What are the optimal first-line therapies for patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer?
Editorial Commentary

What are the optimal first-line therapies for patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer?

Koichi Taira^, Akie Kimura, Akinobu Nakata, Fumio Tanaka, Yasuaki Nagami, Yasuhiro Fujiwara

Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan

^ORCID: 0000-0002-4932-508X.

Correspondence to: Koichi Taira. Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku Osaka City, 545-8585 Osaka, Japan. Email: koichit0802@gmail.com.

Comment on: Lenz HJ, Van Cutsem E, Luisa Limon M, et al. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J Clin Oncol 2022;40:161-70.


Keywords: Immunotherapy; colorectal cancer (CRC); first-line treatment


Submitted Dec 09, 2022. Accepted for publication Feb 06, 2023. Published online Feb 27, 2023.

doi: 10.21037/apm-22-1391


Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer death worldwide (1). Among people diagnosed with CRC, 20% have metastatic CRC (mCRC) (2). The prognosis of mCRC is poor with a 5-year survival rate of less than 15% (3).

However, the number of chemotherapies available to clinicians is gradually increasing. We can use standard chemotherapies as first-line treatment, including fluoropyrimidines, oxaliplatin, irinotecan, anti-vascular endothelial growth factor (VEGF) agents (bevacizumab), and anti-epidermal growth factor receptor (EGFR) agents (cetuximab and panitumumab), alone or in combination (4). Currently, the median overall survival (OS) in mCRC is about to reach approximately 30 months (5,6).

Microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR)/colorectal tumors are identified in approximately 15% of stage I–III CRC patients (7). They show a favorable prognosis compared with that of microsatellite stable/mismatch repair proficient (MSS/pMMR) CRC. In contrast, patients with dMMR/MSI-H mCRC in stage IV account for approximately 5% of all patients with CRC and have poor outcomes with a median OS of only 13.6–16.8 months to benefit from standard chemotherapy (8). Recently, immune-checkpoint inhibitors (ICIs) have been widely used for many cancers and shown to be effective (9-11). For mCRC, various clinical trials are underway, and ICIs have already shown a clinical benefit (12,13). ICIs consist of antagonists of programmed death-1 (PD-1), programmed death ligand-1 (PD-L1), and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) (2).

Recently, two pivotal trials (CheckMate-142 and KEYNOTE-177) have been conducted to investigate first-line therapy for patients with MSI-H/dMMR mCRC (14-19).

CheckMate-142 is a phase II, multicohort, non-randomized study of nivolumab (the fully human immunoglobulin G4 monoclonal antibody inhibitor of PD-1)-based therapies in patients with MSI-H (previously treated and untreated) and non-MSI-H mCRC (14,15). One study has reported on a previously treated cohort that underwent nivolumab monotherapy and nivolumab plus ipilimumab (the fully human immunoglobulin G1 monoclonal antibody that targets the CTLA-4 checkpoint receptor) therapy (14). The previously treated cohort showed a durable response and manageable safety profile. The primary endpoint was the investigator-assessed objective response rate (ORR). In the nivolumab monotherapy cohort of CheckMate-142, at a median follow-up of 12 months, the investigator-assessed ORR was 31.1% [95% confidence interval (CI), 20.8–42.9%], and the disease control rate for ≥12 weeks was 69% (95% CI, 57–79%). Median progression-free survival (PFS) was 14.3 months [95% CI, 4.30–not estimable (NE)], and the 12-month PFS was 50% (95% CI, 38–61%). The 12-month OS was 73% (95% CI, 62–82%). The rates of any-grade and grade 3 to 4 treatment-related adverse events (TRAEs) were 70% and 20%, respectively. In the nivolumab plus ipilimumab cohort, at a median follow-up of 13.4 months, the investigator-assessed ORR was 55% (95% CI, 45.2–63.8%), and the disease control rate for ≥12 weeks was 79% (95% CI, 70.6–85.9%). The 9- and 12-month PFS rates were 76% (95% CI, 67.0– 82.7%) and 71% (95% CI, 61.4–78.7%), respectively (15). The 9- and 12-month OS rates were 87% (95% CI, 80.0–92.2%) and 85% (95% CI, 77.0–90.2%), respectively. The rates of any-grade and grade 3 to 4 TRAEs were 73% and 32%, respectively. The any-grade TRAEs were comparable to those in the nivolumab monotherapy cohort. In general, nivolumab plus ipilimumab therapy was reported to result in more adverse events than nivolumab monotherapy. Nivolumab plus ipilimumab therapy has been investigated using different doses and intervals to solve these problems. The CheckMate-142 previously treated cohort underwent nivolumab therapy (3 mg/kg) plus low-dose (1 mg/kg) ipilimumab every 3 weeks. This low-dose ipilimumab led to decreased adverse events.

