Skip to main content
Erschienen in: Targeted Oncology 4/2017

Open Access 01.07.2017 | Original Research Article

Pooled Analysis of Clinical Outcome of Patients with Chemorefractory Metastatic Colorectal Cancer Treated within Phase I/II Clinical Studies Based on Individual Biomarkers of Susceptibility: A Single-Institution Experience

verfasst von: Andrea Sartore-Bianchi, Alessio Amatu, Erica Bonazzina, Stefano Stabile, Laura Giannetta, Giulio Cerea, Ilaria Schiavetto, Katia Bencardino, Chiara Funaioli, Riccardo Ricotta, Tiziana Cipani, Michele Schirru, Valentina Gambi, Laura Palmeri, Giulia Carlo-Stella, Francesca Rusconi, Sara Di Bella, Giovanni Burrafato, Andrea Cassingena, Emanuele Valtorta, Calogero Lauricella, Federica Pazzi, Alessandra Gambaro, Silvia Ghezzi, Giovanna Marrapese, Emiliana Tarenzi, Silvio Veronese, Mauro Truini, Angelo Vanzulli, Salvatore Siena

Erschienen in: Targeted Oncology | Ausgabe 4/2017

Abstract

Background

Patients with metastatic colorectal cancer (mCRC) refractory to standard therapies have a poor prognosis. In this setting, recruitment into clinical trials is warranted, and studies driven by selection according to individual tumor molecular characteristics are expected to provide added value.

Objective

We retrospectively analyzed data from patients with mCRC refractory to or following failure of standard therapies who were enrolled into phase I/II clinical studies at the Niguarda Cancer Center based on the presence of a specific molecular profile expected to represent the target of susceptibility to the experimental drug(s).

Patients and Methods

From June 2011 to May 2016, 2044 patients with mCRC underwent molecular screening. Eighty patients (3.9%) were enrolled in ad hoc studies; the median age was 60 years (range 36–86) and the median number of previous treatment lines was five (range 2–8). Molecular characteristics exploited within these studies were MGMT promoter hypermethylation (48.7%), HER2 amplification (28.8%), BRAF V600E mutation (20%), and novel gene fusions involving ALK or NTRK (2.5%).

Results

One patient (1%) had RECIST (Response Evaluation Criteria In Solid Tumors) complete response (CR), 13 patients (16.5%) experienced a partial response (PR), and 28 (35%) stable disease (SD). Median progression-free survival (PFS) was 2.8 months (range 2.63–3.83), with 24% of patients displaying PFS >5 months. Median growth modulation index (GMI) was 0.85 (range 0–15.61) and 32.5% of patients had GMI >1.33. KRAS exon 2 mutations were found in 38.5% of patients, and among the 78 patients with known KRAS status, those with wild-type tumors had longer PFS than those with mutated tumors (3.80 [95% CI 2.80–5.03] vs. 2.13 months [95% CI 1.77–2.87], respectively, p = 0.001). Median overall survival (OS) was 7.83 months (range 7.17–9.33) for all patients, and patients with KRAS wild-type tumors had longer OS than those with mutated tumors (7.83 [95% CI 7.33–10.80] vs. 7.18 months [95% CI 5.63–9.33], respectively, p = 0.06).

Conclusions

This single-institution retrospective study indicates that in a heavily pretreated population approximately 4% of mCRC tumors display a potential actionable molecular context suitable for therapeutic intervention. Application of molecular selection is challenging but improves clinical outcome even in later lines of treatment.

1 Introduction

Colorectal cancer (CRC) remains a significant cause of morbidity and mortality worldwide and many patients present with metastatic disease despite large-scale screening programs in various countries [1]. With the only exception of patients presenting with oligometastatic lesions confined to the liver or lung amenable to resection [2, 3], metastatic disease is considered incurable. In cases where treatment with curative intent is excluded, patients are typically given a combination of cytotoxic chemotherapy, often in conjunction with a targeted therapy.
Although advances in systemic therapy have been made, the 5-year survival rate in this setting is still 12.5% [4], with acquired resistance to therapy being the main reason for treatment failure [5]. Indeed, resistance to targeted therapy, as displayed by disease progression, is observed after a median of 4 months on epidermal growth factor receptor (EGFR) antagonists [45]. The discovery of RAS mutations in CRC as a mechanism of innate resistance to these therapies has been an important advance and has ameliorated their clinical use. However, there is an unmet need for effective therapeutic strategies after secondary resistance.
We have previously demonstrated that different molecular alterations that drive resistance can occur simultaneously in the same patient [7]. Identifying relevant molecular subtypes within this heterogeneous disease and matching patients with appropriate single agents or combinations of targeted therapies at resistance is crucial to therapeutic progress [8]. Therefore, recruitment into precision oncology clinical trials based on selection according to individual tumor molecular characteristics is expected to provide added value.
We retrospectively collected data from patients with metastatic CRC (mCRC) resistant to standard therapies treated at the Niguarda Cancer Center (NCC) (Milan, Italy) in phase I/II clinical studies based on the presence of specific tumor molecular profiles conferring susceptibility to experimental drugs, and performed a pooled analysis for measuring results according to main clinical and other molecular variables.

