Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1534 2022-08-05 09:41:57 |
2 format correct + 41 word(s) 1575 2022-08-08 03:24:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Damato, A.;  Rotolo, M.;  Caputo, F.;  Borghi, E.;  Iachetta, F.;  Pinto, C. Oncogene Driver Mutations and Therapeutic Implications in CRC. Encyclopedia. Available online: https://encyclopedia.pub/entry/25887 (accessed on 27 July 2024).
Damato A,  Rotolo M,  Caputo F,  Borghi E,  Iachetta F,  Pinto C. Oncogene Driver Mutations and Therapeutic Implications in CRC. Encyclopedia. Available at: https://encyclopedia.pub/entry/25887. Accessed July 27, 2024.
Damato, Angela, Martina Rotolo, Francesco Caputo, Eleonora Borghi, Francesco Iachetta, Carmine Pinto. "Oncogene Driver Mutations and Therapeutic Implications in CRC" Encyclopedia, https://encyclopedia.pub/entry/25887 (accessed July 27, 2024).
Damato, A.,  Rotolo, M.,  Caputo, F.,  Borghi, E.,  Iachetta, F., & Pinto, C. (2022, August 05). Oncogene Driver Mutations and Therapeutic Implications in CRC. In Encyclopedia. https://encyclopedia.pub/entry/25887
Damato, Angela, et al. "Oncogene Driver Mutations and Therapeutic Implications in CRC." Encyclopedia. Web. 05 August, 2022.
Oncogene Driver Mutations and Therapeutic Implications in CRC
Edit

Genetic alterations in advanced colorectal cancer (CRC) have shown a negative predictive and prognostic role in specific target therapies. The assessment of RAS and BRAF genes and mismatch repair (MMR)/microsatellite status should be evaluated as molecular panel upfront in all cases of CRC to drive patients’ selection toward biological approved treatments.

genetic mutations immune-biomarkers colorectal cancer

1. Introduction

The assessment of RAS and BRAF genes and mismatch repair (MMR)/microsatellite status should be evaluated as molecular panel upfront in all cases of advanced colorectal cancer (CRC) to drive patients’ selection toward biological approved treatments, although the standard cytotoxic drugs (fluoropyrimidines, oxaliplatin, and irinotecan), remain the backbone in most of the cases without specific targets [1]. However, in the early stages of CRC (I–III), the choice of adjuvant treatment is not based on molecular targets, but people know that in stage II CRC, microsatellite instability high (MSI-H)/MMR deficiency is associated with a lower recurrence rate than MSI-low/MMR proficient tumors ( hazard ratio (HR) 0.65; 95% confidence interval (CI) 0.59–0.71) [2].
In this regard, there is a clinical need to improve molecular targets, regardless of those having received strict histology-agnostic approval.
In the last few decades, one of the most studied mechanisms involved in cancer progression concerns the immune system and how it interacts with cancer cells. This research led to revolutionary approaches with the immune checkpoint inhibitors (ICIs) by blocking the immune checkpoint proteins or their ligands to reactivate the antitumor immune response [3][4].
Plenty of biomarkers are appearing on the scientific landscape in order to identify those patients who could really benefit from immunotherapy. Among these, certainly, the most validated in CRC is microsatellite status, which occurs as MSI in 5% of sporadic CRC, while the remaining 95% are classified as stable (MSS) or MSI-low, and the efficacy of immunotherapy is yet to be defined [3].
Further biomarkers involved in immune system regulation should be identified.
Considering the role of immune checkpoints in tumor development and immune escape, the use of anti-programmed cell death protein 1 (PD-1)/PD-Ligand 1(PD-L1) monoclonal antibodies, stimulating the immune response against the tumor, has routinely become part of clinical practice [4].
The prognostic and/or predictive role of PD-1/PD-L1 and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) expression was recognized in various types of cancer, such as melanoma, lung, head and neck, urothelial cancer, and others [4]. ICIs are used in these tumors in different settings, often according to different cut-off values of PD-L1 expression evaluated by immunohistochemistry (IHC) and established by different scores [5].
Most equivocal is the role of the tumor mutational burden (TMB) due to massive molecular heterogeneity resulting in dissimilar correlations between TMB levels and prognostic outcomes such as overall survival (OS) and progression-free survival (PFS) [6][7][8][9].
The attention over other immune checkpoints increased significantly in the last years, and new potential targets were identified, such as Lymphocyte-activation gene 3 (LAG3), which is highly expressed on tumor-infiltrating lymphocytes (TILs) [10]. In some tumors, such as ovarian, melanoma, non-small-cell lung cancer (NSCLC), and gastrointestinal cancers, PD-1 was usually co-expressed with LAG3 [11][12][13][14]. Simultaneous activation of the LAG3 and PD-1 pathways in TILs results in greater T-cell exhaustion, and restraining both pathways may improve T-cell activity and immune response [15]. Elevated expression of LAG3 has seen positive impacts on PFS in estrogen receptor-negative breast cancer [16]. Hald et al. [17] reported that intraepithelial-LAG3 and stromal-LAG3 were both associated with improved disease-specific survival (DSS) and OS in NSCLC. Additionally, in esophageal squamous cell carcinoma, higher LAG3 expression was positively correlated with a better OS and PFS, especially in the patients at early stage I–II [14].
In this scenario, recognizing unresponsive patients before starting the immunotherapy would help the potential development of personalized treatment and allow patients to avoid unnecessary ICIs and toxicities. Nevertheless, the prognostic significance of all these immune checkpoint molecules remains controversial.

