman D, Janne PA (2007) Rationale for a Phase I Trial of Erlotinib and the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) for Patients with Relapsed Non Small Cell Lung Cancer. Clin Cancer Res 13:4628s–4631CrossRef Johnson BE, Jackman D, Janne PA (2007) Rationale for a Phase I Trial of Erlotinib and the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) for Patients with Relapsed Non Small Cell Lung Cancer. Clin Cancer Res 13:4628s–4631CrossRef
30.
Zurück zum Zitat Tabernero J, Rojo F, Calvo E, Burris H, Judson I, Hazell K, Martinelli E, Cajal S, Jones S, Vidal L, Shand N, Macarulla T, Ramos FJ, Dimitrijevic S, Zoellner U, Tang P, Stumm M, Lane HA, Lebwohl D, Baselga J (2008) Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. J Clin Oncol 26:1603–1610PubMedCrossRef Tabernero J, Rojo F, Calvo E, Burris H, Judson I, Hazell K, Martinelli E, Cajal S, Jones S, Vidal L, Shand N, Macarulla T, Ramos FJ, Dimitrijevic S, Zoellner U, Tang P, Stumm M, Lane HA, Lebwohl D, Baselga J (2008) Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. J Clin Oncol 26:1603–1610PubMedCrossRef
31.
Zurück zum Zitat Klumpen HJ, Beijnen JH, Gurney H, Schellens JH (2010) Inhibitors of mTOR. Oncologist 15:1262–1269PubMedCrossRef Klumpen HJ, Beijnen JH, Gurney H, Schellens JH (2010) Inhibitors of mTOR. Oncologist 15:1262–1269PubMedCrossRef
32.
Zurück zum Zitat Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ (2000) Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res 60:2926–293PubMed Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ (2000) Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res 60:2926–293PubMed
Metadaten
Titel
A phase I/II, non-randomized, feasibility/safety and efficacy study of the combination of everolimus, cetuximab and capecitabine in patients with advanced pancreatic cancer
verfasst von
Sil Kordes
Dick J. Richel
Heinz-Josef Klümpen
Mariëtte J. Weterman
Arnoldus J. W. M. Stevens
Johanna W. Wilmink
Publikationsdatum
01.02.2013
Verlag
Springer US
Erschienen in
Investigational New Drugs / Ausgabe 1/2013
Print ISSN: 0167-6997
Elektronische ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-012-9802-1

Weitere Artikel der Ausgabe 1/2013

Investigational New Drugs 1/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.