Next Article in Journal
Distinct Signatures of Tumor-Associated Microbiota and Metabolome in Low-Grade vs. High-Grade Dysplastic Colon Polyps: Inference of Their Role in Tumor Initiation and Progression
Next Article in Special Issue
Cetuximab as a Key Partner in Personalized Targeted Therapy for Metastatic Colorectal Cancer
Previous Article in Journal
Resistance of Leukemia Cells to 5-Azacytidine: Different Responses to the Same Induction Protocol
Previous Article in Special Issue
Pretreatment Plasma Circulating Tumor DNA RAS/BRAF Mutational Status in Refractory Metastatic Colorectal Cancer Patients Who Are Candidates for Anti-EGFR Rechallenge Therapy: A Pooled Analysis of the CAVE and VELO Clinical Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Irinotecan- vs. Oxaliplatin-Based Doublets in KRASG12C-Mutated Metastatic Colorectal Cancer—A Multicentre Propensity-Score-Matched Retrospective Analysis

by
Vincenzo Formica
1,*,†,
Cristina Morelli
1,†,
Veronica Conca
2,
Maria Alessandra Calegari
3,
Jessica Lucchetti
4,
Emanuela Dell’Aquila
5,
Marta Schirripa
6,
Marco Messina
7,
Lisa Salvatore
3,8,
Federica Lo Prinzi
4,
Giovanni Dima
2,
Giovanni Trovato
3,8,
Silvia Riondino
1,
Mario Roselli
1,
Ferdinandos Skoulidis
9,
Hendrik-Tobias Arkenau
10 and
Chiara Cremolini
2
1
Medical Oncology Unit, Department of Medicine of the Systems, University of Rome Tor Vergata, 00133 Rome, Italy
2
Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
3
Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
4
Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy
5
Regina Elena National Cancer Institute, Medical Oncology 1, 00144 Rome, Italy
6
Department of Oncology and Hematology, Belcolle Hospital, Azienda Sanitaria Locale Viterbo, 01100 Viterbo, Italy
7
UOC Oncologia Medica, ARNAS Ospedali Civico Di Cristina Benefratelli, 90127 Palermo, Italy
8
Oncologia Medica, Università Cattolica del Sacro Cuore, 00186 Rome, Italy
9
Department of Thoracic-Head & Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
10
Ellipses Pharma, London W1J 8LG, UK
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work as first author.
Cancers 2023, 15(11), 3064; https://doi.org/10.3390/cancers15113064
Submission received: 17 April 2023 / Revised: 22 May 2023 / Accepted: 30 May 2023 / Published: 5 June 2023
(This article belongs to the Special Issue Cetuximab and Cancer)

Abstract

:

Simple Summary

The sensitivity to chemotherapy of KRASG12C-mutated colorectal cancer has been investigated to verify whether the combination of chemotherapy plus a KRASG12C-inhibitor might become the standard of care in the near future. To this aim, the present retrospective study was designed to assess the performance of irinotecan vs. oxaliplatin in the first-line treatment of KRASG12C-mutated mCRC patients and provide support for first-line decision making. In this setting of patients treated with FOLFIRI or FOLFOX +/− bevacizumab, irinotecan and oxaliplatin were compared using a propensity-score-matched analysis. The survival superiority of irinotecan was demonstrated over oxaliplatin in KRASG12C-mutated patients, while no differences were observed in a control cohort of KRASG12D-mutated patients. This should be considered when investigating chemotherapy plus targeted agent combinations.

Abstract

Background: KRASG12C-mutated metastatic colorectal cancer (mCRC) has recently been recognized as a distinct druggable molecular entity; however, there are limited data on its sensitivity to standard chemotherapy. In the near future, the combination of chemotherapy plus a KRASG12C-inhibitor might become the standard of care; however, the optimal chemotherapy backbone is unknown. Methods: A multicentre retrospective analysis was conducted including KRASG12C-mutated mCRC patients treated with first-line FOLFIRI or FOLFOX +/− bevacizumab. Both unmatched and propensity-score-matched analysis (PSMA) were conducted, with PSMA controlling for: previous adjuvant chemotherapy, ECOG PS, use of bevacizumab in first line, timing of metastasis appearance, time from diagnosis to first-line start, number of metastatic sites, presence of mucinous component, gender, and age. Subgroup analyses were also performed to investigate subgroup treatment–effect interactions. KRASG12D-mutated patients were analysed as control. Results: One hundred and four patients treated with irinotecan-(N = 47) or oxaliplatin-based (N = 57) chemotherapy were included. In the unmatched population, objective response rate (ORR) and median (m) progression-free and overall survival (mPFS and mOS) were comparable between the treatment arms. However, a late (>12 months) PFS advantage was observed with irinotecan (HR 0.62, p = 0.02). In the PSMA-derived cohort, a significant improvement with irinotecan vs. oxaliplatin was observed for both PFS and OS: 12- and 24-month PFS rates of 55% vs. 31% and 40% vs. 0% (HR 0.40, p = 0.01) and mOS 37.9 vs. 21.7 months (HR 0.45, p = 0.045), respectively. According to the subgroup analysis, interaction effects between the presence of lung metastases and treatment groups were found in terms of PFS (p for interaction = 0.08) and OS (p for interaction = 0.03), with a higher benefit from irinotecan in patients without lung metastases. No difference between treatment groups was observed in the KRASG12D-mutated cohort (N = 153). Conclusions: First-line irinotecan-based regimens provided better survival results in KRASG12C-mutated mCRC patients and should be preferred over oxaliplatin. These findings should also be considered when investigating chemotherapy plus targeted agent combinations.