2 Methods

2.1 Patients

We retrospectively collected data from patients with mCRC resistant to standard therapies treated at NCC between June 2011 and May 2016 in phase I/II clinical studies, including one phase I first-in-human study, based on the presence of specific biomarkers that confer susceptibility to experimental drugs (Table 1). These included tumor genetic alterations (i.e., gene mutations, amplifications, or fusions) or a certain genetic context (i.e., methylation of specific genes). Consecutive eligible patients were offered participation in clinical trials. All patients gave written informed consent and the study and all treatments were conducted in accordance with the guidelines of the Institutional Review Board at Ospedale Niguarda.
Table 1
Distribution of patients in clinical trials with actionable molecular alterations treated with matched targeted agents included in the pooled analysis
Biomarker
Study drug(s)
EudraCT N°
Patients (n)
MGMT promoter hypermethylation
Temozolomide [9]
2012–003338-17
27
HER2 amplification
Trastuzumab + lapatinib [10]
2012–002128-33
23
MGMT promoter hypermethylation
Dacarbazine [11]
2011–002080-21
12
BRAF mutation
MEK162 + LGX818 [NCT01543698]
2011–005875-17
9
BRAF mutation
MEK162 + panitumumab [NCT01927341]
2013–001986-18
7
NTRK or ALK gene fusions
Entrectinib [12]
2012–000148-88
2
The presence of the particular biomarker was investigated according to specific study protocol criteria or retrieved by medical history, where applicable. Further molecular characterization of Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations, a well-known biomarker associated with poor prognosis, was performed in all available samples of patients included in the studies by the Pathology Department of NCC as per standard of care. The registration of patients in the database, pathology assessment, and additional mutation analyses if needed were performed at NCC.

2.2 Treatment and Evaluation

Treatment was carried out according to the specific requisites of treatment protocols and was continued until disease progression, unacceptable toxicity, or withdrawal of consent. Assessments, including history, physical examination, and laboratory evaluations, were performed as specified in each protocol, typically before the initiation of therapy, and longitudinally during the treatment cycles. Efficacy was assessed from computed tomography scans and/or magnetic resonance imaging at baseline before treatment initiation and then every 6–8 weeks depending on the study protocol. All radiographs were evaluated by the Department of Radiology at NCC and responses were categorized per RECIST (Response Evaluation Criteria In Solid Tumors) 1.1 criteria and were reported as best response [13]. Progression-free survival of the treatment(PFSn)was recorded for each individual patient as in the case report form of the respective trial, whereas progression-free survival achieved with the previous treatment line received by the patient (PFSn–1) was calculated based on available clinical documentation.

2.3 Statistical Analysis

Fisher’s exact test was used to assess the association among categorical variables and the presence of the biomarker used for treatment selection. Progression-free survival (PFS) was defined as the time interval from the start of therapy to the first observation of disease progression or death, whichever occurred first. All tests were 2-sided, and p < 0.05 was considered statistically significant. Multivariate analysis was performed by Cox logistic regression; the model as a whole was evaluated by likelihood ratio test, whereas the significance of the single independent variables was evaluated by means of the Wald test. Results of the multivariate analysis should be interpreted with caution due to the small sample size and sampling bias of our study.
PFS and overall survival (OS) for all patients were computed using the Kaplan-Meier method and differences in PFS and OS were calculated with log-rank test where applicable. The growth modulation index (GMI) has been applied as previously described [14]. We consider a GMI >1.33, i.e., an increase in the PFSn/PFSn–1 ratio of 30%, as clinically meaningful. All statistical analyses were carried out using R software [15] and graphics were generated with ggplot2 [16].