2. Oncogene Driver Mutations and Therapeutic Implications in CRC

2.1. RAS Mutations

In CRC, the RAS gene is mutated in kirsten rat sarcoma viral oncogene homolog (KRAS) in up to 40% of cases, mostly in exon 2 codons 12 and 13, and in NRAS gene in approximately 3–5% of CRCs in exon 3 (codon 61) and exon 2 (codons 12 and 13) [18]. Identification of RAS status is mandatory in clinical practice due to its role as a negative predictive factor in anti- epidermal growth factor receptor (EGFR) antibody response (cetuximab, panitumumab) [19][20][21]. Compared to wild-type tumors, RAS mutant patients have a worse prognosis, both in the adjuvant and metastatic settings [22][23]. Regardless of RAS status, anti-VEGF (vascular endothelial growth factor)/VEGFR (vascular endothelial growth factor receptor) -2/PDGF (Placental-Derived Growth Factor) agents such as bevacizumab, ramucirumab, aflibercept, and regorafenib demonstrated efficacy in further lines of treatment [24][25][26][27].
A dynamic process has been identified regarding further KRAS mutations leaving the opportunity to develop new treatment strategies. For example, the KRAS G12C codon mutation, which represents 3% of all CRC cases, is now susceptible to inhibition by small molecule inhibitors that specifically and irreversibly block KRAS G12C by locking it in an inactive GDP-bound site [28][29]. Two basket phase I/II studies proved that sotorasib (CodeBreaK100) and adagrasib (KRYSTAL-1) are both active in patients with advanced solid tumors harboring a KRASG12C mutation, among which mCRC [30][31][32]. However, EGFR signaling mutations were identified as the dominant mechanism of CRC resistance to KRAS G12C inhibitors [33]. Indeed, the combination of adagrasib and cetuximab improved objective response rate (ORR) to 43% and disease control rate (DCR) to 100% [34], leading to a possible novel histology-tuned targeted treatment for mCRC, also being tested as sotorasib plus panitumumab [35]. A randomized phase III trial (KRYSTAL-10) [36] is ongoing and evaluates second-line agadrasib plus cetuximab versus standard chemotherapy.
Additionally, several other combinations of sotorasib or adagrasib with diverse compounds as well as newer anti-RAS strategies are ongoing.

