Skip to main content
Case Reports in Oncology logoLink to Case Reports in Oncology
. 2015 Feb 11;8(1):83–87. doi: 10.1159/000375483

Clinical Response to Sorafenib in a Patient with Metastatic Colorectal Cancer and FLT3 Amplification

Raphael Brandão Moreira 1,*, Renata D'Alpino Peixoto 1, Marcelo Rocha de Sousa Cruz 1
PMCID: PMC4361904  PMID: 25848357

Abstract

Background

A considerable number of patients with metastatic colorectal cancer progress after exhausting all approved standard therapies but maintain an adequate performance status and could be candidates for further treatment. We aim at reviewing our experience with sorafenib treatment of a patient with FLT3 mutation in refractory metastatic colorectal cancer.

Methods

Treatment with sorafenib of a patient with metastatic colorectal cancer and FLT3 translocation who had previously been heavily treated.

Results

The patient with metastatic colorectal cancer, aged 51 years, showed significant symptomatic and laboratory improvement with sorafenib treatment (400 mg twice daily).

Conclusion

The presented case illustrates how an aggressive and refractory colorectal tumor may respond well to targeted therapy.

Key Words: Metastatic colorectal cancer, Sorafenib, FLT3 amplification

Introduction

Colorectal cancer (CRC) is the third most common type of cancer worldwide and the second leading cause of cancer-related mortality in humans [1]. At the time of the diagnosis, approximately 30% of the patients have distant metastases, which predominantly occur in the liver [1]. Surgical removal of metastatic deposits remains the only curative approach [2, 3]. However, most patients with advanced CRC will eventually die of metastatic disease, highlighting the desperate need for new and better therapies [1].

Progress in genome sequencing technology has facilitated the identification of crucial genetic alterations that control cancer cell proliferation by constitutive activation of cell signaling/cell cycling pathways or by inactivation of critical negative regulators of these networks [4, 5, 6, 7, 8]. More recently, a number of antibodies and small molecule inhibitors have been developed that target particular oncogenic drivers [9, 10, 11, 12, 13]. These targeted agents may not work extraordinarily well in an unselected population but may induce dramatic regression in tumors harboring the target, demonstrating the concept of ‘precision’ medicine.

Targeted next-generation sequencing (NGS) sequences the entire coding region of a large number of preselected genes with clinical or preclinical relevance in cancer [14]. It also provides a comprehensive analysis of genes with potential therapeutic and prognostic importance, a quick turnaround time (2–3 weeks in this case), and a standardized analytics pipeline [15]. Some patients with identified potentially actionable mutations may derive clinical benefit by receiving genotype-directed therapy.

We report the case of a patient with K-RAS mutated metastatic CRC who had already progressed on all standard therapies and wanted to pursue further options. A clinical comprehensive targeted genomic profile (FoundationOne) with the goal of finding potential drug-targetable genomic alterations was requested. FLT3 amplification was identified and the patient was started on sorafenib, achieving a great clinical benefit.

Case Report

A 51-year-old, previously healthy man presented in March 2011 with bowel habit alteration. He was investigated with a colonoscopy, which revealed a stenotic adenocarcinoma of the upper rectum. Imaging staging showed metastatic lesions in the liver and lungs. However, given the obstructive symptoms, he underwent rectosigmoidectomy. Pathology was consistent with a pT3pN2 (18 out of 19 lymph nodes positive) rectal adenocarcinoma. Molecular studies revealed the presence of KRAS G12D in the neoplastic tissue. A few weeks later, the patient was started on FOLFOX. Evaluation after 4 cycles showed a partial response on the lung and liver, and at that time he underwent resection of the liver and lung lesions. He was then restarted on FOLFOX and completed a total of 11 cycles. Unfortunately, after having stopped treatment with FOLFOX following 5 months, he was found to have new pulmonary and liver lesions, local recurrence, solitary CNS metastases and multiple lymph node metastases. In order to provide better local control, he received short-course radiotherapy (5 × 500 cGy) to the pelvic area followed by surgery. A few weeks later, he underwent neurosurgery with resection of the posterior fossa lesion followed by stereotactic radiosurgery to the surgical bed. Soon thereafter, he was started on FOLFIRI and bevacizumab, achieving stable disease for 6 months with progression in multiple sites 6 months later. At that time, the patient was cachectic, with an Eastern Cooperative Oncology Group performance status of 3 and with signs of liver insufficiency with hepatic encephalopathy. Blood tests revealed a total bilirubin level of 11 mg/dl. He was then admitted to the hospital for supportive treatment, and given the lack of standard therapies and the rapid progression of the disease, hospice care was discussed. Nevertheless, we decided to order a clinical comprehensive targeted genomic profile (FoundationOne) of his tumor with the goal of finding potential drug-targetable genomic alterations. Paraffin-embedded tumor tissue was evaluated using NGS (Foundation Medicine, Cambridge, Mass., USA). Hybridization capture of 3,769 exons from 236 cancer-related genes and of 47 introns from 19 genes commonly rearranged in cancer was applied to ≥50 ng of DNA extracted from the tumor specimens and sequenced to high, uniform coverage. FoundationOne identified 6 genomic alterations (KRAS G12D, FLT3 amplification, BRCA1 Q1756fs*74, TP53 splice site 920-1G>T, APC E1379* and CDK8 amplification). Either sunitinib or sorafenib (both FDA-approved drugs) were suggested as therapies associated with potential clinical benefit based on the presence of FLT3 amplification. In November 2013, the patient was started on sorafenib (400 mg twice daily). Within 7 days of therapy, the patient experienced a fast improvement of the encephalopathy and his performance status. The total bilirubin levels went down from 11 mg/dl to normal levels after 30 days. Four months later, the patients presented with disease progression and died of liver insufficiency secondary the metastatic rectal cancer.