3 Results

From June 2011 to May 2016, 2044 patients with mCRC were referred to NCC for molecular screening for the inclusion into phase I/II trials encompassing the targeting of actionable molecular alterations or molecular contexts of susceptibility. In total, 80 patients (3.9%) were enrolled into six ad hoc studies: one phase I/II and five phase II trials.
The median age was 60 years (range 36–86) and the median number of previous treatment lines was five (range 2–8). The six studies included therapies based on MGMT promoter hypermethylation (48.7%), HER2 amplification (28.8%), BRAF V600E mutation (20%), and gene fusions involving ALK or TRKA (2.5%) (Table 1 and Fig. 1). Among the 78 of 80 patients evaluable for KRAS mutations, any KRAS (exon 2) mutation was found in 30 (38.5%) of patients.
According to RECIST 1.1 criteria, one patient (1%) had complete response (CR), 13 patients (16.5%) had partial response (PR), and 28 (35%) had stable disease (SD), accounting for a 52.5% disease control rate (DCR = CR + PR + SD). The DCR was higher in patients with KRAS wild-type tumors (66 vs. 38%, p = 0.02). The median PFS was 2.8 months (range 2.63–3.83) (Fig. 2a), and 24% of patients displayed a PFS >5 months. A multivariate analysis of clinicopathological factors including age, gender, performance status, carcinoembryonic antigen (CEA) levels, and KRAS status showed that KRAS status and age were significantly associated with PFS (p = 0.011 and 0.047, respectively), whereas CEA and age were significantly associated with OS (p = 0.0063 and 0.0279, respectively). Patients with KRAS wild-type tumors had longer PFS than those with mutated tumors (3.80 [95% CI 2.80–5.03] vs. 2.13 months [95% CI 1.77–2.87], respectively) (p = 0.001) (Fig. 2b). Median OS was 7.83 months (range 7.17–9.33) (Fig. 3a), and patients with KRAS wild-type tumors had longer OS than those with mutated tumors (7.83 [95% CI 7.33–10.80] vs. 7.18 months [95% CI 5.63–9.33], respectively, p = 0.06) (Fig. 3b). The median GMI was 0.85 (range 0–15.61) and 32.5% of patients had a GMI >1.33 (Fig. 4). The Spearman Rho between PFSn and PFSn–1 was 0.10 (p = 0.372).