2.2. BRAF Mutations

The second genetic mutation is represented by BRAF V600E, accounting for 8–10% of mCRC cases, and is nearly always mutually exclusive with KRAS, resulting in RAS-independent oncogenic signaling through the mitogen-activated protein kinase (MAPK) pathway (RAS-RAF-MEK-ERK) [37]. This alteration is associated with a worse prognosis and could predict a poorer response to anti-EGFR treatment [38]. Based on the breathtaking results achieved in other solid tumors such as melanoma, several trials explored the use of BRAF inhibitors as single agents or in combination with MEK inhibitors in the mCRC. However, the inhibition in the MAPK/ERK pathway with a BRAF inhibitor results in adaptive feedback reactivation of MAPK signaling mediated by rapid feedback EGFR reactivation [39][40]. In order to overcome primary resistance, anti-EGFR monoclonal antibodies were tested with BRAF V600E inhibitors, with modest activity for this combination [41][42]. Recently, the BEACON phase III trial compared the combination of anti-BRAF V600E (encorafenib), anti-MEK (binimetinib), and anti-EGFR (cetuximab) inhibitors as a triplet or doublet (encorafenib and cetuximab) scheme versus investigator’s choice (FOLFIRI or irinotecan and cetuximab) in pre-treated BRAF V600E mutant mCRC patients [43]. Long-term follow-up updates showed that both double and triple combinations achieved a median OS of 9.3 months, compared with 5.9 months in the control arm. The overall response rate (ORR) was 26.8% with the triplet, 19.5% with the doublet, and 1.8% in the control arm, with prolonged maintenance of the quality of life (QoL) and reducing the risk of QoL deterioration by more than 40% [43]. The phase II ANCHOR-CRC trial is testing the triplet regimen (encorafenib, binimetinib, and cetuximab) in the first-line setting [44], showing an ORR of 47.8% and DCR of 88%; median PFS was 5.8 months (95% CI, 4.6–6.4) and mOS was 17.2 months (95% CI, 14.1-NE).
Furthermore, approximately 20% of BRAFV600E mCRC have high-level MSI (MSI-H). Currently, an anti-PD-1 antibody, pembrolizumab, represents the standard of care for BRAFV600E mutant MSI-H mCRC [45]. In a phase I/II clinical trial, patients with treatment-refractory BRAFV600E MSS mCRC received encorafenib, cetuximab, and nivolumab, showing effective activity in terms of ORR (45%), DCR (95%), mPFS (7.3 months; 95% CI, 5.5-NA), and mOS of 11.4 months (95% CI, 7.6-NA) [46].
Although the best treatment has not yet been identified, an aggressive strategy involving triplet chemotherapy and targeted therapy is currently the standard of care for fit patients. BRAF- therapies combined with other anti-EGFRs, MEK inhibitors, or PI3K inhibitors seem promising [47][48].

2.3. ERBB2 Alterations

Further developments have identified another molecular biomarker in mCRC, as the prevalence of receptor tyrosine-protein kinase erbB-2 (ERBB2) or frequently called human epidermal growth factor receptor 2 (HER2) amplification, in approximately 3–8%, especially in KRAS wild-type CRC [49]. Although retrospective data have demonstrated that ERBB2 amplification represents a negative predictive biomarker for anti-EGFR therapies, prospective data are needed to define the relationship between ERBB2 and anti-EGFR responsiveness [50][51]. Indeed, RAS/BRAF wild-type mCRC should be screened for ERBB2 amplification before treatment with anti-EGFR therapies [52]. The activity of an anti- ERBB2-targeted therapy was demonstrated in the phase II HERACLES-A trial [53], in which a double signaling blockade by trastuzumab and lapatinib achieved an ORR of 30% in KRAS wild-type chemo refractory tumors. ERBB2 positivity was defined as tumors with a 3 + score in more than 50% of cells by immunohistochemistry or a 2 + score and a HER2:CEP17 ratio > 2 in more than 50% of cells by fluorescent in situ hybridization (FISH). Similar results were confirmed in the MyPathway phase II basket trial (trastuzumab plus pertuzumab) [54], and HER2 positivity was defined as amplification (FISH or HER2/CEP17 ratio 2.0 or copy number > 6), overexpression (IHC 3 +), or activating HER2 mutations. Emergent ERBB2 target agents are evolving in mCRC, such as the tyrosine kinase inhibitors, tucatinib, and an antibody-drug conjugate, trastuzumab-deruxtecan, that in the phase II DESTINY-CRC01 trial [55] achieved in the ERBB2 positive RAS wild-type mCRC an ORR of 45.3%. The preliminary results of the ongoing phase II MOUNTAINEER trial [56] showed 55% ORR with tucatinib combined with trastuzumab.
Remains to be clarified the heterogeneity in HER2/ERBB2 assessment in clinical trials and even across different tumor types [49].

