Maintenance strategies in first-line treatment for RAS/BRAF wild-type metastatic colorectal cancer—one size does not fit all
Editorial Commentary

Maintenance strategies in first-line treatment for RAS/BRAF wild-type metastatic colorectal cancer—one size does not fit all

Per Pfeiffer1,2 ORCID logo, Gabor Liposits3 ORCID logo, Line Tarpgaard1,2 ORCID logo

1Department of Oncology, Odense University Hospital, Odense, Denmark; 2Department of Clinical Research, University of Southern Denmark, Odense, Denmark; 3Department of Oncology, Gødstrup Hospital, Herning, Denmark

Correspondence to: Per Pfeiffer, MD, PhD. Department of Oncology, Odense University Hospital, J. B. Winsløws Vej 4, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark. Email: per.pfeiffer@rsyd.dk.

Comment on: Pinto C, Orlandi A, Normanno N, et al. Fluorouracil, Leucovorin, and Irinotecan Plus Cetuximab Versus Cetuximab as Maintenance Therapy in First-Line Therapy for RAS and BRAF Wild-Type Metastatic Colorectal Cancer: Phase III ERMES Study. J Clin Oncol 2024;42:1278-87.


Keywords: Metastatic colorectal cancer (mCRC); RAS wild-type; epidermal growth factor receptor inhibitors (EGFRi); maintenance therapy


Received: 03 April 2024; Accepted: 13 June 2024; Published online: 18 July 2024.

doi: 10.21037/actr-24-30


Colorectal cancer (CRC) is the third most prevalent cancer worldwide with close to 1.9 million new patients diagnosed every year (1). In almost half of these patients, metastatic CRC (mCRC) will be detected; either synchronous mCRC when metastases are present at the time of the primary diagnosis or metachronous disease when relapse occurs after curative intended treatment. Nearly 50% of CRC patients die each year.

Currently, first-line chemotherapy consists of a combination of 5-fluorouracil (5-FU) and irinotecan followed by 5-FU and oxaliplatin upon progression (or vice versa); depending on the RAS, BRAF, and mismatch repair (MMR) status, targeted agents are frequently added. An anti-angiogenic drug (e.g., bevacizumab) is often used in patients independently of the molecular subtypes, whereas epidermal growth factor receptor inhibitors (EGFRi) as cetuximab or panitumumab are designated for selected patients whose tumors are RAS and BRAF wild-type (2,3).

RAS (both KRAS and NRAS) and BRAF wild-type mCRC represents a distinct subtype of CRC concerning both prognosis and therapeutic options manifesting in a median overall survival (OS) of about 36 months for selected fit patients included in clinical trials. According to current guidelines, the EGFRi combined with a 5-FU-based doublet [5-FU, leucovorin, and irinotecan (FOLFIRI)/5-FU, leucovorin, and oxaliplatin (FOLFOX)] are the cornerstones of systemic treatment (2,3). This combination frequently results in remarkable response rates and tumor shrinkage, making metastasectomy, e.g., liver resection and/or a local ablative approach feasible even in initially unresectable metastatic cases, improving OS. The OS benefit of first-line EGFRi treatment is limited to patients with left-sided primary tumors; patients with right-sided tumors might also achieve significant response, however, without OS benefit (2,3).

In patients with incurable diseases, where the aim is to relief symptoms and prolong life, balancing efficacy and quality of life (QoL) is paramount. During the continuum of care, keeping toxicities at an acceptable level is crucial for the patients. Induction therapy with doublet chemotherapy and an EGFRi typically lasts 4–6 months resulting in cytoreduction/tumor shrinkage in most cases; the optimal duration of first-line induction treatment is though unknown yet. Considering the EGFRi, skin and gastrointestinal toxicities are the most prevalent, and patients often need treatment breaks despite the comprehensive supportive care provided. The use of an oxaliplatin containing chemotherapy backbone (most often FOLFOX) causes potentially disabling cumulative neurotoxicity, leading to dose-reduction of oxaliplatin or treatment break mostly after about 4 months of therapy.

In contrast, irinotecan containing regimens (FOLFIRI) are more suitable for continuous treatment given their safety profile.