Recently, the results of nivolumab plus low-dose ipilimumab as first-line therapy for patients with MSI-H/dMMR mCRC have been reported in another CheckMate-142 cohort (17). This is the first report of dual immune-oncology (I-O) combination first-line therapy for MSI-H/dMMR mCRC. The primary endpoint was investigator-assessed ORR. After a median follow-up of 29 months, the investigator-assessed ORR, complete response (CR), and disease control rate were 69% (95% CI, 53–82%), 13%, and 84%, respectively. The ORR benefit was observed in all evaluated subgroups, including those with BRAF or KRAS mutation status. Median PFS and OS were not reached. The 24-month PFS and OS rates were 73.6% (95% CI, 57.2–84.5%) and 79.4% (95% CI, 64.1–88.7%), respectively, and the progressive disease (PD) rate was 13%. The rates of any-grade and grade 3 to 4 TRAEs were 80% and 22%, respectively. The rates of grade 3 to 4 TRAEs were comparable to those of the nivolumab monotherapy cohort in previously treated MSI-H/dMMR mCRC. This cohort used nivolumab (3 mg/kg) every 2 weeks plus low-dose ipilimumab (1 mg/kg) every 6 weeks. The less frequent and low-dose ipilimumab was manageable and resulted in to fewer serious TRAEs.

The two main limitations of this study are its nonrandomized design and its small sample size. However, the therapy investigated in this study showed robust and durable clinical benefits and was well tolerated. Based on these results, the National Comprehensive Cancer Network guidelines recommend nivolumab plus low-dose ipilimumab as a preferred regimen for first-line therapy in patients with MSI-H/dMMR mCRC.

There is also a report on first-line therapy using other ICIs for patients with MSI-H/dMMR mCRC. KEYNOTE-177 was a multicenter open-label phase III study of pembrolizumab, another PD-1 inhibitor, versus 5-fluorouracil-based chemotherapy alone or in combination with bevacizumab or cetuximab as first-line chemotherapy in MSI-H/dMMR mCRC (18,19). Patients receiving chemotherapy were able to cross over to pembrolizumab therapy after disease progression. The two primary endpoints were PFS and OS. The initial report showed that the median PFS was 16.5 months (95% CI, 5.4–32.4 months) in the pembrolizumab group and 8.2 months (95% CI, 6.1–10.2 months) in the chemotherapy group [hazard ratio (HR), 0.60; 95% CI, 0.45–0.80; P=0.0002), after a median follow-up of 32.4 months. The ORR was observed in 43.8% (95% CI, 35.8–52.0%) of the patients in the pembrolizumab group as compared with 33.1% (95% CI, 25.8–41.1%) in the chemotherapy group, with CR in 11% and 4%, respectively. TRAEs of grade 3 or higher occurred in 22% of the patients in the pembrolizumab group, as compared with 66% in the chemotherapy group. The final analysis showed that the median OS was not reached (NR) (95% CI, 49.2–NR) in the pembrolizumab group vs 36.7 months (27.6–NR) in the chemotherapy group (HR, 0.4; 95% CI, 0.53–1.03; P=0.036), after a median follow-up of 44.5 months (19). However, the pembrolizumab group did not meet the initially set one-sided α boundary of 0.025 required for significance. This lack of OS benefit may have been attributed to the use of subsequent therapy in the chemotherapy group. Most patients (60%) received pembrolizumab or other ICIs after progression with first-line chemotherapy. The PD rate was 29%, which was higher than that with nivolumab plus low-dose ipilimumab as first-line therapy in CheckMate-142 (13%), due to primary resistance, misdiagnosed mismatch repair proficient or microsatellite stable disease, and pseudoprogression. The limitations of this study include the lack of a central confirmation of MSI-H/dMMR status and the heterogeneity of treatment regimens in the chemotherapy group. Based on these results, pembrolizumab was approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) as first-line therapy in patients with MSI-H/dMMR mCRC.

In conclusion, the CheckMate-142 trial supports nivolumab plus low-dose ipilimumab as standard first-line therapy in patients with MSI-H/dMMR mCRC. The CheckMate-142 and KEYNOTE-177 have shown that ICI monotherapy and dual I-O combination treatment provided great effectiveness. However, these two trials cannot be compared because of their different sample characteristics. In CheckMate-142, the patients were mostly Caucasian and only 38% were stage 4. On the other hand, in KEYNOTE-177, the all patients were stage 4. Further trials should be conducted to clarify which therapies are best as first-line therapy for patients with MSI-H/dMMR mCRC: ICI monotherapy, dual I-O combination, or ICI plus chemotherapy with or without targeted therapy. Furthermore, a high rate of PD is observed, with 29% in ICI monotherapy and 13% in dual I-O combination treatment. Therefore, to develop a treatment with a lower PD rate and manageable safety, it is necessary to reduce misdiagnosed MSI-H/dMMR status to unify inspection methods and improve their accuracy. Moreover, it is crucial to identify the low responsiveness to ICIs in advance. Reportedly, low tumor mutational burdens, a phosphatase and tensin homolog (PTEN) mutation, and a high neutrophil-to-lymphocyte ratio might be associated with early disease progression (20,21). The confirmation of these findings necessitates the conduction of larger studies taking into consideration the treatment strategies. Additionally, the intervals between CT scans for the CheckMate-142 and KEYNOTE-177 trials were 6 and 9 weeks, respectively. However, the detection of early disease progression requires a shorter interval of about 4 weeks.