4 Discussion

In this single-institution retrospective analysis we found that 4.0% of mCRC patients refractory to standard therapies were eligible for enrollment into phase I/II clinical trials based on molecular target selection. These patients, who had experienced a median number of five lines of previous treatments, achieved a median PFS of 2.8 months (with approximately one-quarter of patients displaying a PFS >5 months) and a median OS of 7.8 months. These data should be evaluated in the context of advanced mCRC, where the median PFS with newer agents has been less than 2 months [20], and rechallenge with standard chemotherapy [18, 19] or further chemotherapy [17] showed only poor efficacy and was burdened by adverse effects [21]. With regards to objective response rates (ORRs), 14 patients (17.5%) had PR and 28 (35%) SD, accounting for a 52.5% DCR. These data compare favorably with those achieved with licensed third-line therapies such as the multikinase inhibitor regorafenib (ORR = 1–4%) and the trifluridine–tipiracil combination TAS-102 (ORR = 2%), both of which are administered without the selection of a molecular target, or salvage chemotherapy with mitomycin-C or oxaliplatin rechallenge [17, 22, 23].
Following the seminal work by von Hoff et al. [14], several reports investigating the use of molecularly targeted agents on the basis of the identification of molecular alterations provided conflicting results [43, 44]. Provocative findings from Janku et al. highlighted that in phase I studies the lack of matching between molecular alterations and targeted treatment provides only limited clinical benefit in CRC and other solid tumors [24]. Recently, the same authors reported findings of a prospective, single-center ‘navigation’ study conducted in 500 patients with several different refractory cancers who underwent comprehensive genomic profiling by sequencing of 236 genes, offering proof-of-concept for the utility of this approach in assigning therapy to patients with refractory malignancies, especially in those patients with multiple genomic aberrations for whom combination therapies could be implemented [25]. However, patients with CRC were only a small proportion of this cohort and therefore definitive conclusions could not be drawn for this histology. In mCRC, Dienstmann et al. reported a series of 68 patients treated with targeted agents in phase I trials based on molecular profiling, concluding that this approach did not confer a significant clinical benefit on the basis of a comparison between the median time to treatment failure n (TTFn) of 7.9 weeks and TTFn–1 of 16.3 weeks [26]. Similarly, in our present analysis of 80 patients, we observed that PFSn was inferior to PFSn–1 (12.0 vs. 15.6 weeks). However, there are substantial differences between these two cohorts. Our series included only 2.5% patients treated within a phase I study versus 100% of the patients included in the study by Dienstmann et al. [26], and there are differences in the molecular alterations on which the treatments are based (KRAS/BRAF/PIK3CA mutations, PTEN and MET expression vs. HER2, BRAF, MGMT, and gene fusions, respectively). Assessment of clinical benefit by means of the ratio between PFS with the actual targeted therapy and that achieved with the previous line of treatment, referred to as the GMI, has also been proposed [14]. When we applied this index to our cohort, we found that at least one-third of patients benefited from the enrollment in clinical trials with targeted agents matched to selected actionable molecular alterations, making our results meaningful.
Among the molecular abnormalities selected in our cohort, we included gene fusions involving the ALK and NTRK genes. These alterations have only recently been recognized as targets occurring at low prevalence in CRC [27]. In particular, in 2014 we reported the characterization of the TPM3-NTRK1 gene rearrangement as a recurring, although rare, event in CRC. The concomitant discovery of entrectinib (NMS-P626; RXDX-101) as a novel highly potent and selective pan-Trk inhibitor by the group of Ardini et al. [28] enhanced the interest in this target, with further development across different histologies and currently ongoing phase II studies, including in CRC [12, 27]. Interestingly enough, the two gene fusions involving NTRK and ALK described in the present cohort were previously unreported [29, 30], highlighting the challenge of a comprehensive molecular pathology that should not merely include gene fusions together with mutations and amplifications but also elucidate the exact fusion partner and oncogenic potential for treatment with specific inhibitors.
During the last decade, the assessment of KRAS and neuroblastoma RAS viral oncogene homolog (NRAS) gained increasing importance for an optimal management of mCRC [31]. Mutations in KRAS codons 12 or 13 (in exon 2) affect approximately 40% of patients with metastatic CRC [32]. About an additional 5% of CRC patients display mutations in KRAS exons 3 or 4, usually at codon 61 or 146, and another 5% of CRC patients have mutations in NRAS in exons 2, 3, or 4. Extended RAS status (KRAS and NRAS exons 2, 3, and 4 mutations) is the validated biomarker of response to anti-EGFR antibodies [31]. RAS mutant mCRC exhibit a significantly higher cumulative incidence of lung, bone, and brain metastasis [3335]. The prognostic role of RAS mutations is controversial, but increasing evidence shows a negative effect in the adjuvant and metastatic setting [3638]. In our cohort, KRAS mutations showed an overall negative prognostic value, since patients harboring KRAS-mutated tumors had shorter PFS and OS. Also, DCR was more frequent in patients with KRAS wild-type tumors. However, it should be taken into account that half of the selected phase I/II trials were designed for patients with KRAS wild-type tumors (Cmek2110 and  Cmek2116 for BRAF-mutated tumors, HER2 Amplification for Colo-rectaL Cancer Enhanced Stratification-HERACLES for HER2 amplified tumors) and most of the KRAS-mutated patients were treated in the two studies (DETECT and TEMECT with dacarbazine and temozolomide, respectively) that were not based on directly targeting an oncogenic driver but rather exploiting a molecular context of susceptibility.
Limitations of our study include the small sample size and potential sampling bias, the notion that the use of GMI as an index of clinical benefit in CRC has been criticized as there is a lack of correlation between PFSn and PFSn–1 that is the statistical assumption for applying this method [39, 40], and that KRAS-limited instead of extended RAS analysis was performed in our cohort.
In conclusion, our single-institution study indicates that in a heavily pretreated mCRC population about 4% of tumors display a potential actionable molecular context suitable for phase I/II trials with matched therapeutics. Comprehensive molecular selection is challenging, encompassing inclusion of actionable alterations occurring at low frequency in CRC such as HER2 amplification [10] and rare gene fusions involving the ALK and NTRK genes [41], but enhances therapeutic options to be exploited in the advanced setting. This knowledge, coupled with recent advancements in the understanding of acquired resistance mechanisms to targeted agents through liquid biopsy [8, 42] is expanding opportunities of precision oncology therapies for CRC.

Acknowledgments

The Authors are profoundly grateful to Professor Alberto Bardelli who provided knowledge and cultural motivation to the Precision Oncology Program at Niguarda Cancer Center.

Compliance with Ethical Standards

Funding

The study was partly supported by the grant “Terapia Molecolare dei Tumori” by Fondazione Oncologia Niguarda, by Associazione Italiana per la Ricerca sul Cancro (AIRC) Special program 5 × 1000, and by the European Union Horizon 2020 research and innovation programme 2014–2020 under grant agreement number 635342 (MoTriColor project).