Guidelines recommend continuous treatment of patients with unresectable mCRC until progressive disease (PD), severe toxicity, or the patient’s wish for a treatment break (2,3). In clinical practice, this strategy is frequently followed, but even though progression-free survival (PFS) might reach 12 months in selected patients included in clinical trials, the median duration of therapy rarely reach 9 months due to adverse events and frequent, time-consuming visits to healthcare facilities.

To maintain efficacy while ensuring the best possible QoL for the patients, different strategies regarding the duration and intensity of treatment have been investigated. The following three main strategies are well established:

  • Continuous strategy of applying induction treatment until PD with dose reduction if needed;
  • Maintenance strategy using induction treatment for a planned period followed by a less intensive regimen until PD;
  • Intermittent strategy where treatment is completely stopped after a planned period of induction therapy, patients obtain treatment-free interval (TFI) with re-introduction of the same regimen when progression occurs.

Regarding the maintenance strategy in RAS/BRAF wild-type mCRC, clinical trials have focused on the following approaches: EGFRi monotherapy, 5-FU alone, or the combination of those: EGFRi and 5-FU.

The recently published ERMES trial is the largest randomized trial testing the importance of a maintenance strategy in chemo-naïve patients with RAS/BRAF wild-type mCRC (4). Close to 600 patients started induction therapy with FOLFIRI + cetuximab and patients were randomized upfront to continuous induction therapy (arm A) or maintenance cetuximab after eight cycles of induction regimen (arm B), if no sign of PD. The co-primary endpoints of the ERMES study were non-inferior PFS [hazard ratio (HR): 1.33] in the modified per-protocol (mPP) population (patients receiving more than eight cycles of therapy) and superior safety profile. However, differences in incidence rate of severe adverse events would only have been tested if the null-hypothesis of PFS inferiority was rejected.

A higher than expected number of patients dropped out before starting cycle 9 (at the time of maintenance) and for unknown reason more patients dropped out in arm A 48% vs. 38%. Typically, in randomized trials, 60–70% of patients starting induction therapy continue with therapy beyond 4 months.

In the mPP population, there was a difference in PFS {12.2 (arm A) vs. 10.0 months (arm B); HR: 1.3 [95% confidence interval (CI): 1.03 to 1.64]} but non-inferiority could not be confirmed (the upper limit of 95% CI of HR crossed the non-inferiority boundary of 1.33 with a non-inferiority P value of 0.43). On the contrary, the median OS was numerically but not significantly longer in patients receiving maintenance therapy with cetuximab monotherapy (30.7 vs. 35.7 months, P=0.12). There was no difference in response rates 68% vs. 72% as expected as randomized trials have shown that most and deepest responses occurred within the first 4 months of therapy (5). As projected, patients in arm B had a lower incidence of severe adverse events during maintenance therapy 20% vs. 35%. The authors concluded that the ERMES study did not confirm the non-inferiority of maintenance with cetuximab monotherapy.

Thus, the results of the ERMES study suggest that maintenance strategy should contain a cytotoxic component and this is in line with the current European Society for Medical Oncology (ESMO) guidelines (3). Interestingly, when FOLFIRI is combined with EGFRi in first line, ESMO recommends continuous treatment until PD, given FOLFIRI regimen is not associated with cumulative toxicity.

A number of comparable (Table 1) but also slightly different randomized trials have evaluated different schedules of maintenance therapy with EGFRi.

Table 1

Overview of randomized trials evaluating maintenance therapy after 12–16 weeks of induction therapy including EGFRi in patients with (K)RAS wild-type mCRC