Ongoing clinical trials in MSI-H/dMMR mCRC are evaluating different treatment combinations. For example, randomized phase III studies COMMIT (NCT02997228) and CheckMate 8HW (NCT04008030) are currently ongoing. COMMIT evaluates the activity of the combination between standard chemotherapy and bevacizumab with or without PD-L1, and CheckMate 8HW evaluates nivolumab monotherapy, nivolumab plus ipilimumab, or investigator’s choice. The results are expected in the near future.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Annals of Palliative Medicine. The article did not undergo external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://apm.amegroups.com/article/view/10.21037/apm-22-1391/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. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Ganesh K, Stadler ZK, Cercek A, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol 2019;16:361-75. [Crossref] [PubMed]
  3. National Cancer Institute Surveillance Epidemiology and End Results Program. Cancer stat facts: colorectal cancer. [Cited November 23, 2022]. Available online: https://www.cancer.gov/statfacts/html/colorect.html
  4. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: rectal cancer. version 2 2022. [Cited November 23, 2022]. Available online: https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf
  5. Venook AP, Niedzwiecki D, Lenz HJ, et al. Effect of First-Line Chemotherapy Combined With Cetuximab or Bevacizumab on Overall Survival in Patients With KRAS Wild-Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2017;317:2392-401. [Crossref] [PubMed]
  6. Yamazaki K, Nagase M, Tamagawa H, et al. Randomized phase III study of bevacizumab plus FOLFIRI and bevacizumab plus mFOLFOX6 as first-line treatment for patients with metastatic colorectal cancer (WJOG4407G). Ann Oncol 2016;27:1539-46. [Crossref] [PubMed]
  7. Sinicrope FA, Sargent DJ. Molecular pathways: microsatellite instability in colorectal cancer: prognostic, predictive, and therapeutic implications. Clin Cancer Res 2012;18:1506-12. [Crossref] [PubMed]
  8. Venderbosch S, Nagtegaal ID, Maughan TS, et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014;20:5322-30. [Crossref] [PubMed]
  9. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  10. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med 2015;373:1627-39. [Crossref] [PubMed]
  11. Choueiri TK, Larkin J, Oya M, et al. Preliminary results for avelumab plus axitinib as first-line therapy in patients with advanced clear-cell renal-cell carcinoma (JAVELIN Renal 100): an open-label, dose-finding and dose-expansion, phase 1b trial. Lancet Oncol 2018;19:451-60. [Crossref] [PubMed]
  12. Hirano H, Takashima A, Hamaguchi T, et al. Current status and perspectives of immune checkpoint inhibitors for colorectal cancer. Jpn J Clin Oncol 2021;51:10-9. [Crossref] [PubMed]
  13. Zhou C, Cheng X, Tu S. Current status and future perspective of immune checkpoint inhibitors in colorectal cancer. Cancer Lett 2021; Epub ahead of print. [Crossref] [PubMed]
  14. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol 2017;18:1182-91. [Crossref] [PubMed]
  15. Overman MJ, Lonardi S, Wong KYM, et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J Clin Oncol 2018;36:773-9. [Crossref] [PubMed]
  16. André T, Lonardi S, Wong KYM, et al. Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: 4-year follow-up from CheckMate 142. Ann Oncol 2022;33:1052-60. [Crossref] [PubMed]
  17. Lenz HJ, Van Cutsem E, Luisa Limon M, et al. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J Clin Oncol 2022;40:161-70. [Crossref] [PubMed]
  18. André T, Shiu KK, Kim TW, et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N Engl J Med 2020;383:2207-18. [Crossref] [PubMed]
  19. Diaz LA Jr, Shiu KK, Kim TW, et al. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol 2022;23:659-70. [Crossref] [PubMed]
  20. Sui Q, Zhang X, Chen C, et al. Inflammation promotes resistance to immune checkpoint inhibitors in high microsatellite instability colorectal cancer. Nat Commun 2022;13:7316. [Crossref] [PubMed]
  21. Chida K, Kawazoe A, Kawazu M, et al. A Low Tumor Mutational Burden and PTEN Mutations Are Predictors of a Negative Response to PD-1 Blockade in MSI-H/dMMR Gastrointestinal Tumors. Clin Cancer Res 2021;27:3714-24. [Crossref] [PubMed]
Cite this article as: Taira K, Kimura A, Nakata A, Tanaka F, Nagami Y, Fujiwara Y. What are the optimal first-line therapies for patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer? Ann Palliat Med 2023;12(3):607-610. doi: 10.21037/apm-22-1391

Download Citation