U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Pratt VM, Scott SA, Pirmohamed M, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012-.

Cover of Medical Genetics Summaries

Medical Genetics Summaries [Internet].

Show details

Cetuximab Therapy and RAS and BRAF Genotype

, MD and , PhD.

Author Information and Affiliations

Created: .

Estimated reading time: 24 minutes

Introduction

Cetuximab (brand name Erbitux) is a monoclonal antibody used in the treatment of metastatic colorectal cancer (mCRC) and cancer of the head and neck. Cetuximab is an epidermal growth factor receptor (EGFR) antagonist, which works by blocking the growth of cancer cells. It is administered as a weekly intravenous (IV) infusion, but in practice, is often given every other week to coincide with chemotherapy (for example, FOLFIRI or FOLFOX). Cetuximab has several off-label uses as well, which include non-small cell lung cancer, squamous cell carcinoma of the skin, and Menetrier’s disease.

Interestingly, for colorectal cancer, the location of the primary tumor influences whether an individual with mCRC will respond to anti-EGFR therapy, and influences prognosis. Individuals with left-sided tumors are more likely to respond well to anti-EGFR therapy and have a better prognosis. Individuals with right-sided tumors have a worse prognosis and respond poorly to anti-EGFR therapy. However, currently only the mutation status of the tumor, and not the location of the tumor, is discussed in the drug label’s dosing recommendations.

Resistance to cetuximab is associated with specific RAS mutations. The RAS family of oncogenes includes the KRAS and NRAS genes. When mutated, these genes have the ability to transform normal cells into cancerous cells. The KRAS mutations are particularly common, being detectable in 40% of metastatic colorectal tumors.

The KRAS mutations often lead to constitutive activation of the mitogen-activated protein kinase (MAPK) pathway and are associated with resistance to anti-EGFR drugs such as cetuximab. In addition, mutations in NRAS and another gene, BRAF, have been associated with poor response to anti-EGFR therapy; however, BRAF mutation does not explicitly preclude anti-EGFR therapy. Combination therapies targeting both BRAF and EGFR have shown to improve survival for individuals with wild-type RAS and mutant BRAF.

The 2018 FDA-approved drug label for cetuximab states that for mCRC, cetuximab is indicated for K- and N-RAS wild-type (no mutation), EGFR-expressing tumors. The label states that an FDA-approved test must be used to confirm the absence of a RAS mutation (in either KRAS or NRAS) prior to starting cetuximab (Table 1) (1). While the FDA label also states that EGFR expression should also be confirmed by an approved test prior to starting therapy for mCRC, this is largely not implemented in practice, nor is it recommended by professional oncology society guidelines.

Similarly, the 2015 Update from the American Society of Clinical Oncology (ASCO) states that anti-EGFR therapy should only be considered for the treatment of individuals whose tumor is determined to not have mutations detected after extended RAS testing (Table 2) (2).

The 2020 National Comprehensive Cancer Network (NCCN) guideline also strongly recommends KRAS/NRAS genotyping of tumor tissue in all individuals with mCRC. In addition, the guideline states the V600E mutation in the BRAF gene makes a response to cetuximab (and panitumumab) highly unlikely unless given a BRAF inhibitor (Table 3) (3).

Table 1.

The FDA-Approved Cetuximab Label: Dosage and Administration (2020)

Genes to be testedRecommendations for metastatic colorectal cancer
NRAS Cetuximab is not indicated for the treatment of individuals with colorectal cancer that harbor somatic mutations in exon 2 (codons 12 and 13), exon 3 (codons 59 and 61), and exon 4 (codons 117 and 146) of either K-Ras or N- Ras and hereafter is referred to as “RAS” or when the Ras status is unknown.
Confirm the absence of a RAS mutation prior to initiation of treatment with cetuximab.
Information on FDA-approved tests for the detection of K-Ras mutations in individual with metastatic colorectal cancer is available here.
KRAS
EGFR Determine EGFR expression status using FDA-approved tests prior to initiating treatment with cetuximab.

This table is created from (1). EGFR, epidermal growth factor receptor

Table 2.

The ASCO RAS Mutational Testing of Colorectal Carcinoma Tissue (2015)

Genes to be testedRecommendation
KRAS RAS mutational testing of colorectal carcinoma tissue should be performed for all individuals who are being considered for anti-EGFR monoclonal antibody therapy (currently cetuximab and panitumumab).
Before treatment with anti-EGFR antibody therapy, individuals with mCRC should have their tumor tested for mutations in:
  • KRAS exons 2 (codons 12 and 13), 3 (codons 59 and 61) and 4 (codons 117 and 146)
  • NRAS exons 2 (codons 12 and 13), 3 (codons 59 and 61), and 4 (codons 117 and 146)
Anti-EGFR monoclonal antibody therapy should only be considered for treatment of individuals with mCRC carcinoma who are identified as having tumors with no mutations detected after such extended RAS mutation analysis.
NRAS

This table is adapted from (2). EGFR, epidermal growth factor receptor; mCRC, metastatic colorectal cancer; ASCO, American Society of Clinical Oncology

Table 3.

The NCCN KRAS, NRAS, and BRAF Mutation Testing in Metastatic Colorectal Cancer (2020)

Genes to be testedRecommendations for colorectal cancer
KRAS All individuals with metastatic colorectal cancer should have tumor tissue genotyped for RAS (KRAS and NRAS) and BRAF mutations.
individuals with any known KRAS mutation (exon 2, 3, 4) or NRAS mutation (exon 2, 3, 4) should not be treated with either cetuximab or panitumumab.
NRAS
BRAF BRAF V600E mutation makes response to cetuximab or panitumumab highly unlikely unless given with a BRAF inhibitor.

This table is created from (3). NCCN, National Comprehensive Cancer Network

Drug: Cetuximab

Cetuximab is an EGFR antagonist. It is used for the treatment of mCRC, and squamous cell carcinoma of the head and neck. Cetuximab, and the related drug panitumumab (brand name Vectibix, approved only for mCRC), are monoclonal antibodies that specifically target the extracellular domain of EGFR. They act by blocking endogenous ligand binding to the extracellular domain of EGFR, and by enhancing receptor internalization and degradation (4). Cetuximab has also been used for off-label indications that include non-small cell lung cancer (5), squamous cell carcinoma of the skin (6), and Menetrier’s disease (7).

