Korean J Radiol.  2017 Feb;18(1):6-17. 10.3348/kjr.2017.18.1.6.

Evolving Cancer Classification in the Era of Personalized Medicine: A Primer for Radiologists

Affiliations
  • 1Department of Imaging, Dana-Farber Cancer Institute, Boston, MA 02215, USA. ailbheconeill@yahoo.co.uk

Abstract

Traditionally tumors were classified based on anatomic location but now specific genetic mutations in cancers are leading to treatment of tumors with molecular targeted therapies. This has led to a paradigm shift in the classification and treatment of cancer. Tumors treated with molecular targeted therapies often show morphological changes rather than change in size and are associated with class specific and drug specific toxicities, different from those encountered with conventional chemotherapeutic agents. It is important for the radiologists to be familiar with the new cancer classification and the various treatment strategies employed, in order to effectively communicate and participate in the multi-disciplinary care. In this paper we will focus on lung cancer as a prototype of the new molecular classification.

Keyword

Lung cancer; NSCLC; Adenocarcinoma; EGFR mutation; ALK rearrangement

MeSH Terms

Antineoplastic Agents/therapeutic use
Carcinoma, Non-Small-Cell Lung/*classification/diagnostic imaging/drug therapy/genetics
Humans
Lung Neoplasms/*classification/diagnostic imaging/drug therapy/genetics
Molecular Targeted Therapy/methods
Mutation
Precision Medicine/*methods
Receptor Protein-Tyrosine Kinases/genetics
Receptor, Epidermal Growth Factor/genetics
Tomography, X-Ray Computed
Antineoplastic Agents
Receptor Protein-Tyrosine Kinases
Receptor, Epidermal Growth Factor

Figure

  • Fig. 1 Timeline of events in lung cancer molecular targeted therapies. ALK = anaplastic lymphoma kinase, EGFR = epidermal growth factor receptor, FDA = Food and Drug Administration

  • Fig. 2 Pie chart demonstrating percentage of driver alterations in lung adenocarcinoma. Adapted from Sholl et al. J Thorac Oncol 2015;10:768-777 (2). ALK = anaplastic lymphoma kinase, EGFR = epidermal growth factor receptor

  • Fig. 3 Axial contrast enhanced CT on soft tissue windows at four different time points in 57-year-old woman with EGFR exon 19 deletion lung adenocarcinoma. A. Initial image demonstrates large right upper lobe mass (arrow). B. Second image demonstrates response to first generation EGFR TKI (erlotinib) after 3 months with significant decrease in size of mass (arrow). C. Third image was performed 6 months later demonstrates further decrease in size of right upper lobe mass (arrow). D. Final image demonstrates gradual increase in size (arrow). Biopsy demonstrated T790M mutation that has known resistance to EGFR TKI (erlotinib) therapy. EGFR = epidermal growth factor receptor, TKI = tyrosine kinase inhibitor

  • Fig. 4 Axial CECT on lung windows in 53-year-old woman with L8585R point mutation in exon 21 adenocarcinoma. Patient had been treated with EGFR tyrosine kinase inhibitor (erlotinib) for 11 months but was progressing on serial CT studies. Initial CT (A) demonstrates multiple small right-sided pulmonary metastases and almost confluent opacification in left lung. Patient was taken off therapy and CT performed two weeks later due to patients dyspnea demonstrates now complete opacification in left hemithorax and interval increase in size in right sided pulmonary metastases, several had doubled in size (B). Appearances and clinical presentation were consistent with ‘flare’ phenomenon. CECT = contrast enhanced CT, EGFR = epidermal growth factor receptor

  • Fig. 5 Axial CECT on lung windows in 68-year-old woman with exon 19 EGFR mutant adenocarcinoma. A. Patient had dominant masses in right and left lower lobes (arrows) on initial CT. Patient was treated with EGFR tyrosine kinase inhibitor (erlotinib) and follow up CT performed 2 months later (B) demonstrates decrease in size of lower lobe masses (arrows). C. Patient also had developed multifocal faint bilateral subpleural ground glass changes (arrowheads) consistent with pneumonitis. Patient was asymptomatic and remained on treatment with changes on pneumonitis resolving on follow up studies. CECT = contrast enhanced CT, EGFR = epidermal growth factor receptor