References

  1. Goodwin, R.A.; Asmis, T.R. Overview of Systemic Therapy for Colorectal Cancer. Clin. Colon Rectal Surg. 2009, 22, 251–256.
  2. Popat, S.; Hubner, R.; Houlston, R.S. Systematic Review of Microsatellite Instability and Colorectal Cancer Prognosis. J. Clin. Oncol. 2005, 23, 609–618.
  3. Overman, M.J.; Ernstoff, M.S.; Morse, M.A. Where we stand with immunotherapy in colorectal cancer: Deficient mismatch repair, proficient mismatch repair, and toxicity management. Am. Soc. Clin. Oncol. Educ. Book 2018, 38, 239–247.
  4. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074.
  5. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018, 362, eaar3593.
  6. Addeo, A.; Friedlaender, A.; Banna, G.L.; Weiss, G.J. TMB or not TMB as a biomarker: That is the question. Crit. Rev. Oncol. Hematol. 2021, 163, 103374.
  7. Prasad, V.; Addeo, A. The FDA approval of pembrolizumab for patients with TMB >10 mut/Mb: Was it a wise decision? No. Ann. Oncol. 2020, 31, 1112–1114.
  8. Marcus, L.; Fashoyin-Aje, L.A.; Donoghue, M.; Mengdie Yuan, L.R. Pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin. Cancer Res. 2021, 27, 4685–4689.
  9. Wang, F.; Wei, X.L.; Wang, F.H.; Xu, N.; Shen, L.; Dai, G.H.; Yuan, X.L.; Chen, Y.; Yang, S.J.; Shi, J.H.; et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann. Oncol. 2019, 30, 1479–1486.
  10. Ruffo, E.; Wu, R.C.; Bruno, T.C.; Workman, C.J.; Vignalia, D.A.A. Lymphocyte-activation gene 3 (LAG3): The next immune check- point receptor. Semin. Immunol. 2019, 42, 101305.
  11. Zelba, H.; Bedke, J.; Hennenlotter, J.; Mostböck, S.; Zettl, M.; Zichner, T.; Chandran, A.; Stenzl, A.; Rammensee, H.-G.; Gouttefangeas, C. PD-1 and LAG-3 dominate checkpoint receptor-mediated T-cell inhibition in renal cell carcinoma. Cancer Immunol. Res. 2019, 7, 1891–1899.
  12. Whitehair, R.; Peres, L.C.; Mills, A.M. Expression of the immune checkpoints LAG-3 and PD-L1 in high-grade serous ovarian carcinoma: Relationship to tumor-associated lymphocytes and germline BRCA status. Int. J. Gynecol. Pathol. 2020, 39, 558–566.
  13. He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J. Thorac. Oncol. 2017, 12, 814–823.
  14. Zhang, Y.; Liu, Y.D.; Luo, Y.L.; Liu, B.; Huang, Q.; Wang, F.; Zhong, Q. Prognostic value of lymphocyte activation gene-3 (LAG-3) expression in esophageal squamous cell carcinoma. J. Cancer 2018, 9, 4287–4293.
  15. Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1- specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880.
  16. Burugu, S.; Gao, D.; Leung, S.; Chia, S.K.; Nielsen, T.O. LAG-3+ tumor infiltrating lymphocytes in breast cancer: Clinical correlates and association with PD-1/PD-L1+ tumors. Ann. Oncol. 2017, 28, 2977–2984.
  17. Hald, S.M.; Rakaee, M.; Martinez, I.; Richardsen, E.; Al-Saad, S.; Paulsen, E.; Blix, E.S.; Kilvaer, T.; Andersen, S.; Busund, L.T.; et al. LAG-3 in Non-Small-cell Lung Cancer: Expression in Primary Tumors and Metastatic Lymph Nodes Is Associated with Improved Survival. Clin. Lung Cancer 2018, 19, 249–259.
  18. Han, C.B.; Li, F.; Ma, J.T.; Zou, H.W. Concordant KRAS mutations in primary and metastatic colorectal cancer tissue specimens: A meta-analysis and systematic review. Cancer Investig. 2012, 30, 741–747.