Author, journal, year Trial name N Therapy Dur (weeks) Maintenance PFS (months), median OS (months), median
Wasan (6), Lancet Oncol, 2014 COIN-B 78 Randomization 12 TFI (3+) 3.1 16.8
KRASwt (retro) 91 FOLFOX + Cet ×6 Cet (3+) 5.8 22.2
Aranda (7), Eur J Cancer, 2018 MACRO2 64 Randomization 16 Cet 9 23
KRASwt 129 FOLFOX + Cet ×8 FOLFOX + Cet 10 27
Pietrantonio (8), JAMA Oncol, 2019 Valentino 115 Randomization 16 FOLF + Pan 12.0 ~28.0
RASwt 111 FOLFOX + Pan ×8 Pan 9.9 ~28.0
Modest (9), Int J Cancer, 2019 PanaMa 188 FOLFOX + Pan ×6 12 FOLF + Pan (3+) 8.8 (3+) 28.7
RASwt 189 Randomization FOLF (3+) 5.7 (3+) 25.7
Boige (10), J Clin Oncol, 2021 PRODIGE 28 214 FOLFIRI + Cet ×8 16 Cet (4+) 5.3 (4+) 24.8
RASwt Randomization TFI (4+) 2.0 (4+) 19.7
Pinto (4), J Clin Oncol, 2024 ERMES 296 Randomization 16 FOLFIRI + Cet 12.2 30.7
RASwt 297 FOLFIRI + Cet ×8 Cet 10.0 35.7
Avallone (11), J Clin Oncol, 2023 IMPROVE 69 Randomization 16 FOLFIRI + Cet 11.4 31.0
RASwt 68 FOLFIRI + Cet ×8 TFI 18.1 32.2
Munemoto (12), Eur J Cancer, 2019 SAPPHIRE 82 FOLFOX + Pan 12 FOLFOX + Pan 9.1 NR
RASwt (retro) 82 Randomization FOLF + Pan 9.3 NR
Cremolini (13), JAMA Oncol, 2018 Macbeth 59 Randomization 16 Cet 10.1 33.2
RASwt 57 FOLFOXIRI + Cet Bev 9.3 32.2

EGFR, epidermal growth factor receptor inhibitors; mCRC, metastatic colorectal cancer; N, number of patients starting induction therapy; Dur, duration of induction therapy; PFS, progression-free survival; OS, overall survival; wt, wild-type; retro, retrospective; FOLFOX, 5-FU, leucovorin, and oxaliplatin; Cet, cetuximab; TFI, treatment-free interval; FOLF, 5-FU and leucovorin; Pan, panitumumab; FOLFIRI, 5-FU, leucovorin, and irinotecan; NR, not reported or not tested; Bev, bevacizumab.

In six studies (4,6-8,13) patients were randomized before induction therapy, whereas in others (9,10,12), patients without any sign of PD after 12–16 weeks of induction therapy were randomized to EGFRi with or without chemotherapy, or even TFI.

Interpretation of efficacy data from these randomized trials is not straightforward; therefore, it would be obvious but also challenging to perform a systematic review with a meta-analysis to reach a conclusion. Four recent meta-analysis (14-17) included different number of studies, the conclusions were slightly different and none included the ERMES trial. Briefly, Raimondi et al. and Parisi et al. were in favor of the use of 5-FU plus EGFRi (14,15), while Petrelli et al. recommended doublet chemotherapy + EGFRi until PD as the best strategy concerning OS (16). Finally, Mastrantoni et al. concluded that doublet cytotoxic therapy + EGFRi, 5-FU + EGFRi, and EGFRi monotherapy resulted in similar OS (17). However, no study showed a difference in OS.

The development of resistance may be a drawback with continuous exposure to EGFRi therapy (18,19) but this was not supported by two published studies (6,10) which both favored continuous exposure to EGFRi therapy (numerically longer PFS and OS) compared to TFI (after 12 and 16 weeks of induction therapy, respectively). These data are absolutely in contrast to the unpublished data from the IMPROVE study (11). IMPROVE is a small randomized non-comparative phase II study which evaluated induction therapy (FOLFIRI + panitumumab) until PD or an intermittent treatment strategy with eight cycles of induction therapy followed by TFI until PD when another eight cycles of induction regimen was restarted and so on, until PD during therapy. PFS on treatment favored the intermittent arm (11.4 vs. 18.1 months) suggesting that treatment break might be beneficial regarding the tumor biology. Of note, patients receiving intermittent treatment also experienced fewer toxicities. There was no difference in OS (31.0 vs. 32.2 months).