Conflicts of Interest

The authors declare no conflicts of interest.
Open Access This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

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.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, et al. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66:271–89.CrossRefPubMed Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, et al. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66:271–89.CrossRefPubMed
2.
Zurück zum Zitat Abdalla EK, Vauthey J-N, Ellis LM, Ellis V, Pollock R, Broglio KR, et al. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Ann Surg. 2004;239:818–25.CrossRefPubMedPubMedCentral Abdalla EK, Vauthey J-N, Ellis LM, Ellis V, Pollock R, Broglio KR, et al. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Ann Surg. 2004;239:818–25.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Adam R, Delvart V, Pascal G, Valeanu A, Castaing D, Azoulay D, et al. Rescue surgery for unresectable colorectal liver metastases downstaged by chemotherapy: a model to predict long-term survival. Ann Surg. 2004;240:644–57.CrossRefPubMedPubMedCentral Adam R, Delvart V, Pascal G, Valeanu A, Castaing D, Azoulay D, et al. Rescue surgery for unresectable colorectal liver metastases downstaged by chemotherapy: a model to predict long-term survival. Ann Surg. 2004;240:644–57.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–17.CrossRefPubMed Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–17.CrossRefPubMed
5.
Zurück zum Zitat Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol. 2005;205:275–92.CrossRefPubMed Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol. 2005;205:275–92.CrossRefPubMed
6.
Zurück zum Zitat Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.CrossRefPubMed Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.CrossRefPubMed
7.
Zurück zum Zitat Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801.CrossRefPubMedPubMedCentral Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Sartore-Bianchi A, Loupakis F, Argilés G, Prager GW. Challenging chemoresistant metastatic colorectal cancer: therapeutic strategies from the clinic and from the laboratory. Ann Oncol. 2016;27:1456–66.CrossRefPubMed Sartore-Bianchi A, Loupakis F, Argilés G, Prager GW. Challenging chemoresistant metastatic colorectal cancer: therapeutic strategies from the clinic and from the laboratory. Ann Oncol. 2016;27:1456–66.CrossRefPubMed
9.
Zurück zum Zitat Amatu A, Barault L, Moutinho C, Cassingena A, Bencardino K, Ghezzi S, et al. Tumor MGMT promoter hypermethylation changes over time limit temozolomide efficacy in a phase II trial for metastatic colorectal cancer. Ann Oncol. 2016;27:1062–7.CrossRefPubMed Amatu A, Barault L, Moutinho C, Cassingena A, Bencardino K, Ghezzi S, et al. Tumor MGMT promoter hypermethylation changes over time limit temozolomide efficacy in a phase II trial for metastatic colorectal cancer. Ann Oncol. 2016;27:1062–7.CrossRefPubMed
10.
Zurück zum Zitat Sartore-Bianchi A, Trusolino L, Martino C, Bencardino K, Lonardi S, Bergamo F, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17:738–46.CrossRefPubMed Sartore-Bianchi A, Trusolino L, Martino C, Bencardino K, Lonardi S, Bergamo F, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17:738–46.CrossRefPubMed
11.
Zurück zum Zitat Amatu A, Sartore-Bianchi A, Moutinho C, Belotti A, Bencardino K, Chirico G, et al. Promoter CpG island hypermethylation of the DNA repair enzyme MGMT predicts clinical response to dacarbazine in a phase II study for metastatic colorectal cancer. Clin Cancer Res. 2013;19:2265–72.CrossRefPubMed Amatu A, Sartore-Bianchi A, Moutinho C, Belotti A, Bencardino K, Chirico G, et al. Promoter CpG island hypermethylation of the DNA repair enzyme MGMT predicts clinical response to dacarbazine in a phase II study for metastatic colorectal cancer. Clin Cancer Res. 2013;19:2265–72.CrossRefPubMed
12.
Zurück zum Zitat Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017;4:400–9. Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017;4:400–9.
13.
Zurück zum Zitat 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 1990. 2009;45:228–47. 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 1990. 2009;45:228–47.
14.
Zurück zum Zitat Von Hoff DD, Stephenson JJ, Rosen P, Loesch DM, Borad MJ, Anthony S, et al. Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28:4877–83.CrossRef Von Hoff DD, Stephenson JJ, Rosen P, Loesch DM, Borad MJ, Anthony S, et al. Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28:4877–83.CrossRef