  19. Douillard, J.Y.; Oliner, K.S.; Siena, S.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Panitumumab–FOLFOX4 Treatment and RAS Mutations in Colorectal Cancer. N. Engl. J. Med. 2013, 369, 1023–1034.
  20. Van Cutsem, E.; Lenz, H.J.; Köhne, C.H.; Heinemann, V.; Tejpar, S.; Melezínek, I.; Beier, F.; Stroh, C.; Rougier, P.; van Krieken, J.H.; et al. Fluorouracil, Leucovorin, and Irinotecan Plus Cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol. 2015, 33, 692–700.
  21. Bokemeyer, C.; Kohne, C.H.; Ciardiello, F.; Lenz, H.-J.; Heinemann, V.; Klinkhardt, U.; Beier, F.; Duecker, K.; van Krieken, J.H.; Tejpar, S. FOLFOX4 plus cetuximab treatment and RAS mutations in colorectal cancer. Eur. J. Cancer 2015, 51, 1243–1252.
  22. Yokota, T. Are KRAS/BRAF mutations potent prognostic and/or predictive biomarkers in colorectal cancers? Anticancer Agents Med. Chem. 2012, 12, 163–171.
  23. Schirripa, M.; Cremolini, C.; Loupakis, F.; Morvillo, M.; Bergamo, F.; Zoratto, F.; Salvatore, L.; Antoniotti, C.; Marmorino, F.; Sensi, E.; et al. Role of NRAS mutations as prognostic and predictive markers in metastatic colorectal cancer. Int. J. Cancer 2015, 136, 83–90.
  24. Loupakis, F.; Cremolini, C.; Masi, G.; Lonardi, S.; Zagonel, V.; Salvatore, L.; Cortesi, E.; Tomasello, G.; Ronzoni, M.; Spadi, R.; et al. Initial Therapy with FOLFOXIRI and Bevacizumab for Metastatic Colorectal Cancer. N. Engl. J. Med. 2014, 371, 1609–1618.
  25. Verdaguer, H.; Tabernero, J.; Macarulla, T. Ramucirumab in metastatic colorectal cancer: Evidence to date and place in therapy. Ther. Adv. Med. Oncol. 2016, 8, 230–242.
  26. Syed, Y.Y.; McKeage, K. Aflibercept: A Review in Metastatic Colorectal Cancer. Drugs 2015, 75, 1435–1445.
  27. Grothey, A.; Van Cutsem, E.; Sobrero, A.; Siena, S.; Falcone, A.; Ychou, M.; Humblet, Y.; Bouché, O.; Mineur, L.; Barone, C.; et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013, 381, 303–312.
  28. Henry, J.T.; Coker, O.; Chowdhury, S.; Shen, J.P.; Morris, V.K.; Dasari, A.; Raghav, K.; Nusrat, M.; Kee, B.; Parseghian, C.; et al. Comprehensive clinical and molecular characterization of KRAS G12C-mutant colorectal cancer. JCO Precis Oncol. 2021, 5, 613–621.
  29. Ostrem, J.M.; Peters, U.; Sos, M.L.; Wells, J.A.; Shokat, K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013, 503, 548–551.
  30. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRAS G12C Inhibition with Sotorasib in advanced solid tumors. N. Engl. J. Med. 2020, 383, 1207–1217.
  31. Fakih, M.G.; Kopetz, S.; Kuboki, Y.; Kim, T.W.; Munster, P.N.; Krauss, J.C.; Falchook, G.S.; Han, S.; Heinemann, V.; Muro, K.; et al. Sotorasib for previously treated colorectal cancers with KRASG12C Mutation (CodeBreaK100): A prespecified analysis of a single-arm, phase 2 trial. Lancet Oncol. 2022, 23, 115–124.
  32. Johnson, M.L.; Ou, S.H.I.; Barve, M.; Rybkin, I.I.; Papadopoulos, K.P.; Leal, T.A.; Velastegui, K.; Christensen, J.G.; Kheoh, T.; Chao, R.C.; et al. KRYSTAL-1: Activity and Safety of Adagrasib (MRTX849) in Patients with Colorectal Cancer (CRC) and Other Solid Tumors Harboring a KRAS G12C Mutation. Eur. J. Cancer 2020, 138, S2.
  33. Amodio, V.; Yaeger, R.; Arcella, P.; Cancelliere, C.; Lamba, S.; Lorenzato, A.; Arena, S.; Montone, M.; Mussolin, B.; Bian, Y.; et al. EGFR blockade reverts resistance to KRAS G12C inhibition in colorectal cancer. Cancer Discov. 2020, 10, 1129–1139.