Cetuximab is a chimeric monoclonal antibody composed of regions of both mouse and human antibody, whereas panitumumab is a fully human monoclonal antibody. Both biological agents have been shown to provide a clear clinical benefit in the treatment of RAS wild-type mCRC (8, 9).

Colorectal cancer is the third leading cause of cancer death for men and women in the US, and the second in Europe (10). Radiation therapy is generally used for early stage rectal cancer. Surgery is the most common treatment for localized colorectal cancer that has not spread, and chemotherapy is given before (neoadjuvant) or after (adjuvant) surgery to most individuals with cancer that has penetrated the bowel wall deeply or spread to the lymph nodes (11).

Treatment regimens for advanced or metastatic colorectal carcinoma include chemotherapy such as folinic acid, fluorouracil, irinotecan, capecitabine, and oxaliplatin. Targeted biological agents may be added to such regimens, such as cetuximab, panitumumab, and bevacizumab. Bevacizumab (brand name Avastin) is a monoclonal antibody that targets vascular endothelial growth factor, VEGF. Similar FDA-approved biologics include aflibercept (a VEGF inhibitor monoclonal antibody) and regorafenib (a receptor tyrosine kinase inhibitor with activity against VEGF receptors).

Cetuximab is used in combination with FOLFIRI (FOLinic acid, Fluorouracil, IRInotecan) or FOLFOX (FOLinic acid, Fluorouracil, Oxaliplatin) for first-line treatment; or in combination with irinotecan in individuals who are refractory to irinotecan-based chemotherapy (1, 12). Cetuximab may also be used as a single agent (monotherapy) in individuals who either did not respond to oxaliplatin- and irinotecan-based chemotherapy or are intolerant to irinotecan (3).

Interestingly, the location of the primary colorectal tumor is a predictor of the prognosis for metastatic disease. Left-sided tumors derive from the embryonic hindgut (which gives rise to the splenic flexure, descending colon, sigmoid colon, rectum, and one-third of the transverse colon). Whereas right-sided tumors derive from the embryonic midgut (which gives rise to the appendix, cecum, ascending colon, hepatic flexure, and two-thirds of the transverse colon) (13). These differences in embryologic origin correlate with common genetic alterations. Right-sided tumors are more likely to have mutated RAS and BRAF, while left-sided tumors may have upregulated EGFR and/or ERBB2 (HER2) (14). Thus, individuals with left-sided tumors benefit more from EGFR therapy than individuals with right-sided tumors (15, 16, 17).

Multiple professional guidelines suggest that cetuximab has limited benefit in first-line therapy for right-sided tumors (18, 19). Right-sided tumors may respond to bevacizumab in combination with chemotherapy, with potentially longer overall survival compared with cetuximab combination treatment (15, 16, 20). A recent review highlighted multiple retrospective studies regarding the prognostic and predictive power of right- versus left-sidedness of the primary tumor. The authors concluded that in first-line treatment, left-sided tumors were distinctly more likely to respond to anti-EGFR treatment. However, there was no clear consensus for the implications of tumor sidedness with respect to second-line (and beyond) treatment. (21)

Administration of IV anti-EGFR therapy may be associated with severe infusion reactions, including anaphylaxis (3% for cetuximab and 1% for panitumumab) and these reactions are more common in cetuximab treatment versus panitumumab (22). Other toxicities include cardiopulmonary arrest, severe skin rashes (the severity of which may predict an increased response and survival, regardless of RAS mutational status (23, 24)), and an increased risk of venous thrombosis and embolism (2, 11, 25). Additionally, a higher rate of cetuximab-induced infusion reactions has been reported in head and neck cancer treatment (5.4%) as compared with mCRC treatment (26). Within the United States, there appears to be a higher risk of anaphylaxis reaction in areas of the Southeast including North Carolina, Virginia, Tennessee, Florida as well as Missouri and Kansas (reviewed by (27)). Evidence suggests these infusion reactions are due to the presence of immunoglobulin E antibodies targeting a specific glycosylation moiety found on cetuximab (28). The presence of these antibodies in cetuximab-naïve individuals may be due to prior bites from the lone star tick (Amblyomma americanum) (29).

Cetuximab can cause fetal harm when administered during pregnancy. There are no studies in pregnant women, but in animal studies (cynomolgus monkeys) the administration of IV cetuximab in pregnancy resulted in an increased risk of fetal death. Women should be advised to use effective contraception during cetuximab therapy and for 2 months after the last dose. Women should also be advised of the potential risks to the fetus, and to inform their healthcare provider if they know or suspect they are pregnant.

An important role in the progression of mCRC is thought to involve the impaired regulation of EGFR function, resulting in activation of the associated MAPK pathway. Cetuximab and panitumumab are important agents in metastatic disease because they can block the activation of the MAPK pathway. However, resistance to these agents can arise through constitutive activation of the MAPK pathway, which is caused by mutations in downstream signaling proteins, such as KRAS, NRAS and BRAF. Approximately 40% of cases of mCRC are found to have activating mutations in KRAS.

The efficacy of cetuximab in treating mCRC is confined to individuals with wild-type KRAS tumors. Specifically, tumors that do not harbor specific mutations in exons 2, 3, and 4 of the KRAS gene. The NRAS gene is highly similar (homologous) to KRAS, and mutations in the same exons—2, 3, and 4—are also associated with a lack of response to cetuximab (17, 30, 31, 32, 33).

Therefore, expanded RAS testing (of KRAS and NRAS) is the standard of care to determine which individuals with mCRC may benefit from anti-EGFR therapy (34, 35).

Epidermal growth factor receptor overexpression is seen commonly in squamous cell head and neck cancers and is associated with poor survival outcomes (36, 37). Trials with cetuximab in local or metastatic head and neck cancers have shown this anti-EGFR therapy to have most benefit as part of combination therapies in metastatic cancer, with limited application in local, unresectable disease (reviewed by (18)). Neither the FDA-approved drug label nor NCCN guidelines recommend RAS genetic testing before initiating therapy with cetuximab in head and neck cancers (1, 18).