Resistant tumor clones may rapidly grow during EGFRi therapy and decline during EGFRi treatment breaks (20-22). Clinical evidence supports such a strategy in late-line EGFRi rechallenge or re-introduction after prior exposure to EGFRi in first-line therapy and the same biological rationale could be argued for TFI as part of first-line treatment (23). The intermittent strategy of IMPROVE resulted in a remarkable PFS with reduced skin toxicity. A small subgroup was tested with circulating tumor DNA (ctDNA) but only a minor fraction (23%) developed a classic pattern (RAS, PI3K) of resistance. Based on the encouraging results of the IMPROVE study, the authors are planning a confirmatory phase 3 study.

The understanding of resistance mechanisms against anti-EGFR monoclonal antibodies in mCRC is continually evolving. Clinically, anti-EGFR rechallenge, both with and without ctDNA monitoring has been adopted (24). Enhancing patient selection and ctDNA analysis require a thorough understanding of the nature of resistance to anti-EGFR agents considering both the therapy line and the agents anti-EGFR are combined with. The findings are promising but their generalizability is limited due to small sample sizes. To effectively translate these findings into clinical practice, further prospective evidence from large-scale clinical trials is needed to better understand the underlying mechanism and develop strategies to overcome genomic and nongenomic resistance.

In conclusion, several randomized trials and meta-analyses evaluated the effect of maintenance treatment but the optimal strategy still remains unknown. In addition, none of the studies tested or discussed the optimal duration of induction therapy, however, 4 months seems to be a reasonable choice balancing effect and toxicity. Even with the publication of the large ERMES trial, there are still “no size that fits all patients”. Maintenance strategy with 5-FU and EGFRi after 4 months of induction therapy is a safe option, but an intermittent EGFRi-based strategy could be an interesting alternative, especially in patients with left-sided disease. In any case, potential side effects and patient preference should also be an important consideration when deciding whether or not to use maintenance therapy.

There is a need for randomized studies that follow the development of resistance (ctDNA) but until that time, shared decision-making on maintenance treatment must be based on tumor biology, clinical characteristics, and patient preferences.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, AME Clinical Trials Review. The article has undergone external peer review.

Peer Review File: Available at https://actr.amegroups.com/article/view/10.21037/actr-24-30/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-30/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2016;27:1386-422. [Crossref] [PubMed]
  3. Cervantes A, Adam R, Roselló S, et al. Metastatic colorectal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023;34:10-32. [Crossref] [PubMed]
  4. Pinto C, Orlandi A, Normanno N, et al. Fluorouracil, Leucovorin, and Irinotecan Plus Cetuximab Versus Cetuximab as Maintenance Therapy in First-Line Therapy for RAS and BRAF Wild-Type Metastatic Colorectal Cancer: Phase III ERMES Study. J Clin Oncol 2024;42:1278-87. [Crossref] [PubMed]
  5. Stintzing S, Modest DP, Rossius L, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer (FIRE-3): a post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet Oncol 2016;17:1426-34. [Crossref] [PubMed]
  6. Wasan H, Meade AM, Adams R, et al. Intermittent chemotherapy plus either intermittent or continuous cetuximab for first-line treatment of patients with KRAS wild-type advanced colorectal cancer (COIN-B): a randomised phase 2 trial. Lancet Oncol 2014;15:631-9. [Crossref] [PubMed]
  7. Aranda E, García-Alfonso P, Benavides M, et al. First-line mFOLFOX plus cetuximab followed by mFOLFOX plus cetuximab or single-agent cetuximab as maintenance therapy in patients with metastatic colorectal cancer: Phase II randomised MACRO2 TTD study. Eur J Cancer 2018;101:263-72. [Crossref] [PubMed]
  8. Pietrantonio F, Morano F, Corallo S, et al. Maintenance Therapy With Panitumumab Alone vs Panitumumab Plus Fluorouracil-Leucovorin in Patients With RAS Wild-Type Metastatic Colorectal Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2019;5:1268-75. [Crossref] [PubMed]
  9. Modest DP, Rivera F, Bachet JB, et al. Panitumumab-based maintenance after oxaliplatin discontinuation in metastatic colorectal cancer: A retrospective analysis of two randomised trials. Int J Cancer 2019;145:576-85. [Crossref] [PubMed]
  10. Boige V, Francois E, Ben Abdelghani M, et al. Maintenance treatment with cetuximab versus observation in RAS wild-type metastatic colorectal cancer: Results of the randomized phase II PRODIGE 28-time UNICANCER study. J Clin Oncol 2021;39:15. [Crossref]
  11. Avallone A, Giuliani F, Nasti G, et al. Intermittent or continuous panitumumab plus FOLFIRI (FOLFIRI/PANI) for first-line treatment of patients (pts) with RAS/BRAF wild-type (wt) metastatic colorectal cancer (mCRC): An update of survival/toxicity and preliminary results of genomic alterations from IMPROVE study. J Clin Oncol 2023;41:3571. [Crossref]
  12. Munemoto Y, Nakamura M, Takahashi M, et al. SAPPHIRE: a randomised phase II study of planned discontinuation or continuous treatment of oxaliplatin after six cycles of modified FOLFOX6 plus panitumumab in patients with colorectal cancer. Eur J Cancer 2019;119:158-67. [Crossref] [PubMed]
  13. Cremolini C, Antoniotti C, Lonardi S, et al. Activity and Safety of Cetuximab Plus Modified FOLFOXIRI Followed by Maintenance With Cetuximab or Bevacizumab for RAS and BRAF Wild-type Metastatic Colorectal Cancer: A Randomized Phase 2 Clinical Trial. JAMA Oncol 2018;4:529-36. [Crossref] [PubMed]
  14. Parisi A, Ghidini M, Giampieri R, et al. Post-induction Strategies in Metastatic Colorectal Cancer Patients Treated With First-Line Anti-EGFR-Based Treatment: A Systematic Review and Meta-Analysis. Clin Colorectal Cancer 2022;21:e162-70. [Crossref] [PubMed]
  15. Raimondi A, Nichetti F, Stahler A, et al. Optimal maintenance strategy following FOLFOX plus anti-EGFR induction therapy in patients with RAS wild type metastatic colorectal cancer: An individual patient data pooled analysis of randomised clinical trials. Eur J Cancer 2023;190:112945. [Crossref] [PubMed]
  16. Petrelli F, Barni S, Coinu A, et al. The Modified Glasgow Prognostic Score and Survival in Colorectal Cancer: A Pooled Analysis of the Literature. Rev Recent Clin Trials 2015;10:135-41. [Crossref] [PubMed]
  17. Mastrantoni L, Beccia V, Caira G, et al. Maintenance strategies after anti-EGFR-based induction in metastatic colorectal cancer: A systematic review and bayesian network meta-analysis. Crit Rev Oncol Hematol 2023;191:104106. [Crossref] [PubMed]
  18. Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012;486:532-6. [Crossref] [PubMed]
  19. Diaz LA Jr, Williams RT, Wu J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012;486:537-40. [Crossref] [PubMed]
  20. Siravegna G, Bardelli A. Failure is not final: ctDNA-guided rechallenge therapy in colorectal cancer. Ann Oncol 2019;30:1671. Erratum for Ann Oncol 2019;30:157-9. [Crossref] [PubMed]
  21. Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med 2015;21:795-801. [Crossref] [PubMed]
  22. Parseghian CM, Loree JM, Morris VK, et al. Anti-EGFR-resistant clones decay exponentially after progression: implications for anti-EGFR re-challenge. Ann Oncol 2019;30:243-9. [Crossref] [PubMed]
  23. Mauri G, Pizzutilo EG, Amatu A, et al. Retreatment with anti-EGFR monoclonal antibodies in metastatic colorectal cancer: Systematic review of different strategies. Cancer Treat Rev 2019;73:41-53. [Crossref] [PubMed]
  24. Parseghian C, Eluri M, Kopetz S, et al. Mechanisms of resistance to EGFR-targeted therapies in colorectal cancer: more than just genetics. Front Cell Dev Biol 2023;11:1176657. [Crossref] [PubMed]
doi: 10.21037/actr-24-30
Cite this article as: Pfeiffer P, Liposits G, Tarpgaard L. Maintenance strategies in first-line treatment for RAS/BRAF wild-type metastatic colorectal cancer—one size does not fit all. AME Clin Trials Rev 2024;2:61.

Download Citation