  • Fig. 6 Axial contrast enhanced CT of thorax on lung windows (A, B) and axial T1 post gadolinium contrast MRI of brain (C) in 40-year-old man with metastatic lung adenocarcinoma with ALK rearrangement. On initial CT (A) patient has 2.5 × 2.3 cm mass in left upper lobe (arrow). Patient was treated with first generation ALK tyrosine kinase inhibitor (crizotinib) and had durable response in his lungs with second CT (B) post 5 year of treatment demonstrating small 1.2 × 0.6 cm ill-defined nodule at site of primary dominant mass (arrow). Patient unfortunately developed brain metastases due to poor blood brain barrier penetration with ALK inhibitor (crizotinib) and MRI (C) demonstrates ring enhancing 1.0 cm metastasis in right occipital lobe (arrowhead). ALK = anaplastic lymphoma kinase

  • Fig. 7 Axial contrast enhanced CT on soft tissue (A, B) and lung windows (C) in 38-year-old man lifelong non smoker with metastatic ALK rearranged lung adenocarcinoma. A, B. Initial CT demonstrates extensive left pleural nodularity and circumferential thickening with example of pleural thickening in left lower lobe included (arrows). C. Follow up performed 8 weeks after treatment with alectinib (second generation ALK inhibitor) demonstrated decrease in left pleural metastatic disease but interval development of new right lower lobe ground glass changes (arrowheads) consistent with pneumonitis. Patient was not symptomatic and therapy was continued. ALK = anaplastic lymphoma kinase

  • Fig. 8 Axial (A–D) and coronal enhanced (E, F) CT in 58-year-old woman with metastatic lung adenocarcinoma with ALK rearrangement. A, B. Initial CT demonstrates post obstructive atelectasis in left upper lobe from left hilar lymphadenopathy (not shown) and bilateral lung metastases, largest in left upper lobe measuring 1.3 x 1.3 cm (arrow) and in right lower lobe (arrow) measuring 1.6 × 1.4 cm. Patient was treated with crizotinib and follow up CT performed after 8 weeks of therapy (C, D) demonstrates response with resolution of metastases with only mild ground glass changes at site of dominant masses. E, F. Coronal CT demonstrates complication of crizotinib therapy with development of osteopenia best seen on CT as decreased sclerosis after 2 years of therapy within blastic bone metastasis (arrow). ALK = anaplastic lymphoma kinase

  • Fig. 9 Axial CECT at floor of mouth in 58-year-old man with EGFR mutant squamous cell carcinoma of tongue. A. Patient had been treated with right sided hemiglossectomy but developed large enhancing mass in surgical site on right (arrow) and level IIa low density left cervical chain lymph node (arrowhead) on CT. B. Patient was treated with EGFR monoclonal antibody (cetuximab) and follow up CT performed 2 months later demonstrates resolution of mass on right and significant decrease in left cervical lymph node. CECT = contrast enhanced CT, EGFR = epidermal growth factor receptor

  • Fig. 10 MIP (A, C) and axial non contrast fused 18F-FDG PET/CT (B) in 20-year-old man with ALK positive anaplastic large cell lymphoma. A. Patient had been diagnosed one year earlier and treated with brentuximab but had relapsed 6 months after completing treatment. New baseline PET/CT demonstrated residual lymphadenopathy in right external iliac and inguinal lymph nodes (arrows). B. Axial fused PET/CT demonstrates FDG avid right inguinal lymph node (arrow) with SUVmax 5.5. C. Patient was treated with salvage chemotherapy (gemcitabine, vinorelbine and doxil) with response to treatment and resolution of right pelvic lymphadenopathy on PET/CT performed 8 weeks later. ALK = anaplastic lymphoma kinase, MIP = maximum intensity projection, PET = positron emission tomography