Discussion

The identification of relevant molecular targets for cancer initiation and/or progression is an important focus for the development of targeted therapies for CRC, and significant advances have recently been made as a result of projects such as the National Cancer Institute's Cancer Genome Atlas Network [16]. Patients whose tumors depend on particular targets can then be selected to avoid or minimize primary resistance to therapy.

Several critical genes and pathways important in the initiation and progression of CRC have already been identified, such as WNT, RAS/MAPK, PI3K, TGF-β, P53 and DNA mismatch repair pathways [17]. Large-scale sequencing analyses have found numerous recurrently mutated genes and a recurrent chromosomal translocation [18, 19, 20]. In order to better characterize somatic alterations in CRC, the Cancer Genome Atlas Network conducted a genome scale analysis of 276 samples, analyzing exome sequences, DNA copy number, promoter methylation and messenger RNA and microRNA expression [16]. Twenty-four genes were significantly mutated, and in addition to the expected APC, TP53, SMAD4, PIK3CA and KRAS mutations, frequent mutations in ARID1A, SOX9 and FAM123B were discovered [16]. When we requested a targeted genomic profile for the presented case, FLT3 amplification was identified and sorafenib was suggested as a potential targeted therapy.

FLT3 encodes a receptor tyrosine kinase. FLT3 mutations are the most common somatic mutations observed in acute myeloid leukemia, and their presence may be a prognostic factor for poor outcome. Activating mutations of the FLT3 receptor gene lead to the constitutive activation of the FLT3 receptor tyrosine kinase and cause autonomous, cytokine-independent proliferation in vitro [21]. Signaling through the FLT3 pathway leads to the phosphorylation of Shc1 and Akt1 and the activation of mTOR, as well as RAS activation and phosphorylation of ERK1 and 2 [22, 23, 24]. FLT3 amplification has been reported at low levels in several tumor types, including colon and rectum adenocarcinoma (4.2%, 21/489) [25]. However, the frequency of FLT3 gene amplification in CRC and its biological significance remain unknown. In cancer cells with activating mutations in FLT3, Flt3 inhibitors or tyrosine kinase inhibitors have been shown to be effective [26, 27]. Several Flt3 inhibitors, including sunitinib and sorafenib, have been approved by the FDA for use in other indications. These and other inhibitors are under clinical investigation in several cancer types. However, it remains unknown if the rationale for the use of these Flt3 inhibitors applies to tumors with amplifications in FLT3.

Sorafenib has shown preclinical activity against a variety of tumor types [28, 29, 30, 31, 32] and is a standard treatment for advanced hepatocellular and renal cell carcinomas [33, 34]. It is an orally available multikinase inhibitor that targets Raf serine/threonine kinases (Raf-1, wild-type B-Raf and B-Raf V600E), vascular endothelial growth factor receptor (VEGFR)−1, −2 and −3, platelet-derived growth factor receptor (PDGFR)-β and FLT3, c-Kit and p38 tyrosine kinases. Sorafenib has a dual action that targets serine/threonine and receptor tyrosine kinases, inhibiting not only the Raf cascade, preventing the downstream mediation of cell growth and proliferation, but also the VEGFR-2, −3/PDGFR-β signaling cascade, inhibiting the activation of angiogenesis. Therefore, sorafenib acts by inhibiting tumor growth and disrupting tumor microvasculature through antiproliferative, antiangiogenic and proapoptotic effects [35, 36, 37, 38]. Amplification of FLT3 may predict sensitivity to sorafenib, although it is unclear if high-level amplification of FLT3 results in activation. Sorafenib has been shown to inhibit activated Flt3 in preclinical studies of acute myeloid leukemia [27].