Proto-oncogenes

Proto-oncogenes are a group of genes that, when mutated or expressed at abnormally high levels, can contribute to normal cells becoming cancerous cells. The mutated version of the proto-oncogene is called an oncogene.

Proto-oncogenes typically encode proteins that stimulate cell division, inhibit cell differentiation, and halt cell death. All these are important biological processes; however, the increased production of these proteins, caused by oncogenes, can lead to the proliferation of poorly differentiated cancer cells (11). Members of the RAS family and the EGFR gene are all proto-oncogenes.

The RAS family contains three genes, HRAS, NRAS, and KRAS, and they are essential components of signaling pathways. They act as signal transducers –– coupling cell surface receptors to intracellular signaling pathways.

The RAS proteins regulate cell signal transduction by acting as a switch –– they cycle between "on" (GTP-bound) or "off" (GDP-bound) conformations. In the "on" position, RAS proteins transmit extracellular growth signals to the nucleus, primarily by the MAPK pathway. Cells are subsequently stimulated to grow, divide, mature, and differentiate.

Mutations in RAS genes leads to RAS proteins that are resistant to GTPase, so that GTP-remains permanently bound and the receptor remains "on" –– providing a continual growth stimulus to cells. Such activating RAS mutations are common in colorectal cancers.

The EGFR gene is a member of the human epidermal growth factor receptors family (HER) along with ERBB2 (HER2), ERBB3 (HER3) and ERBB4 (HER4). Overexpression of these genes has been associated with multiple cancer types. These receptors dimerize upon binding of an extracellular ligand and activate the downstream signaling pathways, including Ras/Raf/Mek/Erk proteins. In some contexts, overexpression of HER2 can promote dimerization without an extracellular signal, leading to constitutive activation. (38, 39)

Gene: KRAS

KRAS is the most frequently mutated RAS gene found in metastatic colorectal carcinoma. The most frequent individual mutations occur in KRAS exon 2, in codons 12 (G12D, G12V) and 13 (G13D). Collectively, these mutations account for 40% of all RAS mutations in mCRC (40). Individuals with mCRC that harbor KRAS mutations do not benefit from anti–EGFR therapy (either cetuximab or panitumumab therapy) (3, 41, 42, 43, 44, 45, 46).

Gene: NRAS

NRAS is highly homologous to KRAS, and mutations have been reported in exons 2, 3, and 4. Although NRAS mutations are not as frequent as KRAS in mCRC, occurring in approximately 2% of tumors, NRAS influences the response to treatment with anti-EGFR drugs (2, 47, 48, 49).

Individuals with NRAS-mutated tumors are less likely to respond to cetuximab or panitumumab (19, 25). Panitumumab may even have a detrimental effect in individuals with NRAS or KRAS mutations (2).

Gene: BRAF

RAF is a family of serine/threonine kinases that are downstream effectors of KRAS, within the MAPK signaling pathway. The RAF family has 3 members, ARAF, BRAF and CRAF (50).

BRAF mutations are detectable in approximately 5–15% of mCRCs. They tend to only occur in tumors that do not have KRAS exon 2 mutations (51). It is therefore unlikely that tumors with KRAS mutations will respond to either anti-BRAF treatment (which targets mutant BRAF) or anti-EGFR treatment (because of the presence of KRAS mutations) (52).

By far the most common BRAF mutation is known as V600E, which accounts for approximately 90% of BRAF mutations. The mutated BRAF protein is constitutively active and is a highly potent oncogene, acting downstream in the EGFR pathway, thus bypassing inhibition of EGFR by cetuximab or panitumumab (10). Constitutively active BRAF can then activate the downstream kinases MEK1 and MEK2, which ultimately activate ERK kinases at the terminus of the MAP kinase signaling pathway (53).

The BRAF V600E mutation is associated with a poorer prognosis for individuals with mCRC, as well as with resistance to anti-EGFR treatment. It is also possible that other BRAF mutations contribute to anti-EGFR resistance. In BRAF V600E-mutant mCRC, BRAF inhibition results in rapid feedback activation of EGFR, a likely mechanistic explanation for limited clinical utility of this monotherapy (54). Alternative treatments may include the use of drug combinations, such as the addition of a BRAF inhibitor to anti-EGFR, to overcome resistance (35, 55). Indeed, utilization of BRAF inhibitor therapy in combination with anti-EGFR (with or without additional targeting of MEK kinases) showed improved survival in the BEACON trial, with the greatest overall survival in the group targeting BRAF, EGFR and MEK simultaneously (54, 56). Guidelines from the NCCN recommend this triple therapy as one approach for BRAF V600E mutation-positive disease (3). The NCCN guidelines recommend additional combination therapies for BRAF V600E positive colorectal cancer of either vemurafenib, irinotecan and anti-EGFR monoclonal antibodies (cetuximab or panitumumab) or dabrafenib, trametinib and anti-EGFR monoclonal antibodies (3).

The NCCN Colon/Rectal Cancer Panel states that evidence increasingly suggests that the BRAF V600E variant makes response to panitumumab or cetuximab, as single agents or in combination with cytotoxic chemotherapy, highly unlikely unless it is also given with a BRAF inhibitor. Therefore, the panel recommends BRAF genotyping of tumor tissue (either primary tumor or metastasis) at diagnosis of stage IV disease (3).

Gene: EGFR

The HER family consists of 4 members: ERBB2 (HER2), ERBB3 (HER3) and ERBB4 (HER4). All 4 members are transmembrane tyrosine kinase receptors, and they regulate a number of important cellular processes, such as cell growth, survival, and differentiation.

The EGFR protein is expressed in many different tissues, and is activated by the binding of a ligand, such as epithelial growth factor (EGF) or transforming growth factor α (TGFα). Binding induces receptor dimerization, either homodimers or heterodimers with other HER family members, and triggers autophosphorylation of the intracellular tyrosine kinase domain.