15.
17.
Zurück zum Zitat Petrelli F, Ghidini A, Inno A, Barni S. Mitomycin-C+fluoropyrimidines in heavily pretreated metastatic colorectal cancer: a systematic review and evidence synthesis. Anti-Cancer Drugs. 2016;27:488–95.CrossRefPubMed Petrelli F, Ghidini A, Inno A, Barni S. Mitomycin-C+fluoropyrimidines in heavily pretreated metastatic colorectal cancer: a systematic review and evidence synthesis. Anti-Cancer Drugs. 2016;27:488–95.CrossRefPubMed
18.
Zurück zum Zitat Townsend AR, Bishnoi S, Broadbridge V, Beeke C, Karapetis CS, Jain K, et al. Rechallenge with oxaliplatin and fluoropyrimidine for metastatic colorectal carcinoma after prior therapy. Am J Clin Oncol. 2013;36:49–52.CrossRefPubMed Townsend AR, Bishnoi S, Broadbridge V, Beeke C, Karapetis CS, Jain K, et al. Rechallenge with oxaliplatin and fluoropyrimidine for metastatic colorectal carcinoma after prior therapy. Am J Clin Oncol. 2013;36:49–52.CrossRefPubMed
19.
Zurück zum Zitat Santini D, Vincenzi B, La Cesa A, Caricato M, Schiavon G, Spalletta B, et al. Continuous infusion of oxaliplatin plus chronomodulated capecitabine in 5-fluorouracil- and irinotecan-resistant advanced colorectal cancer patients. Oncology. 2005;69:27–34.CrossRefPubMed Santini D, Vincenzi B, La Cesa A, Caricato M, Schiavon G, Spalletta B, et al. Continuous infusion of oxaliplatin plus chronomodulated capecitabine in 5-fluorouracil- and irinotecan-resistant advanced colorectal cancer patients. Oncology. 2005;69:27–34.CrossRefPubMed
20.
Zurück zum Zitat Nielsen DL, Palshof JA, Larsen FO, Jensen BV, Pfeiffer P. A systematic review of salvage therapy to patients with metastatic colorectal cancer previously treated with fluorouracil, oxaliplatin and irinotecan +/− targeted therapy. Cancer Treat Rev. 2014;40:701–15.CrossRefPubMed Nielsen DL, Palshof JA, Larsen FO, Jensen BV, Pfeiffer P. A systematic review of salvage therapy to patients with metastatic colorectal cancer previously treated with fluorouracil, oxaliplatin and irinotecan +/− targeted therapy. Cancer Treat Rev. 2014;40:701–15.CrossRefPubMed
21.
Zurück zum Zitat Bencardino K, Mauri G, Amatu A, Tosi F, Bonazzina E, Palmeri L, et al. Oxaliplatin immune-induced syndrome occurs with cumulative administration and Rechallenge: single institution series and systematic review study. Clin Colorectal Cancer. 2016;15:213–21.CrossRefPubMed Bencardino K, Mauri G, Amatu A, Tosi F, Bonazzina E, Palmeri L, et al. Oxaliplatin immune-induced syndrome occurs with cumulative administration and Rechallenge: single institution series and systematic review study. Clin Colorectal Cancer. 2016;15:213–21.CrossRefPubMed
22.
Zurück zum Zitat Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:303–12.CrossRefPubMed Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:303–12.CrossRefPubMed
23.
Zurück zum Zitat Mayer RJ, Van Cutsem E, Falcone A, Yoshino T, Garcia-Carbonero R, Mizunuma N, et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med. 2015;372:1909–19.CrossRefPubMed Mayer RJ, Van Cutsem E, Falcone A, Yoshino T, Garcia-Carbonero R, Mizunuma N, et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med. 2015;372:1909–19.CrossRefPubMed
24.
Zurück zum Zitat Janku F, Lee JJ, Tsimberidou AM, Hong DS, Naing A, Falchook GS, et al. PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One. 2011;6:e22769.CrossRefPubMedPubMedCentral Janku F, Lee JJ, Tsimberidou AM, Hong DS, Naing A, Falchook GS, et al. PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One. 2011;6:e22769.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Wheler JJ, Janku F, Naing A, Li Y, Stephen B, Zinner R, et al. Cancer therapy directed by comprehensive genomic profiling: a single center study. Cancer Res. 2016;76:3690–701.CrossRefPubMed Wheler JJ, Janku F, Naing A, Li Y, Stephen B, Zinner R, et al. Cancer therapy directed by comprehensive genomic profiling: a single center study. Cancer Res. 2016;76:3690–701.CrossRefPubMed
26.
Zurück zum Zitat Dienstmann R, Serpico D, Rodon J, Saura C, Macarulla T, Elez E, et al. Molecular profiling of patients with colorectal cancer and matched targeted therapy in phase I clinical trials. Mol Cancer Ther. 2012;11:2062–71.CrossRefPubMed Dienstmann R, Serpico D, Rodon J, Saura C, Macarulla T, Elez E, et al. Molecular profiling of patients with colorectal cancer and matched targeted therapy in phase I clinical trials. Mol Cancer Ther. 2012;11:2062–71.CrossRefPubMed