Sorafenib has already been tested in a phase IIb trial with an unselected population with metastatic CRC. Patients were randomized to sorafenib (400 mg twice daily) or placebo, combined with mFOLFOX6 (oxaliplatin 85 mg/m2; levoleucovorin 200 mg/m2; fluorouracil 400 mg/m2 bolus and 2,400 mg/m2 continuous infusion) every 14 days. Median progression-free survival for sorafenib plus mFOLFOX6 was 9.1 months versus 8.7 months for placebo plus mFOLFOX6 (p = 0.46). There was also no difference between treatment arms for overall survival. Subgroup analyses of progression-free survival and overall survival were performed, but showed no difference between treatment arms considering KRAS or BRAF status (mutant and wild type). However, a benefit for sorafenib cannot be ruled out in more specific patient populations, such as the presented case. In addition, a phase Ib study reported a modest response rate for the combination of sorafenib, cetuximab and irinotecan in advanced CRC [39].

The presented case illustrates how an aggressive and refractory colorectal tumor may respond well to targeted therapy. We showed that the use of targeted NGS in clinical settings is practical and can benefit patients because of the ability to define tumors genetically. Despite notable success, effective genetically targeted therapies currently remain unavailable for most patients. Moreover, some patients with potentially actionable alterations do not respond to genotype-directed therapy, highlighting the still underdeveloped understanding of the pathophysiologic implications of many genetic alterations. Systematic evaluation of the predictive value of these genomic alterations is critically important for further progress in this field.