By activating downstream signaling pathways, EGFR has many different biological roles, including stimulating the cell cycle, cell growth, division, differentiation, as well as increased cell invasiveness, apoptosis, and angiogenesis. Therefore, overexpression of EGFR is thought to be an important step in tumor progression, making EGFR a target for anticancer drugs (57, 58, 59).

Currently, there are 2 classes of drug that target EGFR: tyrosine kinase inhibitors (for example, gefitinib and erlotinib) and anti-EGFR monoclonal antibodies (for example, cetuximab and panitumumab) (4).

The EGFR protein is overexpressed in several cancers, including squamous cell carcinoma of the head and neck, squamous cell lung cancer, and colorectal cancer. The EGFR protein is overexpressed in approximately 50–80% of colorectal tumors (2, 60). The FDA-approved drug label for cetuximab states that the drug is licensed for EGFR-expressing mCRC, and mentions in animal studies using human tumor xenografts that lacked EGFR expression, that no anti-tumor effects of cetuximab were observed (1). However, for colorectal cancer, EGFR expression has not been associated with efficacy of anti-EGFR therapy (61).

The NCCN Colon/Rectal Cancer Panel states that EGFR testing of colorectal tumor cells has no proven predictive value in determining likelihood of response to either cetuximab or panitumumab. Therefore, the panel does not recommend routine EGFR testing, and states that no individual should be considered for, or excluded from, cetuximab or panitumumab therapy based on EGFR test results (3).

Gene: ERBB2 / HER2

HER2 belongs to the same family of signaling kinase receptors as EGFR and is encoded by the gene ERBB2, also called HER2. Monoclonal antibodies that target HER2, such as pertuzumab and trastuzumab, are used in the treatment of breast cancer. However, HER2 is rarely expressed in colorectal tumors (approximately 3% overall), though the prevalence is higher in RAS/BRAF wild-type tumors (5–14%) (3). Initial evidence suggested that HER2 overexpression may be predictive of resistance to anti-EGFR therapy, yet some evidence suggested that HER2 status is not a biomarker for cetuximab response (35, 62) A recent review of HER2 retrospective studies found a consistent correlation between ERBB2 amplification and resistance to anti-EGFR treatment (38).

The NCCN Colon/Rectal Cancer Panel recommends ERBB2 amplification/overexpression testing for individuals with mCRC. However, if the tumor is known to have a RAS or BRAF mutation, ERBB2 testing is not required. Based on the outcome of HER2 expression testing, the individual may be eligible for enrollment in one of the on-going clinical trials investigating targeted HER2 therapy in mCRC.(3) The NCCN guidelines emphasize that HER2 overexpression is not prognostic, but can be used to predict success of HER2-targeted therapy and resistance to anti-EGFR antibodies, including cetuximab (3). A dual tyrosine kinase inhibitor targeting HER2 and EGFR called lapatinib is also available and can be used in combination with anti-HER2 monoclonal antibodies for ERBB2-amplified mCRC (3).

Linking gene variation with treatment response

It has been established that specific variants in the genes KRAS and NRAS result in resistance to cetuximab therapy. In addition, the presence of the BRAF V600E mutation makes a beneficial response to treatment unlikely, unless given with a BRAF inhibitor (3, 56, 63). Specific point-mutation variants in ERBB2 and EGFR do not appear to be associated with cetuximab resistance. However, ERBB2 overexpression has been associated with decreased success of anti-EGFR therapies (38, 39).

Genetic Testing

The NIH Genetic Testing Registry, GTR, displays genetic tests that are available for the cetuximab drug response, and the genes KRAS, NRAS, EGFR, BRAF and ERBB2.

The 2020 NCCN Guideline for Colon Cancer (Version 4.2020) provides the following recommendations for genetic testing:

KRAS, NRAS, and BRAF Mutation and HER2 Testing

  • All [individuals] with metastatic colorectal cancer should have tumor tissue genotyped for RAS (KRAS and NRAS) and BRAF mutations. [Individuals] with any known KRAS mutation (exon 2, 3, 4) or NRAS mutation (exon 2, 3, 4) should not be treated with either cetuximab or panitumumab. BRAF V600E mutation makes response to panitumumab or cetuximab highly unlikely unless given with a BRAF inhibitor. WT RAS/BRAF tumors should also be screened for HER2 overexpression/amplification.
  • No specific methodology is recommended (e.g., sequencing, hybridization) for testing KRAS, NRAS, and BRAF mutations.
  • The testing can be performed on formalin-fixed paraffin-embedded tissue. The testing can be performed on the primary colorectal cancers and/or the metastasis, as literature has shown that the KRAS, NRAS, and BRAF mutations are similar in both specimen types.

Microsatellite Instability (MSI) or Mismatch Repair (MMR) Testing

  • Universal MMR* or MSI* testing is recommended in all newly diagnosed [individuals] with colon cancer. See NCCN Guidelines for Genetic/Familial High-Risk Assessment: Colorectal (*IHC for MMR and DNA analysis for MSI are different assays and measure different biological effects caused by deficient MMR function)
  • The presence of a BRAF V600E mutation in the setting of MLH1 absence would preclude the diagnosis of Lynch syndrome (LS) in the vast majority of cases. However, approximately 1% of cancers with BRAF V600E mutation (and loss of MLH-1) are LS. Caution should be exercised in excluding cases with a strong family history from germline screening in the case of BRAF V600E mutations
  • Stage II MSI-H [individuals] may have a good prognosis […]
  • Testing for MSI may be accomplished with a validated NGS panel, especially in [individuals] with metastatic disease who require genotyping of RAS and BRAF (3).

Therapeutic Recommendations based on Genotype

This section contains excerpted1 information on gene-based dosing recommendations. Neither this section nor other parts of this review contain the complete recommendations from the sources.

2020 Statement from the US Food and Drug Administration (FDA)

2.2 Recommended Dosage for Colorectal Cancer (CRC)

Determine EGFR-expression status using FDA-approved tests prior to initiating treatment. Also confirm the absence of a Ras mutation prior to initiation of treatment with cetuximab. Information on FDA-approved tests for the detection of K-Ras mutations in patients with metastatic CRC is available at: http://www.fda.gov/medicaldevices/productsandmedicalprocedures/invitrodiagnostics/ucm301431.htm.

[...]

5.7 Increased Tumor Progression, Increased Mortality, or Lack of Benefit in Patients with Ras- Mutant mCRC

Cetuximab is not indicated for the treatment of patients with CRC that harbor somatic mutations in exon 2 (codons 12 and 13), exon 3 (codons 59 and 61), and exon 4 (codons 117 and 146) of either K-Ras or N- Ras and hereafter is referred to as “Ras” or when the Ras status is unknown.

Retrospective subset analyses of Ras-mutant and wild-type populations across several randomized clinical trials, including CRYSTAL, were conducted to investigate the role of Ras mutations on the clinical effects of anti-EGFR-directed monoclonal antibodies. Use of cetuximab in patients with Ras mutations resulted in no clinical benefit with treatment related toxicity. Confirm Ras mutation status in tumor specimens prior to initiating cetuximab.

Please review the complete therapeutic recommendations that are located here: (1)

2020 Clinical Practice Guidelines in Oncology: Colon Cancer, from the National Comprehensive Cancer Network (NCCN)

Version 4.2020 – Discussion update in progress.

A sizable body of literature has shown that tumors with a mutation in codon 12 or 13 of exon 2 of the KRAS gene are essentially insensitive to cetuximab or panitumumab therapy. More recent evidence shows mutations in KRAS outside of exon 2 and mutations in NRAS are also predictive for a lack of benefit to cetuximab and panitumumab.

The panel therefore strongly recommends RAS (KRAS/NRAS) genotyping of tumor tissue (either primary tumor or metastasis) in all patients with metastatic colorectal cancer. Patients with known KRAS or NRAS mutations should not be treated with either cetuximab or panitumumab, either alone or in combination with other anticancer agents, because they have virtually no chance of benefit and the exposure to toxicity and expense cannot be justified. ASCO released a Provisional Clinical Opinion Update on extended RAS testing in patients with metastatic colorectal cancer (mCRC) that is consistent with the NCCN panel’s recommendations. A guideline on molecular biomarkers for CRC developed by the ASCP, CAP, AMP and ASCO also recommends resting consistent with the NCCN recommendations.

The recommendation for RAS testing, at this point, is not meant to indicate a preference regarding regimen selection in the first-line setting. Rather, this early establishment of RAS status is appropriate to plan for the treatment continuum, so that the information may be obtained in a non- time–sensitive manner and the patient and provider can discuss the implications of a RAS mutation, if present, while other treatment options still exist. Note that because anti-EGFR agents have no role in the management of stage I, II, or III disease, RAS genotyping of colorectal cancers at these earlier stages is not recommended.

KRAS mutations are early events in colorectal cancer formation, and therefore a very tight correlation exists between mutation status in the primary tumor and the metastases. For this reason, RAS genotyping can be performed on archived specimens of either the primary tumor or a metastasis. Fresh biopsies should not be obtained solely for the purpose of RAS genotyping unless an archived specimen from either the primary tumor or a metastasis is unavailable.

The panel recommends that KRAS, NRAS, and BRAF gene testing be performed only in laboratories that are certified under the Clinical Laboratory Improvement Amendments of 1988 (CLIA-88) as qualified to perform highly complex molecular pathology testing. No specific testing methodology is recommended. The three genes can be tested individually or as part of an NGS panel.

[…]

HER2 is a member of the same family of signalling kinase receptors as EGFR and has been successfully targeted in breast cancer in both the advanced and adjuvant settings. HER2 is rarely amplified/overexpressed in CRC (approximately 3% overall), but the prevalence is higher in RAF/BRAF-wild type tumors (reported at %5-14%). Specific molecular diagnostic methods have been proposed for HER2 testing in CRC and HER2-targeted therapies are now recommended as subsequent therapy options in patients with tumors that have HER2 overexpression. Based on this, the NCCN Guidelines recommend testing for HER2 amplifications for patients with mCRC. If the tumor is already known to have a KRAS/NRAS or BRAF mutations, HER2 testing is not required. As HER2-targeted therapies are still under investigation, enrollment in a clinical trial is encouraged.

Evidence does not support a prognostic role of HER2 overexpression. In addition to its role as a predictive marker for HER2-targeted therapy, initial results indicated HER2 amplification/overexpression may be predictive of resistance to EGFR-targeting monoclonal antibodies.

Please review the complete therapeutic recommendations that are located here: (3).

2015 Provisional Clinical Opinion from the American Society of Clinical Oncology (ASCO)
All patients with metastatic colorectal cancer who are candidates for anti-EGFR antibody therapy should have their tumor tested in a Clinical Laboratory Improvement Amendments–certified laboratory for mutations in both KRAS and NRAS exons 2 (codons 12 and 13), 3 (codons 59 and 61), and 4 (codons 117 and 146). The weight of current evidence indicates that anti-EGFR MoAb therapy should only be considered for treatment of patients whose tumor is determined to not have mutations detected after such extended RAS testing.

What’s New and Different?

In addition to testing for mutations in KRAS exon 2 (codons 12 and 13) as recommended previously, before treatment with anti- EGFR antibody therapy, patients with mCRC should have their tumor tested for mutations in:

  • KRAS exons 3 (codons 59 and 61) and 4 (codons 117 and 146)
  • NRAS exons 2 (codons 12 and 13), 3 (codons 59 and 61), and 4 (codons 117 and 146)

Please review the complete therapeutic recommendations that are located here: (2)

Allele Nomenclature

Selected KRAS Somatic Variants

Common allele nameAlternative namesHGVS reference sequencedbSNP reference identifier for allele location
CodingProtein
G12D p.Gly12Asp NM_004985.4:c.35G>A

NP_004976.2:p.Gly12Asp

rs121913529
G12V p.Gly12Val

NM_004985.4:c.35G>T

NP_004976.2:p.Gly12Val

rs121913529
G13D p.Gly13Asp

NM_033360.3:c.38G>A

NP_004976.2:p.Gly13Asp rs112445441

Selected NRAS Somatic Variants

Common allele nameAlternative namesHGVS reference sequencedbSNP reference identifier for allele location
CodingProtein
NRAS G12V p.Gly12Val NM_002524.4:c.35G>T NP_002515.1:p.Gly12Val rs121913237
NRAS G13R p.Gly13Arg NM_002524.4:c.37G>C NP_002515.1:p.Gly13Arg rs121434595
NRAS Q61R p.Gln61Arg NM_002524.4:c.182A>G NP_002515.1:p.Gln61Arg rs11554290
NRAS Q61K p.Gln61Lys NM_002524.4:c.181C>A NP_002515.1:p.Gln61Lys rs121913254

Selected BRAF Somatic Variants

Common allele nameAlternative namesHGVS reference sequencedbSNP reference identifier for allele location
CodingProtein
V600E p.Val600Glu NM_004333.4:c.1799T>C NP_004324.2:p.Val600Glu rs113488022

Guidelines for the description and nomenclature of gene variations are available from the Human Genome Variation Society (HGVS).

Acknowledgments

The authors would like to thank Carmen J. Allegra, MD, Professor of Medicine, University of Florida Health, Gainesville, FLA, USA; Thomas M. Delate, PhD, MS, Clinical Pharmacy Research Scientist, Drug Use Management, Kaiser Permanente National Pharmacy, Clinical Instructor, Clinical Pharmacy Department, Associate Member of University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Jared Freml, PharmD, Pharmacy Department, Kaiser Permanente Colorado, Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy & Pharmaceutical Sciences, Aurora, CO, USA; and Jesus Hermosillo-Rodriguez, MD, Oncology Department, Colorado Permanente Medical Group, Denver, CO, USA for reviewing this summary.

References

1.
ERBITUX- cetuximab solution [package insert]. Branchburg, NJ: ImClone LLC; 2020. Available from: https://dailymed​.nlm​.nih.gov/dailymed/drugInfo​.cfm?setid=8bc6397e-4bd8-4d37-a007-a327e4da34d9.
2.
Allegra C.J., Rumble R.B., Hamilton S.R., Mangu P.B., et al. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J Clin Oncol. 2016;34(2):179–85. [PubMed: 26438111]
3.
NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Colon Cancer: NCCN Guidelines. Version 4.2020. 15 June 2020 2020]; Available from: https://www​.nccn.org​/guidelines/category_1#colon.
4.
Hodoglugil U., Carrillo M.W., Hebert J.M., Karachaliou N., et al. PharmGKB summary: very important pharmacogene information for the epidermal growth factor receptor. Pharmacogenet Genomics. 2013;23(11):636–42. [PMC free article: PMC3966564] [PubMed: 23962910]
5.
Pirker R., Filipits M. Cetuximab in non-small-cell lung cancer. Transl Lung Cancer Res. 2012;1(1):54–60. [PMC free article: PMC4367590] [PubMed: 25806155]
6.
Montaudie H., Viotti J., Combemale P., Dutriaux C., et al. Cetuximab is efficient and safe in patients with advanced cutaneous squamous cell carcinoma: a retrospective, multicentre study. Oncotarget. 2020;11(4):378–385. [PMC free article: PMC6996917] [PubMed: 32064041]
7.
Fiske W.H., Tanksley J., Nam K.T., Goldenring J.R., et al. Efficacy of cetuximab in the treatment of Menetrier's disease. Sci Transl Med. 2009;1(8):8ra18. [PMC free article: PMC3638759] [PubMed: 20368185]
8.
Sorich M.J., Wiese M.D., Rowland A., Kichenadasse G., et al. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol. 2015;26(1):13–21. [PubMed: 25115304]
9.
Pietrantonio F., Cremolini C., Petrelli F., Di Bartolomeo M., et al. First-line anti-EGFR monoclonal antibodies in panRAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 2015;96(1):156–66. [PubMed: 26088456]
10.
Puerta-Garcia E., Canadas-Garre M., Calleja-Hernandez M.A. Molecular biomarkers in colorectal carcinoma. Pharmacogenomics. 2015;16(10):1189–222. [PubMed: 26237292]
11.
Weinstein I.B., Joe A.K. Mechanisms of disease: Oncogene addiction--a rationale for molecular targeting in cancer therapy. Nat Clin Pract Oncol. 2006;3(8):448–57. [PubMed: 16894390]
12.
Folprecht G., Gruenberger T., Bechstein W.O., Raab H.R., et al. Tumour response and secondary resectability of colorectal liver metastases following neoadjuvant chemotherapy with cetuximab: the CELIM randomised phase 2 trial. Lancet Oncol. 2010;11(1):38–47. [PubMed: 19942479]
13.
Tejpar S., Stintzing S., Ciardiello F., Tabernero J., et al. Prognostic and Predictive Relevance of Primary Tumor Location in Patients With RAS Wild-Type Metastatic Colorectal Cancer: Retrospective Analyses of the CRYSTAL and FIRE-3 Trials. JAMA Oncol. 2016 [PMC free article: PMC7505121] [PubMed: 27722750]
14.
Glebov O.K., Rodriguez L.M., Nakahara K., Jenkins J., et al. Distinguishing right from left colon by the pattern of gene expression. Cancer Epidemiol Biomarkers Prev. 2003;12(8):755–62. [PubMed: 12917207]
15.
Boeckx N., Koukakis R., Op de Beeck K., Rolfo C., et al. Effect of Primary Tumor Location on Second- or Later-line Treatment Outcomes in Patients With RAS Wild-type Metastatic Colorectal Cancer and All Treatment Lines in Patients With RAS Mutations in Four Randomized Panitumumab Studies. Clin Colorectal Cancer. 2018;17(3):170–178 e3. [PubMed: 29627309]
16.
Weinberg B.A., Hartley M.L., Salem M.E. Precision Medicine in Metastatic Colorectal Cancer: Relevant Carcinogenic Pathways and Targets-PART 1: Biologic Therapies Targeting the Epidermal Growth Factor Receptor and Vascular Endothelial Growth Factor. Oncology (Williston Park). 2017;31(7):539–48. [PubMed: 28712098]
17.
Aljehani M.A., Morgan J.W., Guthrie L.A., Jabo B., et al. Association of Primary Tumor Site With Mortality in Patients Receiving Bevacizumab and Cetuximab for Metastatic Colorectal Cancer. JAMA Surg. 2018;153(1):60–67. [PMC free article: PMC5833618] [PubMed: 28975237]
18.
NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Head & Neck Cancer: NCCN Guidelines. Version 2.2020. June 9, 2020 2020]; Available from: https://www​.nccn.org​/professionals/physician_gls​/pdf/head-and-neck.pdf.
19.
De Roock W., Claes B., Bernasconi D., De Schutter J., et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11(8):753–62. [PubMed: 20619739]
20.
Venook A.P., Niedzwiecki D., Innocenti F., Fruth B., et al. Impact of primary (1º) tumor location on overall survival (OS) and progression-free survival (PFS) in patients (pts) with metastatic colorectal cancer (mCRC): Analysis of CALGB/SWOG 80405 (Alliance). Journal of Clinical Oncology. 2016;34(15) suppl:3504–3504.
21.
Bahl A., Talwar V., Sirohi B., Mehta P., et al. Primary Tumor Location as a Prognostic and Predictive Marker in Metastatic Colorectal Cancer (mCRC). Front Oncol. 2020;10:964. [PMC free article: PMC7309590] [PubMed: 32612957]
22.
Bylsma L.C., Dean R., Lowe K., Sangare L., et al. The incidence of infusion reactions associated with monoclonal antibody drugs targeting the epidermal growth factor receptor in metastatic colorectal cancer patients: A systematic literature review and meta-analysis of patient and study characteristics. Cancer Med. 2019;8(12):5800–5809. [PMC free article: PMC6745824] [PubMed: 31376243]
23.
Jaka A., Gutierrez-Rivera A., Lopez-Pestana A., del Alcazar E., et al. Predictors of Tumor Response to Cetuximab and Panitumumab in 116 Patients and a Review of Approaches to Managing Skin Toxicity. Actas Dermosifiliogr. 2015;106(6):483–92. [PubMed: 25798804]
24.
Popa C.M., Lungulescu C., Ianosi S.L., Cherciu I., et al. Molecular Profiling of EGFR Status to Identify Skin Toxicity in Colorectal Cancer: A Clinicopathological Review. Curr Health Sci J. 2019;45(2):127–133. [PMC free article: PMC6778291] [PubMed: 31624638]
25.
De Mattos-Arruda L., Dienstmann R., Tabernero J. Development of molecular biomarkers in individualized treatment of colorectal cancer. Clin Colorectal Cancer. 2011;10(4):279–89. [PubMed: 21729679]
26.
Palomar Coloma V., Bravo P., Lezghed N., Mayache-Badis L., et al. High incidence of cetuximab-related infusion reactions in head and neck patients. ESMO Open. 2018;3(5):e000346. p. [PMC free article: PMC6069910] [PubMed: 30094066]
27.
Burke E., Rockey M., Grauer D., Henry D., et al. Assessment of cetuximab-induced infusion reactions and administration rechallenge at an academic medical center. Med Oncol. 2017;34(4):51. [PubMed: 28229341]
28.
Chung C.H., Mirakhur B., Chan E., Le Q.T., et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N Engl J Med. 2008;358(11):1109–17. [PMC free article: PMC2361129] [PubMed: 18337601]
29.
Commins S.P., James H.R., Kelly L.A., Pochan S.L., et al. The relevance of tick bites to the production of IgE antibodies to the mammalian oligosaccharide galactose-alpha-1,3-galactose. J Allergy Clin Immunol. 2011;127(5):1286–93 e6. [PMC free article: PMC3085643] [PubMed: 21453959]
30.
Sunakawa Y., Mogushi K., Lenz H.J., Zhang W., et al. Tumor sidedness and enriched gene groups for efficacy of first-line cetuximab treatment in metastatic colorectal cancer. Mol Cancer Ther. 2018 [PMC free article: PMC7497846] [PubMed: 30275242]
31.
Ghidini M., Petrelli F., Tomasello G. Right Versus Left Colon Cancer: Resectable and Metastatic Disease. Curr Treat Options Oncol. 2018;19(6):31. [PubMed: 29796712]
32.
Ottaiano A., De Stefano A., Capozzi M., Nappi A., et al. First Biologic Drug in the Treatment of RAS Wild-Type Metastatic Colorectal Cancer: Anti-EGFR or Bevacizumab? Results From a Meta-Analysis. Front Pharmacol. 2018;9:441. [PMC free article: PMC5943532] [PubMed: 29773991]
33.
Goldberg R.M., Montagut C., Wainberg Z.A., Ronga P., et al. Optimising the use of cetuximab in the continuum of care for patients with metastatic colorectal cancer. ESMO Open. 2018;3(4):e000353. p. [PMC free article: PMC5950648] [PubMed: 29765773]
34.
Bignucolo A., De Mattia E., Cecchin E., Roncato R., et al. Pharmacogenomics of Targeted Agents for Personalization of Colorectal Cancer Treatment. Int J Mol Sci. 2017;18(7) [PMC free article: PMC5536012] [PubMed: 28708103]
35.
Lin P.S., Semrad T.J. Molecular Testing for the Treatment of Advanced Colorectal Cancer: An Overview. Methods Mol Biol. 2018;1765:281–297. [PubMed: 29589315]
36.
Rubin Grandis J., Melhem M.F., Gooding W.E., Day R., et al. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 1998;90(11):824–32. [PubMed: 9625170]
37.
Zhu X., Zhang F., Zhang W., He J., et al. Prognostic role of epidermal growth factor receptor in head and neck cancer: a meta-analysis. J Surg Oncol. 2013;108(6):387–97. [PubMed: 24038070]
38.
De Cuyper A., Van Den Eynde M., Machiels J.P. HER2 as a Predictive Biomarker and Treatment Target in Colorectal Cancer. Clin Colorectal Cancer. 2020;19(2):65–72. [PubMed: 32229076]
39.
Afrasanie V.A., Marinca M.V., Alexa-Stratulat T., Gafton B., et al. KRAS, NRAS, BRAF, HER2 and microsatellite instability in metastatic colorectal cancer - practical implications for the clinician. Radiol Oncol. 2019;53(3):265–274. [PMC free article: PMC6765160] [PubMed: 31553708]
40.
Rowland A., Dias M.M., Wiese M.D., Kichenadasse G., et al. Meta-analysis comparing the efficacy of anti-EGFR monoclonal antibody therapy between KRAS G13D and other KRAS mutant metastatic colorectal cancer tumours. Eur J Cancer. 2016;55:122–30. [PubMed: 26812186]
41.
Amado R.G., Wolf M., Peeters M., Van Cutsem E., et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(10):1626–34. [PubMed: 18316791]
42.
Baselga J., Rosen N. Determinants of RASistance to anti-epidermal growth factor receptor agents. J Clin Oncol. 2008;26(10):1582–4. [PubMed: 18316790]