27.
Zurück zum Zitat Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016;1:e000023.CrossRefPubMedPubMedCentral Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016;1:e000023.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Ardini E, Menichincheri M, Banfi P, Bosotti R, De Ponti C, Pulci R, et al. Entrectinib, a pan-TRK, ROS1, and ALK inhibitor with activity in multiple molecularly defined cancer indications. Mol Cancer Ther. 2016;15:628–39.CrossRefPubMed Ardini E, Menichincheri M, Banfi P, Bosotti R, De Ponti C, Pulci R, et al. Entrectinib, a pan-TRK, ROS1, and ALK inhibitor with activity in multiple molecularly defined cancer indications. Mol Cancer Ther. 2016;15:628–39.CrossRefPubMed
29.
Zurück zum Zitat Amatu A, Somaschini A, Cerea G, Bosotti R, Valtorta E, Buonandi P, et al. Novel CAD-ALK gene rearrangement is drugable by entrectinib in colorectal cancer. Br J Cancer. 2015;113:1730–4.CrossRefPubMedPubMedCentral Amatu A, Somaschini A, Cerea G, Bosotti R, Valtorta E, Buonandi P, et al. Novel CAD-ALK gene rearrangement is drugable by entrectinib in colorectal cancer. Br J Cancer. 2015;113:1730–4.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Sartore-Bianchi A, Ardini E, Bosotti R, Amatu A, Valtorta E, Somaschini A, et al. Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer. J Natl Cancer Inst 2016;108. Sartore-Bianchi A, Ardini E, Bosotti R, Amatu A, Valtorta E, Somaschini A, et al. Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer. J Natl Cancer Inst 2016;108.
31.
Zurück zum Zitat Douillard J-Y, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–34.CrossRefPubMed Douillard J-Y, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–34.CrossRefPubMed
32.
Zurück zum Zitat Moroni M, Veronese S, Benvenuti S, Marrapese G, Sartore-Bianchi A, Di Nicolantonio F, et al. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol. 2005;6:279–86.CrossRefPubMed Moroni M, Veronese S, Benvenuti S, Marrapese G, Sartore-Bianchi A, Di Nicolantonio F, et al. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol. 2005;6:279–86.CrossRefPubMed
33.
Zurück zum Zitat Cejas P, López-Gómez M, Aguayo C, Madero R, de Castro CJ, Belda-Iniesta C, et al. KRAS mutations in primary colorectal cancer tumors and related metastases: a potential role in prediction of lung metastasis. PLoS One. 2009;4:e8199.CrossRefPubMedPubMedCentral Cejas P, López-Gómez M, Aguayo C, Madero R, de Castro CJ, Belda-Iniesta C, et al. KRAS mutations in primary colorectal cancer tumors and related metastases: a potential role in prediction of lung metastasis. PLoS One. 2009;4:e8199.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Andreyev HJN, Norman AR, Cunningham D, Oates J, Dix BR, Iacopetta BJ, et al. Kirsten ras mutations in patients with colorectal cancer: the “RASCAL II” study. Br J Cancer. 2001;85:692–6.CrossRefPubMedPubMedCentral Andreyev HJN, Norman AR, Cunningham D, Oates J, Dix BR, Iacopetta BJ, et al. Kirsten ras mutations in patients with colorectal cancer: the “RASCAL II” study. Br J Cancer. 2001;85:692–6.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Deng Y, Wang L, Tan S, Kim GP, Dou R, Chen D, et al. KRAS as a predictor of poor prognosis and benefit from postoperative FOLFOX chemotherapy in patients with stage II and III colorectal cancer. Mol Oncol. 2015;9:1341–7.CrossRefPubMed Deng Y, Wang L, Tan S, Kim GP, Dou R, Chen D, et al. KRAS as a predictor of poor prognosis and benefit from postoperative FOLFOX chemotherapy in patients with stage II and III colorectal cancer. Mol Oncol. 2015;9:1341–7.CrossRefPubMed
36.
Zurück zum Zitat Kadowaki S, Kakuta M, Takahashi S, Takahashi A, Arai Y, Nishimura Y, et al. Prognostic value of KRAS and BRAF mutations in curatively resected colorectal cancer. World J Gastroenterol. 2015;21:1275–83.CrossRefPubMedPubMedCentral Kadowaki S, Kakuta M, Takahashi S, Takahashi A, Arai Y, Nishimura Y, et al. Prognostic value of KRAS and BRAF mutations in curatively resected colorectal cancer. World J Gastroenterol. 2015;21:1275–83.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Vincenzi B, Cremolini C, Sartore-Bianchi A, Russo A, Mannavola F, Perrone G, et al. Prognostic significance of K-Ras mutation rate in metastatic colorectal cancer patients. Oncotarget. 2015;6:31604–12.CrossRefPubMedPubMedCentral Vincenzi B, Cremolini C, Sartore-Bianchi A, Russo A, Mannavola F, Perrone G, et al. Prognostic significance of K-Ras mutation rate in metastatic colorectal cancer patients. Oncotarget. 2015;6:31604–12.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Tosi F, Magni E, Amatu A, Mauri G, Bencardino K, Truini M, et al. Effect of KRAS and BRAF mutations on survival of metastatic colorectal cancer after liver resection: a systematic review and meta-analysis. Clin Colorectal Cancer. Epub 2017 Jan 25. doi: 10.1016/j.clcc.2017.01.004. Tosi F, Magni E, Amatu A, Mauri G, Bencardino K, Truini M, et al. Effect of KRAS and BRAF mutations on survival of metastatic colorectal cancer after liver resection: a systematic review and meta-analysis. Clin Colorectal Cancer. Epub 2017 Jan 25. doi: 10.1016/j.clcc.2017.01.004.
39.
Zurück zum Zitat Mick R, Crowley JJ, Carroll RJ. Phase II clinical trial design for noncytotoxic anticancer agents for which time to disease progression is the primary endpoint. Control Clin Trials. 2000;21:343–59.CrossRefPubMed Mick R, Crowley JJ, Carroll RJ. Phase II clinical trial design for noncytotoxic anticancer agents for which time to disease progression is the primary endpoint. Control Clin Trials. 2000;21:343–59.CrossRefPubMed
40.
Zurück zum Zitat Buyse M, Quinaux E, Hendlisz A, Golfinopoulos V, Tournigand C, Mick R. Progression-free survival ratio as end point for phase II trials in advanced solid tumors. J Clin Oncol. 2011;29:e451–2.CrossRefPubMed Buyse M, Quinaux E, Hendlisz A, Golfinopoulos V, Tournigand C, Mick R. Progression-free survival ratio as end point for phase II trials in advanced solid tumors. J Clin Oncol. 2011;29:e451–2.CrossRefPubMed
41.
Zurück zum Zitat Overman MJ, Morris V, Kee B, Fogelman D, Xiao L, Eng C, et al. Utility of a molecular prescreening program in advanced colorectal cancer for enrollment on biomarker-selected clinical trials. Ann Oncol. 2016;27:1068–74.CrossRefPubMed Overman MJ, Morris V, Kee B, Fogelman D, Xiao L, Eng C, et al. Utility of a molecular prescreening program in advanced colorectal cancer for enrollment on biomarker-selected clinical trials. Ann Oncol. 2016;27:1068–74.CrossRefPubMed
42.
Zurück zum Zitat Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2016;6:147–53.CrossRefPubMed Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2016;6:147–53.CrossRefPubMed
43.
Zurück zum Zitat Le Tourneau C, Delord J-P, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–34.CrossRefPubMed Le Tourneau C, Delord J-P, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–34.CrossRefPubMed
44.
Zurück zum Zitat Tsimberidou A-M, Iskander NG, Hong DS, Wheler JJ, Falchook GS, Fu S, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res. 2012;18:6373–83.CrossRefPubMedPubMedCentral Tsimberidou A-M, Iskander NG, Hong DS, Wheler JJ, Falchook GS, Fu S, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res. 2012;18:6373–83.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Price TJ, Peeters M, Kim TW, Li J, Cascinu S, Ruff P, et al. Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): a randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 2014;15:569-79. Price TJ, Peeters M, Kim TW, Li J, Cascinu S, Ruff P, et al. Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): a randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 2014;15:569-79.
Metadaten
Titel
Pooled Analysis of Clinical Outcome of Patients with Chemorefractory Metastatic Colorectal Cancer Treated within Phase I/II Clinical Studies Based on Individual Biomarkers of Susceptibility: A Single-Institution Experience
verfasst von
Andrea Sartore-Bianchi
Alessio Amatu
Erica Bonazzina
Stefano Stabile
Laura Giannetta
Giulio Cerea
Ilaria Schiavetto
Katia Bencardino
Chiara Funaioli
Riccardo Ricotta
Tiziana Cipani
Michele Schirru
Valentina Gambi
Laura Palmeri
Giulia Carlo-Stella
Francesca Rusconi
Sara Di Bella
Giovanni Burrafato
Andrea Cassingena
Emanuele Valtorta
Calogero Lauricella
Federica Pazzi
Alessandra Gambaro
Silvia Ghezzi
Giovanna Marrapese
Emiliana Tarenzi
Silvio Veronese
Mauro Truini
Angelo Vanzulli
Salvatore Siena
Publikationsdatum
01.07.2017
Verlag
Springer International Publishing
Erschienen in
Targeted Oncology / Ausgabe 4/2017
Print ISSN: 1776-2596
Elektronische ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-017-0505-6

Weitere Artikel der Ausgabe 4/2017

Targeted Oncology 4/2017 Zur Ausgabe

Update Onkologie

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