References

  • 1.Saltz LB, Meropol NJ, Loehrer PJ, Sr, Needle MN, Kopit J, Mayer RJ. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol. 2004;22:1201–1208. doi: 10.1200/JCO.2004.10.182. [DOI] [PubMed] [Google Scholar]
  • 2.Reissfelder C, Timke C, Schmitz-Winnenthal H, Rahbari NN, Koch M, Klug F, et al. A randomized controlled trial to investigate the influence of low dose radiotherapy on immune stimulatory effects in liver metastases of colorectal cancer. BMC Cancer. 2011;11:419. doi: 10.1186/1471-2407-11-419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Reissfelder C, Rahbari NN, Koch M, Ulrich A, Pfeilschifter I, Waltert A, et al. Validation of prognostic scoring systems for patients undergoing resection of colorectal cancer liver metastases. Ann Surg Oncol. 2009;16:3279–3288. doi: 10.1245/s10434-009-0654-7. [DOI] [PubMed] [Google Scholar]
  • 4.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
  • 5.Bos JL. ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682–4689. [PubMed] [Google Scholar]
  • 6.Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–566. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
  • 7.Nakahara M, Isozaki K, Hirota S, Miyagawa J, Hase-Sawada N, Taniguchi M, et al. A novel gain-of-function mutation of c-kit gene in gastrointestinal stromal tumors. Gastroenterology. 1998;115:1090–1095. doi: 10.1016/s0016-5085(98)70079-4. [DOI] [PubMed] [Google Scholar]
  • 8.Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009;9:749–758. doi: 10.1038/nrc2723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037. doi: 10.1056/NEJM200104053441401. [DOI] [PubMed] [Google Scholar]
  • 11.Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385–2394. doi: 10.1056/NEJMoa1214886. [DOI] [PubMed] [Google Scholar]
  • 12.Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12:735–742. doi: 10.1016/S1470-2045(11)70184-X. [DOI] [PubMed] [Google Scholar]
  • 13.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  • 14.Wagle N, Berger MF, Davis MJ, Blumenstiel B, Defelice M, Pochanard P, et al. High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer Discov. 2012;2:82–93. doi: 10.1158/2159-8290.CD-11-0184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031. doi: 10.1038/nbt.2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi: 10.1038/nature11252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fearon ER. Molecular genetics of colorectal cancer. Annu Rev Pathol. 2011;6:479–507. doi: 10.1146/annurev-pathol-011110-130235. [DOI] [PubMed] [Google Scholar]
  • 18.Bass AJ, Lawrence MS, Brace LE, Ramos AH, Drier Y, Cibulskis K, et al. Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat Genet. 2011;43:964–968. doi: 10.1038/ng.936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314:268–274. doi: 10.1126/science.1133427. [DOI] [PubMed] [Google Scholar]
  • 20.Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108–1113. doi: 10.1126/science.1145720. [DOI] [PubMed] [Google Scholar]
  • 21.Tse KF, Novelli E, Civin CI, Bohmer FD, Small D. Inhibition of FLT3-mediated transformation by use of a tyrosine kinase inhibitor. Leukemia. 2001;15:1001–1010. doi: 10.1038/sj.leu.2402199. [DOI] [PubMed] [Google Scholar]
  • 22.Zhang S, Mantel C, Broxmeyer HE. Flt3 signaling involves tyrosyl-phosphorylation of SHP-2 and SHIP and their association with Grb2 and Shc in Baf3/Flt3 cells. J Leukoc Biol. 1999;65:372–380. doi: 10.1002/jlb.65.3.372. [DOI] [PubMed] [Google Scholar]
  • 23.Mizuki M, Fenski R, Halfter H, Matsumura I, Schmidt R, Muller C, et al. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood. 2000;96:3907–3914. [PubMed] [Google Scholar]
  • 24.Chen W, Drakos E, Grammatikakis I, Schlette EJ, Li J, Leventaki V, et al. mTOR signaling is activated by FLT3 kinase and promotes survival of FLT3-mutated acute myeloid leukemia cells. Mol Cancer. 2010;9:292. doi: 10.1186/1476-4598-9-292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.The cBio Cancer Genomics Portal. 2013.
  • 26.Schittenhelm MM, Yee KW, Tyner JW, McGreevey L, Haley AD, Town A, et al. FLT3 K663Q is a novel AML-associated oncogenic kinase: determination of biochemical properties and sensitivity to Sunitinib (SU11248) Leukemia. 2006;20:2008–2014. doi: 10.1038/sj.leu.2404374. [DOI] [PubMed] [Google Scholar]
  • 27.von Bubnoff N, Engh RA, Aberg E, Sanger J, Peschel C, Duyster J. FMS-like tyrosine kinase 3-internal tandem duplication tyrosine kinase inhibitors display a nonoverlapping profile of resistance mutations in vitro. Cancer Res. 2009;69:3032–3041. doi: 10.1158/0008-5472.CAN-08-2923. [DOI] [PubMed] [Google Scholar]
  • 28.Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–134. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 29.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
  • 30.Suen AW, Galoforo S, Marples B, McGonagle M, Downing L, Martinez AA, et al. Sorafenib and radiation: a promising combination in colorectal cancer. Int J Radiat Oncol Biol Phys. 2010;78:213–220. doi: 10.1016/j.ijrobp.2010.02.064. [DOI] [PubMed] [Google Scholar]
  • 31.Ratain MJ, Eisen T, Stadler WM, Flaherty KT, Kaye SB, Rosner GL, et al. Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol. 2006;24:2505–2512. doi: 10.1200/JCO.2005.03.6723. [DOI] [PubMed] [Google Scholar]
  • 32.Galal KM, Khaled Z, Mourad AM. Role of cetuximab and sorafenib in treatment of metastatic colorectal cancer. Indian J Cancer. 2011;48:47–54. doi: 10.4103/0019-509X.75825. [DOI] [PubMed] [Google Scholar]
  • 33.Lang L. FDA approves sorafenib for patients with inoperable liver cancer. Gastroenterology. 2008;134:379. doi: 10.1053/j.gastro.2007.12.037. [DOI] [PubMed] [Google Scholar]
  • 34.Kane RC, Farrell AT, Saber H, Tang S, Williams G, Jee JM, et al. Sorafenib for the treatment of advanced renal cell carcinoma. Clin Cancer Res. 2006;12:7271–7278. doi: 10.1158/1078-0432.CCR-06-1249. [DOI] [PubMed] [Google Scholar]
  • 35.Ibrahim N, Yu Y, Walsh WR, Yang JL. Molecular targeted therapies for cancer: sorafenib mono-therapy and its combination with other therapies (review) Oncol Rep. 2012;27:1303–1311. doi: 10.3892/or.2012.1675. [DOI] [PubMed] [Google Scholar]
  • 36.Dal Lago L, D'Hondt V, Awada A. Selected combination therapy with sorafenib: a review of clinical data and perspectives in advanced solid tumors. Oncologist. 2008;13:845–858. doi: 10.1634/theoncologist.2007-0233. [DOI] [PubMed] [Google Scholar]
  • 37.Wilhelm SM, Adnane L, Newell P, Villanueva A, Llovet JM, Lynch M. Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129–3140. doi: 10.1158/1535-7163.MCT-08-0013. [DOI] [PubMed] [Google Scholar]
  • 38.Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099–7109. doi: 10.1158/0008-5472.CAN-04-1443. [DOI] [PubMed] [Google Scholar]
  • 39.Azad N, Dasari A, Arcaroli J, Taylor GE, Laheru DA, Carducci MA, et al. Phase I pharmacokinetic and pharmacodynamic study of cetuximab, irinotecan and sorafenib in advanced colorectal cancer. Invest New Drugs. 2013;31:345–354. doi: 10.1007/s10637-012-9820-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Case Reports in Oncology are provided here courtesy of Karger Publishers

RESOURCES