Reference

1. Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC. Tumours of the Lung, Pleura, Thymus and Heart. Lyon: IARC;2004.
2. Sholl LM, Aisner DL, Varella-Garcia M, Berry LD, Dias-Santagata D, Wistuba II, et al. Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: the lung cancer mutation consortium experience. J Thorac Oncol. 2015; 10:768–777.
3. Ha SY, Choi SJ, Cho JH, Choi HJ, Lee J, Jung K, et al. Lung cancer in never-smoker asian females is driven by oncogenic mutations, most often involving EGFR. Oncotarget. 2015; 6:5465–5474.
4. Hsu KH, Ho CC, Hsia TC, Tseng JS, Su KY, Wu MF, et al. Identification of five driver gene mutations in patients with treatment-naïve lung adenocarcinoma in Taiwan. PLoS One. 2015; 10:e0120852.
5. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JH, Beasley MB, et al. The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 2015; 10:1243–1260.
6. Schlessinger J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell. 2002; 110:669–672.
7. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002; 2:489–501.
8. Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 1995; 19:183–232.
9. Mologni L. Inhibitors of the anaplastic lymphoma kinase. Expert Opin Investig Drugs. 2012; 21:985–994.
10. 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.
11. Mologni L. Current and future treatment of anaplastic lymphoma kinase-rearranged cancer. World J Clin Oncol. 2015; 6:104–108.
12. Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004; 101:13306–13311.
13. Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005; 97:339–346.
14. Hasegawa M, Sakai F, Ishikawa R, Kimura F, Ishida H, Kobayashi K. CT Features of epidermal growth factor receptor-mutated adenocarcinoma of the lung: comparison with nonmutated adenocarcinoma. J Thorac Oncol. 2016; 11:819–826.
15. Liu Y, Kim J, Qu F, Liu S, Wang H, Balagurunathan Y, et al. CT features associated with epidermal growth factor receptor mutation status in patients with lung adenocarcinoma. Radiology. 2016; 280:271–280.
16. Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science. 2004; 305:1163–1167.
17. Tracy S, Mukohara T, Hansen M, Meyerson M, Johnson BE, Jänne PA. Gefitinib induces apoptosis in the EGFRL858R non-small-cell lung cancer cell line H3255. Cancer Res. 2004; 64:7241–7244.
18. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007; 7:169–181.
19. 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.
20. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res. 2013; 19:2240–2247.
21. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007; 316:1039–1043.
22. Riely GJ, Kris MG, Zhao B, Akhurst T, Milton DT, Moore E, et al. Prospective assessment of discontinuation and reinitiation of erlotinib or gefitinib in patients with acquired resistance to erlotinib or gefitinib followed by the addition of everolimus. Clin Cancer Res. 2007; 13:5150–5155.
23. Chaft JE, Oxnard GR, Sima CS, Kris MG, Miller VA, Riely GJ. Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFR-mutant lung cancer and acquired resistance to erlotinib or gefitinib: implications for clinical trial design. Clin Cancer Res. 2011; 17:6298–6303.
24. Hughes B, Mileshkin L, Townley P, Gitlitz B, Eaton K, Mitchell P, et al. Pertuzumab and erlotinib in patients with relapsed non-small cell lung cancer: a phase II study using 18F-fluorodeoxyglucose positron emission tomography/computed tomography imaging. Oncologist. 2014; 19:175–176.
25. Howard SA, Rosenthal MH, Jagannathan JP, Krajewski KM, Shinagare AB, Ramaiya NH, et al. Beyond the vascular endothelial growth factor axis: update on role of imaging in nonantiangiogenic molecular targeted therapies in oncology. AJR Am J Roentgenol. 2015; 204:919–932.