1. Introduction

Colorectal cancer (CRC) represents the third most common cancer type and the fourth leading cause of cancer-related death in Western countries [1]. In about half of cases metastatic disease is observed, either at diagnosis or as metachronous occurrence [2]. Median overall survival (mOS) for metastatic CRC (mCRC) is currently 18–24 months in the unselected population, but it varies according to the RAS and BRAF mutational status and primary tumour side [3]. RAS-mutated tumours are associated with a worse prognosis than RAS and BRAF wild-type tumours [4], which is also due to intrinsic resistance to the anti-EGFR agents cetuximab and panitumumab [5]. Activating KRAS mutations are frequent across many solid tumour types, and in CRC they are present in approximately 40% of cases [6]. They have been considered undruggable for a long time due to their unstable inhibition by currently available drugs available.
More recently, stable inhibitors of KRASG12C-mutated protein have been developed and have demonstrated clinical benefit in phase I/II studies, as well as in combination with cetuximab [7], while phase III trials testing these agents against the standard of care in second or later lines of treatment are currently ongoing [8].
Nonetheless, today, the standard first-line therapy of KRASG12C-mutated mCRC patients remains irinotecan- or oxaliplatin-based doublets, with few data exploring possible differences between the two drugs in this specific molecular subtype. The aim of the present study was to retrospectively review the performance of irinotecan vs. oxaliplatin in the first-line treatment of KRASG12C-mutated mCRC patients and provide support for first-line decision making.

2. Patients and Methods

A multicentre retrospective analysis was conducted including patients from 7 Italian institutions with histologically confirmed CRC and measurable metastatic disease treated with a first-line chemotherapy. Only KRASG12C-mutated patients treated between 2009 and 2021 with either FOLFIRI +/− bevacizumab or FOLFOX +/− bevacizumab were selected. KRAS mutational status was determined using either real-time PCR, pyrosequencing, or next-generation sequencing according to local procedure. Patients with microsatellite instability high/mismatch repair deficient tumours and BRAF-mutated tumours were not included. For the present analysis, the cut-off date for survival follow-up of included patients was 25 March 2022. All patients signed informed consent for collection of anonymized data for retrospective analyses at the time of the first-line therapy. The research was carried out in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki).
Outcome measures were progression-free survival (PFS), defined as the time from first-line start until radiological or clinical disease progression or death; OS, defined as the time from first-line start until death from any cause; and radiological objective response rate (ORR) according to RECIST 1.1 criteria.
Survival curves were drawn according to the Kaplan–Meier method and compared with the log-rank test. Accompanying hazard ratios (HR) using a univariate Cox-proportional hazards model were estimated. Chi-square test was used for difference in categorical variables.
Difference between irinotecan and oxaliplatin was firstly assessed in the unmatched KRASG12C-mutated population. Subsequently, to overcome possible selection bias in choosing first-line irinotecan vs. oxaliplatin, a propensity score and exact matching (PSM-EM) analysis was performed, thus ensuring balanced key baseline characteristics between the two treatment groups. Propensity score was estimated upon the following possibly influencing variables: previous oxaliplatin-based adjuvant chemotherapy (yes vs. no), ECOG PS (0 vs. 1 or more), use of bevacizumab in combination with first-line chemotherapy (yes vs. no), timing of metastasis appearance (synchronous vs. metachronous), time elapsed from histological diagnosis to first-line commencement (< vs. >12 months), number of metastatic sites (1 vs. 2 or more), and presence of mucinous component (yes vs. no). Exact matching was performed by gender (male vs. female) and age (< vs. >65 years).
Pairs of irinotecan- and oxaliplatin-treated patients were matched on the logit of the propensity score using the near neighbour method and a calliper of width equal to 0.2 of the standard deviation of the logit of the propensity score.
Subgroup analyses for key outcome measures were also performed to investigate significant treatment effect/subgroup interaction and presented as forest plot with interaction p values.
Treatment activity was also investigated in a control population of patients harbouring the KRASG12D mutation exactly matched for gender and age and selected from the databases of the 7 participating centres in order to validate the KRASG12C-specificity of the primary findings.
A statistically significant finding was defined as a two-sided p value < 0.05. All analyses were carried out with R (version 4.0.3).