  34. Weiss, J.; Yaeger, R.D.; Johnson, M.L.; Spira, A.; Klempner, S.J.; Barve, M.A.; Christensen, J.G.; Chi, A.; Der-Torossian, H.; Velastegui, K.; et al. LBA6 KRYSTAL-1: Adagrasib (MRTX849) as Monotherapy or Combined with Cetuximab (Cetux) in Patients (Pts) with Colorectal Cancer (CRC) Harboring a KRASG12C Mutation. Ann. Oncol. 2021, 32, S1294.
  35. Fakih, M.; Falchook, G.S.; Hong, D.S.; Yaeger, R.D.; Chan, E.; Mather, O.; Cardona, P.; Dai, T.; Strickler, J. CodeBreaK 101 Subprotocol H: Phase Ib Study Evaluating Combination of Sotorasib (Soto), a KRASG12C Inhibitor, and Panitumumab (PMab), an EGFR Inhibitor, in Advanced KRAS p.G12C-Mutated Colorectal Cancer (CRC). Ann. Oncol. 2021, 32, S551.
  36. Tabernero, J.; Bendell, J.; Corcoran, R.; Kopetz, S.; Lee, J.; Davis, M.; Christensen, J.; Chi, A.; Kheoh, T.; Yaeger, R.; et al. KRYSTAL-10: A randomized phase 3 study of adagrasib (MRTX849) in combination with cetuximab vs chemotherapy in patients with previously treated advanced colorectal cancer with KRASG12C mutation. Ann. Oncol. 2021, 32, S121.
  37. Pietrantonio, F.; Petrelli, F.; Coinu, A.; di Bartolomeo, M.; Borgonovo, K.; Maggi, C.; Cabiddu, M.; Iacovelli, R.; Bossi, I.; Lonat, V.; et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A meta-analysis. Eur. J. Cancer 2015, 51, 587–594.
  38. Rowland, A.; Dias, M.M.; Wiese, M.D.; Kichenadasse, G.; McKinnon, R.A.; Karapetis, C.S.; Sorich, M.J. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br. J. Cancer 2015, 112, 1888–1894.
  39. Corcoran, R.B.; Ebi, H.; Turke, A.B.; Coffee, E.M.; Nishino, M.; Cogdill, A.P.; Brown, R.D.; della Pelle, P.; Dias-Santagata, D.; Hung, K.E.; et al. EGFR-mediated reactivation of MAPK signaling contributes to insensitivity of BRAF-mutant colorectal cancers to RAF inhibition with vem.murafenib. Cancer Discov. 2012, 2, 227–235.
  40. Prahallad, A.; Sun, C.; Huang, S.; di Nicolantonio, F.; Salazar, R.; Zecchin, D.; Beijersbergen, R.L.; Bardelli, A.; Bernards, R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012, 483, 100–103.
  41. Atreya, C.E.; Van Cutsem, E.; Bendell, J.C.; Andre, T.; Schellens, J.H.; Gordon, M.S.; McRee, A.J.; O’Dwyer, P.J.; Muro, K.; Tabernero, J.; et al. Updated efficacy of the MEK inhibitor trametinib (T), BRAF inhibitor dab- rafenib (D), and anti-EGFR antibody panitumumab (P) in patients (pts) with BRAF V600E mutated (BRAFm) metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2015, 33, 103.
  42. Van Geel, R.M.J.M.; Tabernero, J.; Elez, E.; Bendell, J.C.; Spreafico, A.; Schuler, M.; Yoshino, T.; Delord, J.; Yamada, Y.; Lolkema, M.P.; et al. A phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov. 2017, 7, 610–619.
  43. Tabernero, J.; Grothey, A.; Van Cutsem, E.; Yaeger, R.; Wasan, H.; Yoshino, T.; Desai, J.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib plus Cetuximab as a new standard of care for previously treated BRAF V600E–mutant metastatic colorectal cancer: Updated survival results and subgroup analyses from the BEACON Study. J. Clin. Oncol. 2021, 39, 273–284.
  44. Van Cutsem, E.; Taieb, J.; Yaeger, R.; Yoshino, T.; Maiello, E.; Elez, E.; Dekervel, J.; Ross, P.; Ruiz Casado, A.; Graham, J.; et al. ANCHOR CRC: Results from a single-arm, phase 2 study of encorafenib, binimetinib plus cetuximab in previously untreated BRAF V600E mutant metastatic colorectal cancer. Ann. Oncol. 2021, 32, S222.
  45. André, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Carci-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in microsatellite instability-high advanced colorectal cancer. N. Engl. J. Med. 2020, 383, 2207–2218.
  46. Morris, V.K.; Perseghian, C.M.; Eschano, M.; Johnson, B.; Raghav, K.P.S.; Dasari, A.; Huey, R.; Overman, M.J.; Willis, J.; Lee, M.S.; et al. Phase I/II trial of encorafenib, cetuximab, and nivolumab in patients with microsatellite stable, BRAFV600E metastatic colorectal cancer. J. Clin. Oncol. 2022, 40, 12.
  47. Taieb, J.; Lapeyre-Prost, A.; Laurent Puig, P.; Zaanan, A. Exploring the best treatment options for BRAF-mutant metastatic colon cancer. Br. J. Cancer 2019, 121, 434–442.
  48. Mauri, G.; Bonazzina, E.; Amatu, A.; Tosi, F.; Bencardino, K.; Gori, V.; Massihnia, D.; Cipani, T.; Spina, F.; Ghezzi, S.; et al. The Evolutionary landscape of treatment for BRAFV600E mutant metastatic colorectal cancer. Cancers 2021, 13, 137.
  49. Valtorta, E.; Martino, C.; Sartore-Bianchi, A.; Penaullt-Llorca, F.; Viale, G.; Risio, M.; Rugge, M.; Grigioni, W.; Bencardino, K.; Lonardi, S.; et al. Assessment of a HER2 scoring system for colorectal cancer: Results from a validation study. Mod. Pathol. 2015, 28, 1481–1491.
  50. Bregni, G.; Sciallero, S.; Sobrero, A. HER2 amplification and anti-EGFR sensitivity in advanced colorectal cancer. JAMA Oncol. 2019, 5, 605.
  51. Pietrantonio, F.; Vernieri, C.; Siravegna, G.; Mennitto, A.; Berenato, R.; Perrone, F.; Gloghini, A.; Tamborini, E.; Lonardi, S.; Morano, F.; et al. Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. Clin. Cancer Res. 2017, 23, 2414–2422.
  52. Raghav, K.P.S.; Overman, M.J.; Yu, R.; Meric-Bernstam, F.; Menter, D.; Kee, B.K.; Muranyi, A.; Singh, S.; Routbort, M.; Chen, K.; et al. HER2 amplification as a negative predictive biomarker for anti-epidermal growth factor receptor antibody therapy in metastatic colorectal cancer. J. Clin. Oncol. 2016, 34, 3517.
  53. Sartore-Bianchi, A.; Trusolino, L.; Martino, C.; Bencardino, K.; Lonardi, S.; Bergamo, F.; Zagonel, V.; Leone, F.; Depetris, I.; Martinelli, E.; 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, 62. phase 2 trial. Lancet Oncol. 2016, 17, 738–746.
  54. Meric-Bernstam, F.; Hurwitz, H.; Raghav, K.P.S.; McWilliams, R.R.; Fakih, M.; VanderWalde, A.; Swanton, C.; Kurzrock, R.; Burris, H.; Sweeney, C.; et al. Pertuzumab plus trastuzumab for HER2-Amplified Metastatic Colorectal Cancer (MyPathway): An updated report from a multicentre, open-label, Phase 2a, Multiple Basket Study. Lancet Oncol. 2019, 20, 518–530.
  55. Siena, S.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Fakih, M.; Elez, E.; et al. Trastuzumab deruxtecan (DS-8201) in Patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-Label, Phase 2 Trial. Lancet Oncol. 2021, 22, 779–789.
  56. Strickler, J.H.; Ng, K.; Cercek, A.; Fountzilas, C.; Sanchez, F.A.; Hubbard, J.M.; Wu, C.; Siena, S.; Tabernero, J.; Van Cutsem, E.; et al. MOUNTAINEER: Open-Label, Phase II Study of Tucatinib Combined with Trastuzumab for HER2-Positive Metastatic Colorectal Cancer (SGNTUC-017, Trial in Progress). J. Clin. Oncol. 2021, 39, TPS153.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 454
Revisions: 2 times (View History)
Update Date: 08 Aug 2022
1000/1000
Video Production Service