43.
Lievre A., Bachet J.B., Boige V., Cayre A., et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26(3):374–9. [PubMed: 18202412]
44.
Dahabreh I.J., Terasawa T., Castaldi P.J., Trikalinos T.A. Systematic review: Anti-epidermal growth factor receptor treatment effect modification by KRAS mutations in advanced colorectal cancer. Ann Intern Med. 2011;154(1):37–49. [PubMed: 21200037]
45.
Tejpar S., Celik I., Schlichting M., Sartorius U., et al. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30(29):3570–7. [PubMed: 22734028]
46.
Van Cutsem E., Lenz H.J., Kohne C.H. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 2015;33(7):692–700. V. Heinemann, et al. p. [PubMed: 25605843]
47.
Chang S.C., Lin P.C., Lin J.K., Lin C.H., et al. Mutation Spectra of Common Cancer-Associated Genes in Different Phenotypes of Colorectal Carcinoma Without Distant Metastasis. Ann Surg Oncol. 2016;23(3):849–55. [PubMed: 26471487]
48.
Vaughn C.P., Zobell S.D., Furtado L.V., Baker C.L., et al. Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosomes Cancer. 2011;50(5):307–12. [PubMed: 21305640]
49.
Janku F., Lee J.J., Tsimberidou A.M., Hong D.S., et al. PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One. 2011;6(7):e22769. p. [PMC free article: PMC3146490] [PubMed: 21829508]
50.
Orlandi A., Calegari A., Inno A., Berenato R., et al. BRAF in metastatic colorectal cancer: the future starts now. Pharmacogenomics. 2015;16(18):2069–81. [PubMed: 26615988]
51.
Rajagopalan H., Bardelli A., Lengauer C., Kinzler K.W., et al. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 2002;418(6901):934. [PubMed: 12198537]
52.
Morkel M., Riemer P., Blaker H., Sers C. Similar but different: distinct roles for KRAS and BRAF oncogenes in colorectal cancer development and therapy resistance. Oncotarget. 2015;6(25):20785–800. [PMC free article: PMC4673229] [PubMed: 26299805]
53.
Roviello G., D'Angelo A., Petrioli R., Roviello F., et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. Transl Oncol. 2020;13(9):100795. p. [PMC free article: PMC7260582] [PubMed: 32470910]
54.
Van Cutsem E., Huijberts S., Grothey A., Yaeger R., et al. Binimetinib, Encorafenib, and Cetuximab Triplet Therapy for Patients With BRAF V600E-Mutant Metastatic Colorectal Cancer: Safety Lead-In Results From the Phase III BEACON Colorectal Cancer Study. J Clin Oncol. 2019;37(17):1460–1469. [PMC free article: PMC7370699] [PubMed: 30892987]
55.
Shinozaki E., Yoshino T., Yamazaki K., Muro K., et al. Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the Biomarker Research for anti-EGFR monoclonal Antibodies by Comprehensive Cancer genomics (BREAC) study. Br J Cancer. 2017;117(10):1450–1458. [PMC free article: PMC5680457] [PubMed: 28972961]
56.
Kopetz S., Grothey A., Yaeger R., Van Cutsem E., et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N Engl J Med. 2019;381(17):1632–1643. [PubMed: 31566309]
57.
Raymond E., Faivre S., Armand J.P. Epidermal growth factor receptor tyrosine kinase as a target for anticancer therapy. Drugs. 2000;60 Suppl 1:15–23. [PubMed: 11129168]
58.
Krause D.S., Van Etten R.A. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2):172–87. [PubMed: 16014887]
59.
Normanno N., De Luca A., Bianco C., Strizzi L., et al. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366(1):2–16. [PubMed: 16377102]
60.
Antonacopoulou A.G., Tsamandas A.C., Petsas T., Liava A., et al. EGFR, HER-2 and COX-2 levels in colorectal cancer. Histopathology. 2008;53(6):698–706. [PubMed: 19102009]
61.
Cunningham D., Humblet Y., Siena S., Khayat D., et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351(4):337–45. [PubMed: 15269313]
62.
Valentini A.M., Cavalcanti E., Di Maggio M., Caruso M.L. RAS-expanded Mutations and HER2 Expression in Metastatic Colorectal Cancer: A New Step of Precision Medicine. Appl Immunohistochem Mol Morphol. 2018;26(8):539–544. [PMC free article: PMC6135466] [PubMed: 30199395]
63.
Kopetz S., McDonough S.L., Morris V.K., Lenz H.-J., et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG 1406). Journal of Clinical Oncology. 2017;35(4) suppl:520–520. [PMC free article: PMC8462593] [PubMed: 33356422]

Footnotes

1

The FDA labels specific drug formulations. We have substituted the generic names for any drug labels in this excerpt. The FDA may not have labeled all formulations containing the generic drug. Certain terms, genes and genetic variants may be corrected in accordance with nomenclature standards, where necessary. We have given the full name of abbreviations, shown in square brackets, where necessary.

Copyright Notice

All Medical Genetics Summaries content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 International (CC BY 4.0) license which permits copying, distribution, and adaptation of the work, provided the original work is properly cited and any changes from the original work are properly indicated. Any altered, transformed, or adapted form of the work may only be distributed under the same or similar license to this one.

Bookshelf ID: NBK564547PMID: 33237690

Views

Related Summaries by Drug Class

Tests in GTR by Condition

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...