3. Results

Out of 4843 screened patients, 122 had KRASG12C-mutated (2.52%) mCRC, and 104 of them met the inclusion criteria and were treated with first-line FOLFIRI (12 patients), FOLFIRI plus bevacizumab (35 patients), FOLFOX (14 patients), or FOLFOX plus bevacizumab (43 patients). Patients’ characteristics are reported in Table 1. As of March 2022, 96 (92%) patients had reached the progression endpoint and 78 (75%) the survival endpoint. Median follow-up of surviving patients was 20.7 months (range 7.8 to 118.2 months). Second-line treatment was received by 52% of patients, in most cases including the cytotoxic agent not received in the first line. No patient received the KRAS-specific inhibitor.
In the whole cohort of 104 patients, ORR, mPFS, and mOS were 58%, 10.2 months (95% CI 9.1–11.6 months), and 24.9 months (95% CI 20.2–29.5 months), respectively.
In the unmatched analysis, ORR was 57% vs. 58% for irinotecan- vs. oxaliplatin-treated patients, respectively.
Furthermore, mPFS was comparable between irinotecan- vs. oxaliplatin-treated patients: 10.5 vs. 9.9 months, respectively. However, at approximately 12 months, the curves separated in favour of irinotecan with a significant improvement in 12-, 18- and 24-month PFS rates were 38% vs. 30%, 32% vs. 13%, and 22% vs. 4%, for irinotecan vs. oxaliplatin, respectively; HR 0.62 (95% CI 0.41–0.94) and p = 0.02 (Figure 1A).
A numerically higher mOS was observed for irinotecan as compared to oxaliplatin, with 28.9 vs. 21.1 months, respectively; HR 0.70 (95% CI 0.44–1.10) and p = 0.12 (Figure 1B).
To account for the imbalance in possible influencing factors, a PSM-EM analysis was performed. Background variables that were taken into account for patients treated with FOLFIRI +/− bevacizumab or FOLFOX +/− bevacizumab were the following: previous oxaliplatin-based adjuvant chemotherapy (yes vs. no), ECOG PS (0 vs. 1 or more), use of bevacizumab in combination with first-line chemotherapy (yes vs. no), timing of metastasis appearance (synchronous vs. metachronous), time elapsed from histological diagnosis to first-line commencement (< vs. >12 months), number of metastatic sites (1 vs. 2 or more), presence of mucinous component (yes vs. no), gender (male vs. female), and age (< vs. >65 years).
In total, 44 patients (22 patients per arm) were found to adequately match and were selected for the matched analysis. The advantage of irinotecan was even more pronounced in the PSM-EM-derived cohort, with 12-, 18- and 24-month PFS rates of 55% vs. 31%, 50% vs. 14%, and 40% vs. 0% for irinotecan vs. oxaliplatin, respectively; HR 0.40 (95% CI 0.20–0.83) and p = 0.01 (Figure 2A). The mOS was 37.9 vs. 21.7 months for irinotecan vs. oxaliplatin, respectively; HR 0.45 (95% CI 0.21–0.98) and p = 0.045 (Figure 2B). ORR was 73% vs. 68%, respectively.
In order to investigate whether the survival advantage of irinotecan was driven by specific clinical features, subgroup analyses were performed. Among all the tested subgroups, interaction effects between the presence or not of lung metastases and treatment groups were found in terms of PFS (p for interaction = 0.08) and OS (p for interaction = 0.03) (Figure 3). By analysing PFS and OS separately in the two subgroups of patients without and with lung metastases, the survival advantage of irinotecan was significant for both PFS and OS only in patients without lung metastases (62 patients): mPFS 11.3 vs. 9.1 months (HR 0.37 (95% CI 0.21–0.66), p = 0.0007) and mOS 27.2 vs. 17.3 months (HR 0.43 (95% CI 0.24–0.78), p = 0.005) for irinotecan and oxaliplatin, respectively (Figure 4A,B). ORR was numerically superior with irinotecan in this subgroup: 69% vs. 47%, with a chi-square p value 0.087. No survival difference by treatment was observed in patients with lung metastases, with p = 0.85 and 0.55 for PFS and OS, respectively (Figure 4C,D).
A population of 153 KRASG12D-mutated mCRC patients treated with either FOLFIRI +/− bevacizumab (61 patients) or FOLFOX +/− bevacizumab (92 patients) in the first-line setting, exactly matched for age and gender with the KRASG12C-mutated cohort, was used as control group. In KRASG12D-mutated patients, no difference was observed between irinotecan- and oxaliplatin-based regimens in terms of both PFS (p = 0.36) and OS (p = 0.29) (Figure 5A,B).