26. Thornton E, Howard SA, Jagannathan J, Krajewski KM, Shinagare AB, O'Regan K, et al. Imaging features of bowel toxicities in the setting of molecular targeted therapies in cancer patients. Br J Radiol. 2012; 85:1420–1426.
27. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 2009; 27:4247–4253.
28. Yamamoto S, Korn RL, Oklu R, Migdal C, Gotway MB, Weiss GJ, et al. ALK molecular phenotype in non-small cell lung cancer: CT radiogenomic characterization. Radiology. 2014; 272:568–576.
29. Ou SH, Bartlett CH, Mino-Kenudson M, Cui J, Iafrate AJ. Crizotinib for the treatment of ALK-rearranged non-small cell lung cancer: a success story to usher in the second decade of molecular targeted therapy in oncology. Oncologist. 2012; 17:1351–1375.
30. Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn MJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013; 368:2385–2394.
31. Ou SH, Bazhenova L, Camidge DR, Solomon BJ, Herman J, Kain T, et al. Rapid and dramatic radiographic and clinical response to an ALK inhibitor (crizotinib, PF02341066) in an ALK translocation-positive patient with non-small cell lung cancer. J Thorac Oncol. 2010; 5:2044–2046.
32. Nishino M, Hatabu H, Johnson BE, McLoud TC. State of the art: response assessment in lung cancer in the era of genomic medicine. Radiology. 2014; 271:6–27.
33. Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, et al. EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 2010; 363:1734–1739.
34. Costa DB, Shaw AT, Ou SH, Solomon BJ, Riely GJ, Ahn MJ, et al. Clinical experience with crizotinib in patients with advanced ALK-rearranged non-small-cell lung cancer and brain metastases. J Clin Oncol. 2015; 33:1881–1888.
35. Kinoshita K, Asoh K, Furuichi N, Ito T, Kawada H, Hara S, et al. Design and synthesis of a highly selective, orally active and potent anaplastic lymphoma kinase inhibitor (CH5424802). Bioorg Med Chem. 2012; 20:1271–1280.
36. Gadgeel SM, Gandhi L, Riely GJ, Chiappori AA, West HL, Azada MC, et al. Safety and activity of alectinib against systemic disease and brain metastases in patients with crizotinib-resistant ALK-rearranged non-small-cell lung cancer (AF-002JG): results from the dose-finding portion of a phase 1/2 study. Lancet Oncol. 2014; 15:1119–1128.
37. Schnell P, Bartlett CH, Solomon BJ, Tassell V, Shaw AT, de Pas T, et al. Complex renal cysts associated with crizotinib treatment. Cancer Med. 2015; 4:887–896.
38. Weickhardt AJ, Rothman MS, Salian-Mehta S, Kiseljak-Vassiliades K, Oton AB, Doebele RC, et al. Rapid-onset hypogonadism secondary to crizotinib use in men with metastatic nonsmall cell lung cancer. Cancer. 2012; 118:5302–5309.
39. Cadoo KA, Fornier MN, Morris PG. Biological subtypes of breast cancer: current concepts and implications for recurrence patterns. Q J Nucl Med Mol Imaging. 2013; 57:312–321.
40. Morrow M. Personalizing extent of breast cancer surgery according to molecular subtypes. Breast. 2013; 22:Suppl 2. S106–S109.
41. Kennecke H, Yerushalmi R, Woods R, Cheang MC, Voduc D, Speers CH, et al. Metastatic behavior of breast cancer subtypes. J Clin Oncol. 2010; 28:3271–3277.
42. Bria E, Cuppone F, Fornier M, Nisticò C, Carlini P, Milella M, et al. Cardiotoxicity and incidence of brain metastases after adjuvant trastuzumab for early breast cancer: the dark side of the moon? A meta-analysis of the randomized trials. Breast Cancer Res Treat. 2008; 109:231–223.
43. Ang KK, Berkey BA, Tu X, Zhang HZ, Katz R, Hammond EH, et al. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002; 62:7350–7356.
44. Seiwert TY, Zuo Z, Keck MK, Khattri A, Pedamallu CS, Stricker T, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res. 2015; 21:632–641.
45. Bonner JA, Harari PM, Giralt J, Cohen RB, Jones CU, Sur RK, et al. Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival. Lancet Oncol. 2010; 11:21–28.
46. Yazdi MH, Faramarzi MA, Nikfar S, Abdollahi M. A comprehensive review of clinical trials on EGFR inhibitors such as cetuximab and panitumumab as monotherapy and in combination for treatment of metastatic colorectal cancer. Avicenna J Med Biotechnol. 2015; 7:134–144.
47. 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; 22:1201–1218.
48. Dong F, Kojiro S, Borger DR, Growdon WB, Oliva E. Squamous cell carcinoma of the vulva: a subclassification of 97 cases by clinicopathologic, immunohistochemical, and molecular features (p16, p53, and EGFR). Am J Surg Pathol. 2015; 39:1045–1053.
49. Santos M, Landolfi S, Olivella A, Lloveras B, Klaustermeier J, Suárez H, et al. p16 overexpression identifies HPV-positive vulvar squamous cell carcinomas. Am J Surg Pathol. 2006; 30:1347–1356.
50. Scheistrøen M, Tropé C, Pettersen EO, Nesland JM. p53 protein expression in squamous cell carcinoma of the vulva. Cancer. 1999; 85:1133–1138.
51. Growdon WB, Boisvert SL, Akhavanfard S, Oliva E, Dias-Santagata DC, Kojiro S, et al. Decreased survival in EGFR gene amplified vulvar carcinoma. Gynecol Oncol. 2008; 111:289–297.
52. Johnson GA, Mannel R, Khalifa M, Walker JL, Wren M, Min KW, et al. Epidermal growth factor receptor in vulvar malignancies and its relationship to metastasis and patient survival. Gynecol Oncol. 1997; 65:425–429.
53. Horowitz NS, Olawaiye AB, Borger DR, Growdon WB, Krasner CN, Matulonis UA, et al. Phase II trial of erlotinib in women with squamous cell carcinoma of the vulva. Gynecol Oncol. 2012; 127:141–146.
54. Brugières L, Pacquement H, Le Deley MC, Leverger G, Lutz P, Paillard C, et al. Single-drug vinblastine as salvage treatment for refractory or relapsed anaplastic large-cell lymphoma: a report from the French Society of Pediatric Oncology. J Clin Oncol. 2009; 27:5056–5061.
55. Sibon D, Fournier M, Brière J, Lamant L, Haioun C, Coiffier B, et al. Long-term outcome of adults with systemic anaplastic large-cell lymphoma treated within the Groupe d'Etude des Lymphomes de l'Adulte trials. J Clin Oncol. 2012; 30:3939–3946.
56. Mossé YP, Lim MS, Voss SD, Wilner K, Ruffner K, Laliberte J, et al. Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children's Oncology Group phase 1 consortium study. Lancet Oncol. 2013; 14:472–480.
57. Coffin CM, Watterson J, Priest JR, Dehner LP. Extrapulmonary inflammatory myofibroblastic tumor (inflammatory pseudotumor). A clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol. 1995; 19:859–872.
58. Coffin CM, Hornick JL, Fletcher CD. Inflammatory myofibroblastic tumor: comparison of clinicopathologic, histologic, and immunohistochemical features including ALK expression in atypical and aggressive cases. Am J Surg Pathol. 2007; 31:509–520.
59. Antonescu CR, Suurmeijer AJ, Zhang L, Sung YS, Jungbluth AA, Travis WD, et al. Molecular characterization of inflammatory myofibroblastic tumors with frequent ALK and ROS1 gene fusions and rare novel RET rearrangement. Am J Surg Pathol. 2015; 39:957–967.
60. Chan JK, Cheuk W, Shimizu M. Anaplastic lymphoma kinase expression in inflammatory pseudotumors. Am J Surg Pathol. 2001; 25:761–768.
61. Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, et al. The genetic landscape of high-risk neuroblastoma. Nat Genet. 2013; 45:279–284.
62. Mossé YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008; 455:930–935.
63. De Brouwer S, De Preter K, Kumps C, Zabrocki P, Porcu M, Westerhout EM, et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin Cancer Res. 2010; 16:4353–4362.
Full Text Links
  • KJR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr