Korean J Radiol.  2017 Feb;18(1):28-41. 10.3348/kjr.2017.18.1.28.

Molecular Targeted Therapy in Modern Oncology: Imaging Assessment of Treatment Response and Toxicities

Affiliations
  • 1Department of Imaging, Dana-Farber Cancer Institute, Boston, MA 02215, USA. kmkrajewski@partners.org
  • 2Department of Radiology, Brigham and Women's Hospital, Boston, MA 02115, USA.

Abstract

Oncology is a rapidly evolving field with a shift toward personalized cancer treatment. The use of therapies targeted to the molecular features of individual tumors and the tumor microenvironment has become much more common. In this review, anti-angiogenic and other molecular targeted therapies are discussed, with a focus on typical and atypical response patterns and imaging manifestations of drug toxicities.

Keyword

Vascular endothelial growth factor; Response Evaluation Criteria in Solid Tumors; Epidermal growth factor receptor; Anaplastic lymphoma kinase; Mammalian target of rapamycin; Human epidermal growth factor receptor 2

MeSH Terms

Angiogenesis Inhibitors/adverse effects/therapeutic use
Antineoplastic Agents/adverse effects/therapeutic use
Humans
Molecular Targeted Therapy/adverse effects/*methods
Neoplasms/*diagnostic imaging/*drug therapy/genetics
Tomography, X-Ray Computed
Treatment Outcome
Angiogenesis Inhibitors
Antineoplastic Agents

Figure

  • Fig. 1 Typical response of metastatic clear cell renal cell carcinoma to VEGF-targeted therapy and subsequent tumor growth off of treatment. In this 55-year-old woman, baseline contrast-enhanced CT prior to start of treatment demonstrates vascularized right hilar metastasis (arrow, A), pelvic serosal deposit (arrow, B) and left iliac bone metastasis (arrowhead, B). After 10 weeks of axitinib treatment, each metastasis is decreased in size and density (C, D). Patient subsequently developed wound infection in her right lower extremity, resulting in hold in axitinib treatment. After 6 weeks of holding treatment (and 18 weeks from first follow-up CT), right hilar mass (E), and pelvic serosal metastasis (arrow, F) are increased in size and enhancement, representing tumor growth. In addition, rim enhancement is newly seen involving left iliac bone lesion (arrowhead), also representing increasing tumor at site. VEGF = vascular endothelial growth factor

  • Fig. 2 Atypical response to VEGF-targeted therapy in colon cancer. In this 57-year-old woman with metastatic colon cancer, baseline contrast-enhanced CT (A) demonstrates multiple heterogeneous liver metastases. Serum CEA at baseline measured 2049. After 12 weeks of therapy with folinic acid, fluorouracil, oxaliplatin (FOLFOX) and avastin, multiple liver metastases have increased in size but have decreased in density (B), showing greater homogeneity and better defined margins. Serum CEA at time of 12 week follow-up CT measured 1126. Findings at follow-up CT represent response according to CT-morphologic criteria. CEA = carcinoembryonic antigen, VEGF = vascular endothelial growth factor

  • Fig. 3 Hepatocellular cancer (HCC) with response to VEGF-targeted therapy according to mRECIST. Baseline contrast-enhanced CT (A) demonstrates vascularized, enhancing tumor thrombus in right anterior and posterior portal vein branches (arrows) in this 59-year-old man with multifocal HCC. Follow-up contrast-enhanced CT after 2 months of VEGF-targeted therapy shows loss of hypervascularity of tumor thrombus (B), in keeping with response to therapy according to mRECIST. mRECIST = modified Response Evaluation Criteria in Solid Tumors, VEGF = vascular endothelial growth factor

  • Fig. 4 Gastrointestinal stromal tumor with response to sunitinib and tumor bowel fistula development. In this 76-year-old woman, baseline axial contrast-enhanced CT (A) demonstrates mass adjacent to small bowel in left lower quadrant (white arrow). Follow-up contrast-enhanced CT (B) demonstrates new oral contrast within tumor lesion, contiguous with small bowel lumen (black arrow).

  • Fig. 5 EGFR-mutant lung cancer with response to treatment, development of resistant disease, flare off of therapy and subsequent response to second-line treatment. Baseline CT in this 62-year-old never smoker demonstrates large left upper lobe perihilar mass with interlobular septal thickening emanating from mass (A), multistation lymphadenopathy is not shown. CT follow-up 5 months after erlotinib treatment shows dramatic shrinkage of left perihilar mass (B). Subsequent follow-up CT 8 months after start of treatment shows mild increase in left perihilar tumor indicative of resistant disease (not shown). Lymph node biopsy confirmed T790M secondary resistance mutation. Erlotinib therapy was discontinued in anticipation of clinical trial participation. One month after erlotinib discontinuation, patient presented with shortness of breath, cough and chest pain. CT chest revealed flare phenomenon, represented by marked increase in tumor burden in both lungs (C, D). Erlotinib was re-initiated with carboplatin and pemetrexed chemotherapy, and tumor burden significantly decreased 8 weeks later (E, F). EGFR = epidermal growth factor receptor

  • Fig. 6 EMF4-ALK rearranged lung cancer response to crizotinib and associated toxicity. Baseline contrast-enhanced CT in this 50-year-old man demonstrates large right lateral chest wall mass (arrow, A) which responded dramatically to crizotinib therapy, with little if any residual mass after 3 months of treatment (B). Thoracic vertebral bodies were normal in height at baseline (C). Demineralization and compression deformities developed during course of treatment (arrow, D), known toxicity of crizotinib related to rapid onset hypogonadism and low testosterone. ALK = anaplastic lymphoma kinase, EMF4= echinoderm microtubule-associated protein-like 4

  • Fig. 7 Response to mTOR-inhibitor treatment in Waldenstrom's macroglobulinemia and lung toxicity. Baseline contrast-enhanced coronal CT images in 59-year-old woman show retroperitoneal lymphadenopathy (black arrows) and mild splenomegaly (A). Five months after start of treatment, coronal contrast-enhanced CT shows decreased adenopathy and splenomegaly (B). At baseline, clear lung bases were present (C). On five month follow-up CT (D), new ground-glass and reticular opacities developed in lung bases (white arrows), in keeping with mTOR-associated pneumonitis. Patient was asymptomatic. mTOR = mammalian target of rapamycin

  • Fig. 8 Response to aromatase inhibitor and cyclin dependent kinase inhibitor in metastatic hormone receptor+, HER2 negative breast cancer. In this 29-year-old woman, baseline coronal CT (A) demonstrates multiple liver metastases (arrow). Follow-up coronal contrast-enhanced CT (B) demonstrates decreased size of multiple liver lesions (arrow) representing response to treatment. HER2 = human epidermal growth factor receptor 2


Cited by  1 articles

Solitary Nodular Invasive Mucinous Adenocarcinoma of the Lung: Imaging Diagnosis Using the Morphologic-Metabolic Dissociation Sign
Min Jae Cha, Kyung Soo Lee, Tae Jung Kim, Hyun Su Kim, Tae Sung Kim, Myung Jin Chung, Byung Tae Kim, Yang Soo Kim
Korean J Radiol. 2019;20(3):513-521.    doi: 10.3348/kjr.2018.0409.


Reference

1. Mendelsohn J. Personalizing oncology: perspectives and prospects. J Clin Oncol. 2013; 31:1904–1911.
2. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971; 285:1182–1186.
3. Lee JC, Chow NH, Wang ST, Huang SM. Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. Eur J Cancer. 2000; 36:748–753.
4. Berger DP, Herbstritt L, Dengler WA, Marmé D, Mertelsmann R, Fiebig HH. Vascular endothelial growth factor (VEGF) mRNA expression in human tumor models of different histologies. Ann Oncol. 1995; 6:817–825.
5. Krieg M, Haas R, Brauch H, Acker T, Flamme I, Plate KH. Upregulation of hypoxia-inducible factors HIF-1alpha and HIF-2alpha under normoxic conditions in renal carcinoma cells by von Hippel-Lindau tumor suppressor gene loss of function. Oncogene. 2000; 19:5435–5443.
6. Ohta Y, Endo Y, Tanaka M, Shimizu J, Oda M, Hayashi Y, et al. Significance of vascular endothelial growth factor messenger RNA expression in primary lung cancer. Clin Cancer Res. 1996; 2:1411–1416.
7. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004; 350:2335–2342.
8. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006; 355:2542–2550.
9. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 2003; 349:427–434.
10. Tewari KS, Sill MW, Long HJ 3rd, Penson RT, Huang H, Ramondetta LM, et al. Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med. 2014; 370:734–743.
11. Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011; 365:2484–2496.
12. Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011; 365:2473–2483.
13. Ciombor KK, Berlin J, Chan E. Aflibercept. Clin Cancer Res. 2013; 19:1920–1925.
14. Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012; 30:3499–3506.
15. Tirumani SH, Fairchild A, Krajewski KM, Nishino M, Howard SA, Baheti AD, et al. Anti-VEGF molecular targeted therapies in common solid malignancies: comprehensive update for radiologists. Radiographics. 2015; 35:455–474.
16. 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.
17. Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med. 2013; 369:722–731.
18. Goodman VL, Rock EP, Dagher R, Ramchandani RP, Abraham S, Gobburu JV, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007; 13:1367–1373.
19. Rehman S, Jayson GC. Molecular imaging of antiangiogenic agents. Oncologist. 2005; 10:92–103.
20. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000; 92:205–216.
21. Thiam R, Fournier LS, Trinquart L, Medioni J, Chatellier G, Balvay D, et al. Optimizing the size variation threshold for the CT evaluation of response in metastatic renal cell carcinoma treated with sunitinib. Ann Oncol. 2010; 21:936–941.
22. Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nat Rev Cancer. 2002; 2:727–739.
23. Nishino M, Jagannathan JP, Krajewski KM, O'Regan K, Hatabu H, Shapiro G, et al. Personalized tumor response assessment in the era of molecular medicine: cancer-specific and therapy-specific response criteria to complement pitfalls of RECIST. AJR Am J Roentgenol. 2012; 198:737–745.
24. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010; 28:1963–1972.
25. Chun YS, Vauthey JN, Boonsirikamchai P, Maru DM, Kopetz S, Palavecino M, et al. Association of computed tomography morphologic criteria with pathologic response and survival in patients treated with bevacizumab for colorectal liver metastases. JAMA. 2009; 302:2338–2344.
26. Crabb SJ, Patsios D, Sauerbrei E, Ellis PM, Arnold A, Goss G, et al. Tumor cavitation: impact on objective response evaluation in trials of angiogenesis inhibitors in non-small-cell lung cancer. J Clin Oncol. 2009; 27:404–410.
27. Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010; 30:52–60.
28. Arizumi T, Ueshima K, Takeda H, Osaki Y, Takita M, Inoue T, et al. Comparison of systems for assessment of post-therapeutic response to sorafenib for hepatocellular carcinoma. J Gastroenterol. 2014; 49:1578–1587.
29. van der Veldt AA, Meijerink MR, van den Eertwegh AJ, Haanen JB, Boven E. Choi response criteria for early prediction of clinical outcome in patients with metastatic renal cell cancer treated with sunitinib. Br J Cancer. 2010; 102:803–809.
30. Smith AD, Shah SN, Rini BI, Lieber ML, Remer EM. Morphology, attenuation, size, and structure (MASS) criteria: assessing response and predicting clinical outcome in metastatic renal cell carcinoma on antiangiogenic targeted therapy. AJR Am J Roentgenol. 2010; 194:1470–1478.
31. Krajewski KM, Guo M, Van den Abbeele AD, Yap J, Ramaiya N, Jagannathan J, et al. Comparison of four early posttherapy imaging changes (EPTIC; RECIST 1.0, tumor shrinkage, computed tomography tumor density, Choi criteria) in assessing outcome to vascular endothelial growth factor-targeted therapy in patients with advanced renal cell carcinoma. Eur Urol. 2011; 59:856–862.
32. Badgwell BD, Camp ER, Feig B, Wolff RA, Eng C, Ellis LM, et al. Management of bevacizumab-associated bowel perforation: a case series and review of the literature. Ann Oncol. 2008; 19:577–582.
33. Shinagare AB, Howard SA, Krajewski KM, Zukotynski KA, Jagannathan JP, Ramaiya NH. Pneumatosis intestinalis and bowel perforation associated with molecular targeted therapy: an emerging problem and the role of radiologists in its management. AJR Am J Roentgenol. 2012; 199:1259–1265.
34. Howard SA, Krajewski KM, Thornton E, Jagannathan JP, O'Regan K, Cleary J, et al. Decade of molecular targeted therapy: abdominal manifestations of drug toxicities--what radiologists should know. AJR Am J Roentgenol. 2012; 199:58–64.
35. Hapani S, Sher A, Chu D, Wu S. Increased risk of serious hemorrhage with bevacizumab in cancer patients: a meta-analysis. Oncology. 2010; 79:27–38.
36. Schutz FA, Je Y, Azzi GR, Nguyen PL, Choueiri TK. Bevacizumab increases the risk of arterial ischemia: a large study in cancer patients with a focus on different subgroup outcomes. Ann Oncol. 2011; 22:1404–1412.
37. Choueiri TK, Schutz FA, Je Y, Rosenberg JE, Bellmunt J. Risk of arterial thromboembolic events with sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. J Clin Oncol. 2010; 28:2280–2285.
38. Nalluri SR, Chu D, Keresztes R, Zhu X, Wu S. Risk of venous thromboembolism with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis. JAMA. 2008; 300:2277–2285.
39. Hurwitz HI, Saltz LB, Van Cutsem E, Cassidy J, Wiedemann J, Sirzén F, et al. Venous thromboembolic events with chemotherapy plus bevacizumab: a pooled analysis of patients in randomized phase II and III studies. J Clin Oncol. 2011; 29:1757–1764.
40. Dienstmann R, Braña I, Rodon J, Tabernero J. Toxicity as a biomarker of efficacy of molecular targeted therapies: focus on EGFR and VEGF inhibiting anticancer drugs. Oncologist. 2011; 16:1729–1740.
41. Singer S, Grommes C, Reiner AS, Rosenblum MK, DeAngelis LM. Posterior reversible encephalopathy syndrome in patients with cancer. Oncologist. 2015; 20:806–811.
42. Tirumani S, Baheti A, Nishino M, Krajewski K, Rosenthal M, Ramaiya N. The cancer imaging radiology report template in the era of molecular-targeted therapy (MTT): what should be included in the radiology interpretation and why. Chicago, IL: Radiological Society of North America 2014 Scientific Assembly and Annual Meeting;Accessed June 29, 2016. Web site. http://archive.rsna.org/2014/14009488.html.
43. Shinagare AB, Jagannathan JP, Krajewski KM, Ramaiya NH. Liver metastases in the era of molecular targeted therapy: new faces of treatment response. AJR Am J Roentgenol. 2013; 201:W15–W28.
44. Herbst RS, Heymach JV, Lippman SM. Lung cancer. N Engl J Med. 2008; 359:1367–1380.
45. Sequist LV, Bell DW, Lynch TJ, Haber DA. Molecular predictors of response to epidermal growth factor receptor antagonists in non-small-cell lung cancer. J Clin Oncol. 2007; 25:587–595.
46. Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004; 304:1497–1500.
47. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004; 350:2129–2139.
48. 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.
49. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010; 363:1693–1703.
50. Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, et al. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014; 370:1189–1197.
51. Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Jänne PA, et al. First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol. 2008; 26:2442–2449.
52. Inoue A, Suzuki T, Fukuhara T, Maemondo M, Kimura Y, Morikawa N, et al. Prospective phase II study of gefitinib for chemotherapy-naive patients with advanced non-small-cell lung cancer with epidermal growth factor receptor gene mutations. J Clin Oncol. 2006; 24:3340–3346.
53. Nishino M, Jackman DM, Hatabu H, Jänne PA, Johnson BE, Van den Abbeele AD. Imaging of lung cancer in the era of molecular medicine. Acad Radiol. 2011; 18:424–436.
54. 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.
55. Nishino M, Dahlberg SE, Fulton LE, Digumarthy SR, Hatabu H, Johnson BE, et al. Volumetric tumor response and progression in EGFR-mutant NSCLC patients treated with erlotinib or gefitinib. Acad Radiol. 2016; 23:329–336.
56. 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.
57. Nishino M, Cardarella S, Dahlberg SE, Jackman DM, Ramaiya NH, Hatabu H, et al. Radiographic assessment and therapeutic decisions at RECIST progression in EGFR-mutant NSCLC treated with EGFR tyrosine kinase inhibitors. Lung Cancer. 2013; 79:283–288.
58. Park K, Yu CJ, Kim SW, Lin MC, Sriuranpong V, Tsai CM, et al. First-line erlotinib therapy until and beyond response evaluation criteria in solid tumors progression in asian patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer: the ASPIRATION study. JAMA Oncol. 2016; 2:305–312.
59. Nishino M, Dahlberg SE, Cardarella S, Jackman DM, Rabin MS, Ramaiya NH, et al. Volumetric tumor growth in advanced non-small cell lung cancer patients with EGFR mutations during EGFR-tyrosine kinase inhibitor therapy: developing criteria to continue therapy beyond RECIST progression. Cancer. 2013; 119:3761–3768.
60. Yang J, Ramalingam SS, Jänne PA, Cantarini M, Mitsudomi T. LBA2_PR: Osimertinib (AZD9291) in pre-treated pts with T790M-positive advanced NSCLC: updated Phase 1 (P1) and pooled Phase 2 (P2) results. J Thorac Oncol. 2016; 11:4 Suppl. S152–S153.
61. Endo M, Johkoh T, Kimura K, Yamamoto N. Imaging of gefitinib-related interstitial lung disease: multi-institutional analysis by the West Japan Thoracic Oncology Group. Lung Cancer. 2006; 52:135–140.
62. Herbst RS, Prager D, Hermann R, Fehrenbacher L, Johnson BE, Sandler A, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol. 2005; 23:5892–5899.
63. 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.
64. 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.
65. Miller JA, Ford DJ, Ahmed MS, Loree TR. Two cases of pneumatosis intestinalis during cetuximab therapy for advanced head and neck cancer. Case Rep Oncol Med. 2015; 2015:214236.
66. Lin YT, Wang YF, Yang JC, Yu CJ, Wu SG, Shih JY, et al. Development of renal cysts after crizotinib treatment in advanced ALK-positive non-small-cell lung cancer. J Thorac Oncol. 2014; 9:1720–1725.
67. 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.
68. Gibbons JJ, Abraham RT, Yu K. Mammalian target of rapamycin: discovery of rapamycin reveals a signaling pathway important for normal and cancer cell growth. Semin Oncol. 2009; 36:Suppl 3. S3–S17.
69. Shinagare AB, Krajewski KM, Jagannathan JP, Ramaiya NH. Genitourinary imaging: part 2, role of imaging in medical management of advanced renal cell carcinoma. AJR Am J Roentgenol. 2012; 199:W554–W564.
70. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007; 356:2271–2281.
71. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008; 372:449–456.
72. Ghobrial IM, Gertz M, Laplant B, Camoriano J, Hayman S, Lacy M, et al. Phase II trial of the oral mammalian target of rapamycin inhibitor everolimus in relapsed or refractory Waldenstrom macroglobulinemia. J Clin Oncol. 2010; 28:1408–1414.
73. Oudard S, Thiam R, Fournier LS, Medioni J, Lamuraglia M, Scotte F, et al. Optimisation of the tumour response threshold in patients treated with everolimus for metastatic renal cell carcinoma: analysis of response and progression-free survival in the RECORD-1 study. Eur J Cancer. 2012; 48:1512–1518.
74. Krajewski KM, Nishino M, Franchetti Y, Ramaiya NH, Van den Abbeele AD, Choueiri TK. Intraobserver and interobserver variability in computed tomography size and attenuation measurements in patients with renal cell carcinoma receiving antiangiogenic therapy: implications for alternative response criteria. Cancer. 2014; 120:711–721.
75. Lamuraglia M, Raslan S, Elaidi R, Oudard S, Escudier B, Slimane K, et al. mTOR-inhibitor treatment of metastatic renal cell carcinoma: contribution of Choi and modified Choi criteria assessed in 2D or 3D to evaluate tumor response. Eur Radiol. 2016; 26:278–285.
76. Maroto JP, Hudes G, Dutcher JP, Logan TF, White CS, Krygowski M, et al. Drug-related pneumonitis in patients with advanced renal cell carcinoma treated with temsirolimus. J Clin Oncol. 2011; 29:1750–1756.
77. Dabydeen DA, Jagannathan JP, Ramaiya N, Krajewski K, Schutz FA, Cho DC, et al. Pneumonitis associated with mTOR inhibitors therapy in patients with metastatic renal cell carcinoma: incidence, radiographic findings and correlation with clinical outcome. Eur J Cancer. 2012; 48:1519–1524.
78. Atkinson BJ, Cauley DH, Ng C, Millikan RE, Xiao L, Corn P, et al. mTOR inhibitor associated noninfectious pneumonitis in patients with renal cell cancer: management, predictors, and outcomes. BJU Int. 2014; 113:376–382.
79. Nishino M, Boswell EN, Hatabu H, Ghobrial IM, Ramaiya NH. Drug-related pneumonitis during mammalian target of rapamycin inhibitor therapy: radiographic pattern-based approach in Waldenström macroglobulinemia as a paradigm. Oncologist. 2015; 20:1077–1083.
80. Nishino M, Brais LK, Brooks NV, Hatabu H, Kulke MH, Ramaiya NH. Drug-related pneumonitis during mammalian target of rapamycin inhibitor therapy in patients with neuroendocrine tumors: a radiographic pattern-based approach. Eur J Cancer. 2016; 53:163–170.
81. Tirumani SH, Krajewski KM, Shinagare AB, Jagannathan JP, Ramaiya NH. Gallbladder complications associated with molecular targeted therapies: clinical and imaging features. Clin Imaging. 2014; 38:50–55.
82. Tirumani SH, Jagannathan JP, Shinagare AB, Kim KW, Krajewski KM, Ramaiya NH. Acute pancreatitis associated with molecular targeted therapies: a retrospective review of the clinico-radiological features, management and outcome. Pancreatology. 2013; 13:461–467.
83. Parithivel K, Ramaiya N, Jagannathan JP, O'Regan K, Krajewski K, Fisher D, et al. Everolimus- and temsirolimus-associated enteritis: report of three cases. J Clin Oncol. 2011; 29:e404–e406.
84. National Clinical Practice Guidelines in Oncology (NCCN guidelines): Breast cancer-version 3. 2015. Accessed November 29, 2016. Web site. http://www.consensocancermamario.com/guias/NCCN_2015.pdf.
85. Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet Oncol. 2015; 16:25–35.
86. Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 2016; 17:425–439.
87. Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, Sahmoud T, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012; 366:520–529.
88. Cooley C, Nishino M, Jagannathan J, Ramaiya N, Di Salvo D, Krajewski KM. Clinical problem solving: utility of sonography in oncologic patients. J Ultrasound Med. 2014; 33:9–22.
89. Hadji P, Aapro MS, Body JJ, Bundred NJ, Brufsky A, Coleman RE, et al. Management of aromatase inhibitor-associated bone loss in postmenopausal women with breast cancer: practical guidance for prevention and treatment. Ann Oncol. 2011; 22:2546–2555.
90. Pegram MD, Lipton A, Hayes DF, Weber BL, Baselga JM, Tripathy D, et al. Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J Clin Oncol. 1998; 16:2659–2671.
91. Baselga J, Cortés J, Kim SB, Im SA, Hegg R, Im YH, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012; 366:109–119.
92. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006; 355:2733–2743.
93. Lin NU, Diéras V, Paul D, Lossignol D, Christodoulou C, Stemmler HJ, et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009; 15:1452–1459.
94. Krop IE, LoRusso P, Miller KD, Modi S, Yardley D, Rodriguez G, et al. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol. 2012; 30:3234–3241.
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