4. Discussion

In the present retrospective multicentre study including 104 KRASG12C-mutated mCRC patients treated with either FOLFIRI or FOLFOX +/− bevacizumab in the first-line setting, we were able to demonstrate a long-term survival benefit of irinotecan-based regimens in this specific molecular subtype. This effect was maintained and even reinforced after adjusting for possible imbalances with a propensity-score-matched analysis based on the receipt of adjuvant oxaliplatin, ECOG PS, bevacizumab within the first-line regimen, timing of metastasis appearance, time elapsed from histological diagnosis to first-line commencement, number of metastatic sites, presence of mucinous component, gender, and age.
The benefit of irinotecan was especially evident in patients without lung metastases, with a mOS > 27 months when KRASG12C-mutated patients were treated with an irinotecan-based first-line chemotherapy.
The superiority of irinotecan-based regimens could not be demonstrated in the control cohort of 153 patients harbouring the KRASG12D mutation.
The choice of a KRASG12D-mutated cohort as control was essentially based on the effort to make the control group as most homogeneous as possible and on the high prevalence of this alteration in CRC. Moreover, G12D-specific inhibitors are already under investigation in phase I trials (e.g., NCT05382559); therefore, collecting more information about patients with tumours harbouring this specific KRAS mutation is of interest.
This is not the first time that irinotecan was shown to be superior to oxaliplatin in KRAS-mutated mCRC. In a phase II randomized trial of the AIO cooperative group, where first-line capecitabine/irinotecan/bevacizumab was compared to capecitabine/oxaliplatin/bevacizumab in unselected patients, the irinotecan-based regimen showed superiority for OS in the KRAS codon 12–13-mutated subgroup (74 patients): mOS 28.7 vs. 18.8 months, respectively, p = 0.03 [9]. Analyses by sub-mutations in general, and for the KRASG12C mutation in particular, were not performed in this study.
In another retrospective series including 128 KRAS-mutated patients, a survival advantage with irinotecan-based first-line chemotherapy was also reported in codon 12 mutant patients (mOS 42.7 months) [10].
In a recent study by Ciardiello et al. including KRASG12C-mutated mCRC patients, a numerically higher mOS was observed in irinotecan-treated (29 patients) vs. oxaliplatin-treated patients (66 patients), at 22 vs. 18 months, respectively [11]. In this study, a superiority of the triplet regimen FOLFOXIRI administered to 16 patients was observed; however, no PSM analysis was performed. In our study, patients treated with FOLFOXIRI were excluded since the analysis was mainly focused on the difference between oxaliplatin and irinotecan in consideration of the fact that, in common clinical practice, most of patients are deemed candidates for a doublet regimen of either FOLFIRI or FOLFOX. The subgroup analyses of randomized trials of FOLFOXIRI versus FOLFIRI (TRIBE) or FOLFOX (TRIBE2) according to KRASG12C mutation would provide useful information about the added value of the intensified chemotherapy regimen in this patients’ subgroup.
A signal of different sensitivity to anticancer therapy based on the metastatic pattern was observed in our analysis. This has recently become a focus of research also in trials with immunotherapy, where lung and liver metastases from CRC seem to show distinct genomic profiles and immune microenvironments, which would explain a differential response to the treatment [12,13,14,15].
Some mutations, such as those affecting TCF7L2, APC, FLT3, TP53, and ZFP36L2, seem to preferentially occur in lung metastases with either a polyphyletic or monophyletic mechanism [16]. It would be of interest to specifically assess whether these mutations might interact with irinotecan or oxaliplatin sensitivity.
Moreover, a more in-depth assessment of baseline CT scans would be useful to work out the influence of disease burden on chemotherapy sensitivity. CT scans were not available for the analysis in our study; however, we used the total number of metastatic sites (‘2 or more’ vs. ‘1’) as a “proxy” of disease burden (Figure 3).
It is well known that rectal cancer more frequently spreads to lungs than colon cancer. In our cohort, only 10 patients had a rectal primary tumour, with half (n = 5) of them presenting with lung metastases. Because of the limited number of patients, no further stratified analyses were specifically conducted for rectal cancer patients.
Differences in the sites of metastatic spread according to KRAS mutational status were previously reported. In a study by Tie et al. [17], a higher prevalence of KRAS mutations in lung versus liver metastases was found (62% vs. 32%, p = 0.003), and a higher risk of lung relapse was evidenced among those with KRAS-mutated primary tumours (HR = 2.1, p = 0.007).
Intra-tumour heterogeneity between primary tumour and lung metastases has also been confirmed in KRASG12C-mutated patients [18]. However, whether this may influence sensitivity to irinotecan, or the interaction between drug pharmacodynamics and host microenvironment, is yet to be assessed [19,20]. The observed difference in chemotherapy sensitivity in KRASG12C-mutated patients is perhaps not due to the KRASG12C mutation itself but rather to co-occurring alterations that are possibly more frequent in KRASG12C-mutated tumours. Following this hypothesis, the study of pathways involving the topoisomerase I enzyme, the target of the active form of irinotecan SN38, would be recommended.
The evidence of interaction between treatment effect and pattern of metastases in KRASG12C-mutated mCRC is completely unprecedented and should be cautiously interpreted given that the present study is retrospective, and the subgroup analysis was unplanned. However, it would deserve further investigation.

5. Conclusions

In conclusion, our data suggest that, currently, in the absence of approved targeted agents in the first-line setting, when a doublet chemotherapy is chosen, irinotecan-based regimens should be preferred, especially in patients without lung metastases. Moreover, possible synergic effects should be taken into consideration if combinations of ‘chemo-targeted’ therapies with novel KRASG12C-inhibitors are tested in clinical trials, since these new drugs might either potentiate the effect of less efficacious regimens, such as oxaliplatin-based doublets, or further enhance the beneficial effect of more efficacious regimens, such as those that are irinotecan-based [21].

Author Contributions

Conceptualization: V.F., C.M., M.A.C., F.S., H.-T.A. and C.C.; methodology: V.F., S.R., M.R., F.S., H.-T.A. and C.C.; validation: V.F., C.M., V.C., M.A.C., J.L., E.D., M.S., M.M., L.S., F.L.P., G.D., G.T., S.R., M.R., F.S., H.-T.A. and C.C.; formal analysis: V.F. and C.M.; data curation: V.C., M.A.C., J.L., E.D., M.S., M.M., F.L.P., G.D. and G.T.; writing—original draft preparation: V.F., C.M., M.A.C., S.R., M.R., F.S., H.-T.A. and C.C.; writing—review and editing: V.F., C.M., V.C., M.A.C., J.L., E.D., M.S., M.M., L.S., F.L.P., G.D., G.T., S.R., M.R., F.S., H.-T.A. and C.C.; supervision: V.F., C.M., V.C., M.A.C., J.L., E.D., M.S., M.M., L.S., F.L.P., G.D., G.T., S.R., M.R., F.S., H.-T.A. and C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been partially funded by a grant from the European Project Horizon 2020 SC1-BHC-02-2019 [REVERT, GA n. 848098].

Institutional Review Board Statement

The study was approved by the Tor Vergata University Hospital IRB and Ethics Committee (record number 4.22 P.U.D.).

Informed Consent Statement

All patients signed informed consent for collection of anonymized data for retrospective analyses at the time of the first-line therapy.

Data Availability Statement

All raw anonymized data are available upon request via email to the corresponding author.

Conflicts of Interest

Formica Vincenzo declares honoraria from Amgen, Bayer, Servier, Pierre Fabre, and MSD. Morelli Cristina declares no conflict of interest. Conca Veronica declares no conflict of interest. Calegari Maria Alessandra declares no conflict of interest. Lucchetti Jessica declares no conflict of interest. Dell’Aquila Emanuela declares no conflict of interest. Schirripa Marta declares no conflict of interest. Messina Marco declares consultant for Sanofi, Elmaresearch, and Iquvia; congress honoraria and advisory board: Sanofi, Amgen, Novartis, AstraZeneca, Merck, Janssen, Archimedes Pharma, BMS, and MSD; congress support and sponsoring: Astellas, Janssen, Sanofi, Novartis, AstraZeneca, Eli Lilly, Roche, Amgen, Merk, BMS, Pfizer, Servier, and BMS. Salvatore Lisa declares speaker’s and consultant’s fee from MSD, Astra-Zeneca, Servier, Bayer, Merck, Amgen, and Pierre-Fabre. Lo Prinzi Federica declares no conflict of interest. Dima Giovanni declares no conflict of interest. Trovato Giovanni declares no conflict of interest. Riondino Silvia declares no conflict of interest. Roselli Mario declares no conflict of interest. Ferdinandos Skoulidis declares consultant for Astra Zeneca, Novartis, BeiGene, Guardant Health, BergenBio, Navire Pharma, Tango Therapeutics, Calithera Biosciences, Intellisphere LLC, and Medscape LLC; research grant from Mirati, Pfizer, and Merck; stock interest in BioNTech and Moderna; speaker’s fee from VSPO McGill Universitè de Montreal and RV Mais Promocao Eventos LTDS. Arkenau Hendrik-Tobias declares employment in Ellipses Pharma and Cremolini Chiara declares honoraria from Roche, Amgen, Merck, MSD, Pierre Fabre, Nordic Pharma, Bayer, Servier, and Organon; research grants from Merck, Servier, and Bayer.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Sauer, A.G.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 145–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Formica, V.; Sera, F.; Cremolini, C.; Riondino, S.; Morelli, C.; Arkenau, H.-T.; Roselli, M. KRAS and BRAF Mutations in Stage II and III Colon Cancer: A Systematic Review and Meta-Analysis. Gynecol. Oncol. 2022, 114, 517–527. [Google Scholar] [CrossRef] [PubMed]
  4. Modest, D.P.; Ricard, I.; Heinemann, V.; Hegewisch-Becker, S.; Schmiegel, W.; Porschen, R.; Stintzing, S.; Graeven, U.; Arnold, D.; von Weikersthal, L.F.; et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: Pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann. Oncol. 2016, 27, 1746–1753. [Google Scholar] [CrossRef] [PubMed]
  5. Yoshino, T.; Arnold, D.; Taniguchi, H.; Pentheroudakis, G.; Yamazaki, K.; Xu, R.-H.; Kim, T.; Ismail, F.; Tan, I.; Yeh, K.-H.; et al. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: A JSMO–ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann. Oncol. 2018, 29, 44–70. [Google Scholar] [CrossRef]
  6. Morris, V.K.; Kennedy, E.B.; Baxter, N.N.; Benson, A.B.; Cercek, A.; Cho, M.; Ciombor, K.K.; Cremolini, C.; Davis, A.; Deming, D.A.; et al. Treatment of Metastatic Colorectal Cancer: ASCO Guideline. J. Clin. Oncol. 2022, 41, 678–700. [Google Scholar] [CrossRef]
  7. Yaeger, R.; Weiss, J.; Pelster, M.S.; Spira, A.I.; Barve, M.; Ou, S.-H.I.; Leal, T.A.; Bekaii-Saab, T.S.; Paweletz, C.P.; Heavey, G.A.; et al. Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated KRAS G12C. N. Engl. J. Med. 2023, 388, 44–54. [Google Scholar] [CrossRef]
  8. Ji, J.; Wang, C.; Fakih, M. Targeting KRASG12C-Mutated Advanced Colorectal Cancer: Research and Clinical Developments. OncoTargets Ther. 2022, 15, 747–756. [Google Scholar] [CrossRef]
  9. Schmiegel, W.; Reinacher-Schick, A.; Arnold, D.; Kubicka, S.; Freier, W.; Dietrich, G.; Geißler, M.; Hegewisch-Becker, S.; Tannapfel, A.; Pohl, M.; et al. Capecitabine/irinotecan or capecitabine/oxaliplatin in combination with bevacizumab is effective and safe as first-line therapy for metastatic colorectal cancer: A randomized phase II study of the AIO colorectal study group. Ann. Oncol. 2013, 24, 1580–1587. [Google Scholar] [CrossRef]
  10. Ergun, Y.; Acikgoz, Y.; Bal, O.; Ucar, G.; Dirikoc, M.; Yildirim, E.C.; Akdeniz, N.; Uncu, D. KRAS codon 12 and 13 mutations may guide the selection of irinotecan or oxaliplatin in first-line treatment of metastatic colorectal cancer. Expert Rev. Mol. Diagn. 2019, 19, 1131–1140. [Google Scholar] [CrossRef]
  11. Ciardiello, D.; Chiarazzo, C.; Famiglietti, V.; Damato, A.; Pinto, C.; Zampino, M.; Castellano, G.; Gervaso, L.; Zaniboni, A.; Oneda, E.; et al. Clinical efficacy of sequential treatments in KRASG12C-mutant metastatic colorectal cancer: Findings from a real-life multicenter Italian study (CRC-KR GOIM). ESMO Open 2022, 7, 100567. [Google Scholar] [CrossRef] [PubMed]
  12. Fakih, M.G.; Sandhu, J.; Lim, D.; Li, S.M.; Wang, C. 320MO A phase I clinical trial of regorafenib, ipilimumab, and nivolumab (RIN) in chemotherapy resistant MSS metastatic colorectal cancer (mCRC). Ann. Oncol. 2022, 33, S684. [Google Scholar] [CrossRef]
  13. Mettu, N.B.; Ou, F.-S.; Zemla, T.J.; Halfdanarson, T.R.; Lenz, H.-J.; Breakstone, R.A.; Boland, P.M.; Crysler, O.V.; Wu, C.; Nixon, A.B.; et al. Assessment of Capecitabine and Bevacizumab with or without Atezolizumab for the Treatment of Refractory Metastatic Colorectal Cancer. JAMA Netw. Open 2022, 5, e2149040. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, C.; Sandhu, J.; Ouyang, C.; Ye, J.; Lee, P.P.; Fakih, M. Clinical Response to Immunotherapy Targeting Programmed Cell Death Receptor 1/Programmed Cell Death Ligand 1 in Patients with Treatment-Resistant Microsatellite Stable Colorectal Cancer with and without Liver Metastases. JAMA Netw. Open 2021, 4, e2118416. [Google Scholar] [CrossRef]
  15. Formica, V.; Cereda, V.; Nardecchia, A.; Tesauro, M.; Roselli, M. Immune reaction and colorectal cancer: Friends or foes? World J. Gastroenterol. 2014, 20, 12407–12419. [Google Scholar] [CrossRef]
  16. Chen, H.-N.; Shu, Y.; Liao, F.; Liao, X.; Zhang, H.; Qin, Y.; Wang, Z.; Luo, M.; Liu, Q.; Xue, Z.; et al. Genomic evolution and diverse models of systemic metastases in colorectal cancer. Gut 2022, 71, 322–332. [Google Scholar] [CrossRef]
  17. Tie, J.; Lipton, L.; Desai, J.; Gibbs, P.; Jorissen, R.N.; Christie, M.; Drummond, K.J.; Thomson, B.N.; Usatoff, V.; Evans, P.M.; et al. KRAS Mutation Is Associated with Lung Metastasis in Patients with Curatively Resected Colorectal Cancer. Clin. Cancer Res. 2011, 17, 1122–1130. [Google Scholar] [CrossRef] [Green Version]
  18. Jo, P.; Bernhardt, M.; Nietert, M.; König, A.; Azizian, A.; Schirmer, M.A.; Grade, M.; Kitz, J.; Reuter-Jessen, K.; Ghadimi, M.; et al. KRAS mutation status concordance between the primary tumor and the corresponding metastasis in patients with rectal cancer. PLoS ONE 2020, 15, e0239806. [Google Scholar] [CrossRef]
  19. Roselli, M.; Formica, V.; Cereda, V.; Jochems, C.; Richards, J.; Grenga, I.; Orlandi, A.; Ferroni, P.; Guadagni, F.; Schlom, J. The association of clinical outcome and peripheral T-cell subsets in metastatic colorectal cancer patients receiving first-line FOLFIRI plus bevacizumab therapy. Oncoimmunology 2016, 5, e1188243. [Google Scholar] [CrossRef] [Green Version]
  20. Formica, V.; Cereda, V.; di Bari, M.-G.; Grenga, I.; Tesauro, M.; Raffaele, P.; Ferroni, P.; Guadagni, F.; Roselli, M. Peripheral CD45RO, PD-1, and TLR4 expression in metastatic colorectal cancer patients treated with bevacizumab, fluorouracil, and irinotecan (FOLFIRI-B). Med Oncol. 2013, 30, 1–8. [Google Scholar] [CrossRef]
  21. Masuishi, T.; Kuboki, Y.; Fakih, M.; Strickler, J.; Furqan, M.; Kim, E.; Cardona, P.; Tran, Q.; Chan, E.; Hong, D. 444TiP Trial in progress: A phase Ib study of sotorasib, a selective KRAS G12C inhibitor, in combination with panitumumab and FOLFIRI in treatment naïve and previously treated metastatic colorectal cancer (CodeBreaK 101). Ann. Oncol. 2022, 33, S737–S738. [Google Scholar] [CrossRef]
Figure 1. PFS (panel (A)) and OS (panel (B)) analysis for first-line irinotecan-based (FOLFIRI +/− bevacizumab) vs. oxaliplatin-based (FOLFOX +/− bevacizumab) chemotherapy doublet in the unmatched primary population of 104 KRASG12C-mutated metastatic colorectal cancer patients. IRI-based: irinotecan-based. OXA-based: oxaliplatin-based. PFS: progression-free survival. OS: overall survival.
Figure 1. PFS (panel (A)) and OS (panel (B)) analysis for first-line irinotecan-based (FOLFIRI +/− bevacizumab) vs. oxaliplatin-based (FOLFOX +/− bevacizumab) chemotherapy doublet in the unmatched primary population of 104 KRASG12C-mutated metastatic colorectal cancer patients. IRI-based: irinotecan-based. OXA-based: oxaliplatin-based. PFS: progression-free survival. OS: overall survival.
Cancers 15 03064 g001
Figure 2. PFS (panel (A)) and OS (panel (B)) analysis for first-line irinotecan-based (FOLFIRI +/− bevacizumab) vs. oxaliplatin-based (FOLFOX +/− bevacizumab) chemotherapy doublet in the propensity score matching analysis (PSMA)-derived population of 44 KRASG12C-mutated metastatic colorectal cancer patients. Propensity score was based on receipt of adjuvant oxaliplatin, ECOG PS, receipt of bevacizumab within the first-line regimen, timing of metastasis appearance, time elapsed from histological diagnosis to first-line commencement, number of metastatic sites, presence of mucinous component, gender, and age. IRI-based: irinotecan-based. OXA-based: oxaliplatin-based. PFS: progression-free survival. OS: overall survival.
Figure 2. PFS (panel (A)) and OS (panel (B)) analysis for first-line irinotecan-based (FOLFIRI +/− bevacizumab) vs. oxaliplatin-based (FOLFOX +/− bevacizumab) chemotherapy doublet in the propensity score matching analysis (PSMA)-derived population of 44 KRASG12C-mutated metastatic colorectal cancer patients. Propensity score was based on receipt of adjuvant oxaliplatin, ECOG PS, receipt of bevacizumab within the first-line regimen, timing of metastasis appearance, time elapsed from histological diagnosis to first-line commencement, number of metastatic sites, presence of mucinous component, gender, and age. IRI-based: irinotecan-based. OXA-based: oxaliplatin-based. PFS: progression-free survival. OS: overall survival.
Cancers 15 03064 g002
Figure 3. Subgroup analysis for treatment effect–key variables interaction for PFS (A) and OS (B). The dashed line and grey areas indicate the log-hazard ratio and 90% confidence intervals in the whole population of 104 patients. PFS: progression-free survival. OS: overall survival. CI: confidence interval. Dx: diagnosis.
Figure 3. Subgroup analysis for treatment effect–key variables interaction for PFS (A) and OS (B). The dashed line and grey areas indicate the log-hazard ratio and 90% confidence intervals in the whole population of 104 patients. PFS: progression-free survival. OS: overall survival. CI: confidence interval. Dx: diagnosis.
Cancers 15 03064 g003
Figure 4. PFS (panels (A,C)) and OS (panels (B,D)) by treatment and absence (panels (A,B)) or presence (panels (C,D)) of lung metastasis in the primary population of 104 KRASG12C-mutated metastatic colorectal cancer patients. w/o: without. w/: with. IRI-based: irinotecan-based chemotherapy. OXA-based: oxaliplatin-based chemotherapy. PFS: progression-free survival. OS: overall survival.
Figure 4. PFS (panels (A,C)) and OS (panels (B,D)) by treatment and absence (panels (A,B)) or presence (panels (C,D)) of lung metastasis in the primary population of 104 KRASG12C-mutated metastatic colorectal cancer patients. w/o: without. w/: with. IRI-based: irinotecan-based chemotherapy. OXA-based: oxaliplatin-based chemotherapy. PFS: progression-free survival. OS: overall survival.
Cancers 15 03064 g004
Figure 5. PFS (panel (A)) and OS (panel (B)) by treatment group in 153 patients with KRASG12D-mutated mCRC treated with first-line chemotherapy. OXA-based: oxaliplatin-based chemotherapy. PFS: progression-free survival. OS: overall survival.
Figure 5. PFS (panel (A)) and OS (panel (B)) by treatment group in 153 patients with KRASG12D-mutated mCRC treated with first-line chemotherapy. OXA-based: oxaliplatin-based chemotherapy. PFS: progression-free survival. OS: overall survival.
Cancers 15 03064 g005
Table 1. Characteristics of the 104 KRASG12C-mutated metastatic colorectal cancer patients included in the study.
Table 1. Characteristics of the 104 KRASG12C-mutated metastatic colorectal cancer patients included in the study.
Characteristics% (N = 104)
Sex
Male40% (42)
Female60% (62)
Age
<6554% (56)
≥6546% (48)
Primary tumour sidedness
Right37% (38)
Left63% (66)
Mucinous histology
Yes14% (15)
No86% (89)
Metastasis at diagnosis
Yes69% (72)
No31% (32)
Number of metastatic sites
141% (43)
≥259% (61)
Metastatic sites
Liver77% (80)
Lung41% (43)
Peritoneum22% (23)
First-line treatment
Oxaliplatin-based doublet55% (57)
Irinotecan-based doublet45% (47)
Bevacizumab use in first line
Yes75% (78)
No25% (26)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Formica, V.; Morelli, C.; Conca, V.; Calegari, M.A.; Lucchetti, J.; Dell’Aquila, E.; Schirripa, M.; Messina, M.; Salvatore, L.; Lo Prinzi, F.; et al. Irinotecan- vs. Oxaliplatin-Based Doublets in KRASG12C-Mutated Metastatic Colorectal Cancer—A Multicentre Propensity-Score-Matched Retrospective Analysis. Cancers 2023, 15, 3064. https://doi.org/10.3390/cancers15113064

AMA Style

Formica V, Morelli C, Conca V, Calegari MA, Lucchetti J, Dell’Aquila E, Schirripa M, Messina M, Salvatore L, Lo Prinzi F, et al. Irinotecan- vs. Oxaliplatin-Based Doublets in KRASG12C-Mutated Metastatic Colorectal Cancer—A Multicentre Propensity-Score-Matched Retrospective Analysis. Cancers. 2023; 15(11):3064. https://doi.org/10.3390/cancers15113064

Chicago/Turabian Style

Formica, Vincenzo, Cristina Morelli, Veronica Conca, Maria Alessandra Calegari, Jessica Lucchetti, Emanuela Dell’Aquila, Marta Schirripa, Marco Messina, Lisa Salvatore, Federica Lo Prinzi, and et al. 2023. "Irinotecan- vs. Oxaliplatin-Based Doublets in KRASG12C-Mutated Metastatic Colorectal Cancer—A Multicentre Propensity-Score-Matched Retrospective Analysis" Cancers 15, no. 11: 3064. https://doi.org/10.3390/cancers15113064

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop