Breast Cancer DOI 10.1007/s12282-015-0613-z

SPECIAL FEATURE

The way to the next generation molecular diagnostics

Molecular imaging using PET for breast cancer Hiroaki Kurihara1 • Chikako Shimizu2 • Yasuji Miyakita3 • Masayuki Yoshida4 Akinobu Hamada5 • Yousuke Kanayama6 • Kan Yonemori2 • Jun Hashimoto2 • Hitomi Tani1 • Makoto Kodaira2 • Mayu Yunokawa2 • Harukaze Yamamoto2 • Yasuyoshi Watanabe6 • Yasuhiro Fujiwara2 • Kenji Tamura2



Received: 26 February 2015 / Accepted: 16 April 2015 Ó The Japanese Breast Cancer Society 2015

Abstract Molecular imaging can visualize the biological processes at the molecular and cellular levels in vivo using certain tracers for specific molecular targets. Molecular imaging of breast cancer can be performed with various imaging modalities, however, positron emission tomography (PET) is a sensitive and non-invasive molecular imaging technology and this review will focus on PET molecular imaging of breast cancer, such as FDG-PET, FLT-PET, hormone receptor PET, and anti-HER2 PET. Keywords Breast cancer  PET  Molecular imaging  Molecular target therapy  HER2

Introduction Molecular imaging is defined by the Society of Nuclear Medicine as the visualization, characterization, and measurement of biological processes at the molecular and & Hiroaki Kurihara [email protected] 1

Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan

2

Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan

3

Department of Neurosurgery, National Cancer Center Hospital, Tokyo, Japan

4

Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan

5

Department of Clinical Pharmacology Group for Translational Research Support Core, National Cancer Center Research Institute, Tokyo, Japan

6

RIKEN Center for Molecular Imaging Science, Kobe, Hyogo, Japan

cellular levels in humans and other living systems [1]. The imaging of specific molecular targets that are associated with cancer will facilitate earlier diagnosis and better management of cancer patients. Molecular imaging of breast cancer can be performed with various imaging modalities, such as magnetic resonance imaging (MRI) using a contrast agent [2–6], positron emission tomography (PET) using a positron-emitting radionuclide [7–9], single photon emission computed tomography (SPECT) using a gamma-emitting radionuclide [10–12], optical imaging using a fluorescent dye [13, 14], or contrastenhanced ultrasound (US) [15, 16]. PET is a highly sensitive and non-invasive molecular imaging technology that has become an indispensable tool in cancer research, clinical trials, and medical practice. This review will focus on the use of PET molecular imaging of breast cancer. PET imaging using 18F-fluorodeoxyglucose (FDG) enables physicians to see where a tumor is located in the body. Using certain radiolabeled tracers, PET imaging is also expected to visualize the expression and activity of particular molecules, cells, and biological processes that influence the behavior of tumors and/or their responsiveness to therapeutic drugs [7–9, 17, 18]. To date, various tracers have been discovered and developed. Recent advances in molecular/cell biology have enabled the design of specific tracers to image molecular targets non-invasively using PET [19]. Here, we highlight recent advances in molecular imaging with PET for breast cancer. We begin with an overview of tracers and targets for molecular PET imaging of breast cancer. Next, we will focus on FDG-PET imaging, FLTPET imaging, hormone receptor PET imaging, and HER2 PET imaging. We will also describe other targets used for PET imaging of breast cancer briefly.

123

Breast Cancer

Tracers for and targets of breast cancer used in molecular PET imaging PET molecular imaging is based on the detection of radiolabeled tumor-specific tracers. Radioisotopes used for PET imaging (e.g., 18F, 11C, 15O, 13N, 64Cu, 124I, 89Zr) emit positrons. For molecular PET imaging of breast cancer, several tracers targeting certain tumor characteristics have been developed (Table 1). To target general phenomena, a certain tracer can be used to visualize either cellular glucose metabolism or DNA synthesis, processes that are both increased in tumor cells compared to normal cells [8, 17]. Most breast cancers express hormone receptors on the surface of tumor cells that provide targets of interest for imaging in these subsets of patients [24]. Moreover, other receptors present at the tumor cell membrane such as human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), insulinlike growth factor-1 receptor (IGF-1R), and plateletderived growth factor receptor b (PDGFR-b) may also be of interest for imaging. In addition, tumor cells secrete Table 1 Molecular targets/tumor characteristics and PET tracers for breast cancer Target/tumor characteristics

PET tracer

References

Glucose metabolism

FDG

[25–33]

DNA synthesis

FLT

[34–36]

ER

FES

[24, 41– 46]

18

[68]

18

[69]

18

[70]

18

[71]

F-fluorotamoxifen

PR

F-FMNP F-FENP F-FPTP

89

[58, 61]

64

[59, 60]

64

[72]

89

[74]

11

[75]

IGF-1R

89

[76]

VEGFR

64

[77]

89

[78]

64

[79]

18

[80]

HER2

Zr-labeled trastuzumab Cu-labeled trastuzumab

EGFR

Cu-labeled trastuzumab fragment 89 Zr-labeled cetuximab Zr-panitumumab C-PD153035 Zr-labeled R1507 Cu-DOTA-VEGF Zr-bevacizumab

Integrin

Cu-DOTA-RGD F-galacto-RGD

[73]

FDG 18F-fluorodeoxyglucose, FLT 18F-fluorothymidine, ER estrogen receptor, FES 16a-18F-fluoro-17b-estradiol, PR progesterone receptor, 18F-FMNP 21-18F-fluoro-16-a-methyl-19-norprogesterone, 18FFENP 21-18F-Fluoro-16a-ethyl-norprogesterone, 18F-FPTP 4-18Ffluoropropyl-tanaproget, HER2 human epidermal growth factor receptor 2, EGFR epidermal growth factor receptor, IGF-1R type 1 insulin-like growth factor receptor, VEGFR vascular endothelial growth factor receptor, RGD arginine–glycine–aspartic acid peptide

123

growth factors, such as vascular endothelial growth factor (VEGF) and transforming growth factor b (TGF-b) that could be tracer target candidates. Targets that are involved in hypoxia and angiogenesis such as VEGF-receptors, aVb3 integrin, fibronectin, and endostatin could also be used as tracer targets since both of these processes are important to tumor growth.

FDG-PET imaging The best studied and most widely used PET tracer for clinical purposes is FDG [25]. FDG-PET visualizes glucose metabolism, which is often increased in tumor cells compared to normal cells. Uptake by the tumor can usually be detected with a PET scanner 45–90 min after FDG injection (Fig. 1). The limited anatomical information obtained from FDG-PET alone has been improved by integrated PET/CT or a PET/MR scanner that provides fusion images of PET with CT or MRI [26–28]. FDG-PET has been found to be helpful for primary tumor detection and diagnosis, staging of locoregional and distant metastases, and in monitoring response to therapy. In a pre-operative setting, high FDG uptake by tumors was observed, particularly in ductal carcinomas [29]. FDG uptake itself, however, is not tumor specific, and it can be difficult to distinguish between malignant and benign breast cells. Moreover, falsepositive results can occur when activated inflammatory cells such as granulocytes and macrophages exist [30]. For early detection and diagnosis of breast cancer, the ability to detect non-palpable, small malignancies less than 1 cm in diameter is desirable; however, whole body FDG-PET has not been extensively used for this purpose because the spatial resolution of previous-generation clinical PET cameras is limited [8, 31]. High-resolution, high-sensitivity positron emission mammography (PEM) has been developed for detection and diagnosis of early-stage breast cancer (Fig. 2). PEM using FDG can detect primary breast cancer lesions as small as 3 mm in diameter [32, 33]. Its clinical utility for screening or as an adjunct to mammography has to be demonstrated by future studies.

FLT-PET imaging The pyrimidine analogue PET tracer 18F-fluoro-L-thymidine (FLT) has been developed to image increased cellular DNA synthesis. FLT-PET imaging shows normal physiological uptake in liver, bone marrow and the urinary tract (Fig. 3). In breast cancer patients, small studies have been reported. In a group of 12 breast cancer patients, FLT uptake was seen in 92 % of primary breast tumors and 87 % of axillary lymph node metastases [34]. Another

Breast Cancer Fig. 1 Typical images of whole body FDG-PET/CT showing breast cancer. Both Maximumintensity projection (MIP) image (a) and PET/CT fusion image (b) showed nodular lesion of 1.5 cm in diameter in lt breast of a 64-year-old woman. Following core needle biopsy revealed invasive ductal carcinoma

study compared FDG and FLT for monitoring and predicting tumor response to chemotherapy in 14 patients with primary or metastatic breast cancer. This study showed a strong correlation between the percentage decrease in FLT uptake within the tumor 2 weeks after initiating chemotherapy and the reduction in tumor size measured by CT at a later interval (average, 3.3 months). No correlation was found between FDG uptake changes over the first 2 weeks and these response measurements [35]. Based on these results, FLT-PET may help predict patient response to therapy [36].

Hormone receptor PET imaging Up to 70 % of breast cancer patients show tumors positive for hormone receptors. The majority of them are positive for estrogen receptor (ER) and more than 95 % of tumors positive for progesterone receptor (PR) are also ER-positive. Immunohistochemical determination of ER and PR expression at the time of primary diagnosis is a part of standard care. Anti-hormonal treatment strategies are based on hormone receptor expression, which is predictive of response to such treatment. Hormone receptor expression, however, can vary between primary and recurrent tumors in approximately 30 % of cases [37, 38]. The other study demonstrated that the discordance between the ER status of the primary tumor and the distant metastases was 41 % for bone marrow metastases, and 44 % for liver metastases [39]. In addition, anti-hormonal treatment induces ER loss in a number of patients with acquired hormonal resistance [40]. Thus, monitoring hormone receptor expression in

primary tumor and metastatic sites throughout the course of the disease might help a physician determine if anti-hormonal treatment should be used during the different phases of treatment. To monitor hormone receptor status in a noninvasive and repeated fashion, the PET tracer 16a-18Ffluoro-17b-estradiol (FES) was developed as a receptor ligand for ER [24]. Several studies using FES-PET have been performed in breast cancer patients and revealed the correlation of FES tumor uptake with immunohistochemical ER tumor density [41–44]. These studies suggested a potential role for FES-PET in assessing ER status, especially in patients with multiple tumors or tumors that are difficult to perform a biopsy on. Comparing the tumor uptakes of FES and FDG, as well as ER status among 43 patients showed an 88 % overall agreement between FES uptake and ER status. In contrast, there was no observed correlation either between FDG uptake and ER status or between FES and FDG uptake [41]. In 47 patients with immunohistochemically ER-positive tumors, FES-PET was performed at baseline either prior to or shortly after the initiation of anti-hormonal therapy. FES uptake by tumors at baseline was subsequently compared with the response determined by a combination of clinical assessment and modified RECIST criteria 6 months after the therapy. In patients with low FES uptake by tumor lesions at baseline (n = 15), no response was shown. In a group comprising 32 patients with a high FES tumor uptake at baseline, 34 % of the patients responded to anti-hormonal therapy [45, 46]. Therefore, FES-PET could not be recommended as a routine imaging technique for the workup of a breast cancer patient. It could, however, be useful for predicting response to anti-hormonal therapy.

123

Breast Cancer Fig. 2 Typical images of PEM showing breast cancer. a Mediolateral oblique (MLO) view of PEM; b cranio-caudal (CC) views of PEM; c MIP image of whole body PET/CT. Within lt breast of a 73-year-old woman, PEM images could visualize a lesion of 7 mm in diameter that whole-body PET/ CT scanner could hardly detect. Courtesy of Dr. Masami Kawamoto, Yuai-Clinic, Yokohama, Japan

HER2 PET imaging HER2, which is overexpressed in 25–30 % of breast cancer patients is involved in tumor cell survival, proliferation, maturation, metastasis, and angiogenesis [47, 48]. Trastuzumab, a humanized monoclonal antibody against HER2 is widely used and targeting HER2 with trastuzumab is a well-established therapeutic strategy for HER2-positive breast cancer [49–53]. HER2 expression is routinely determined using immunohistochemistry (IHC) or fluorescence in situ hybridization [54], however, technical problems can arise when lesions cannot be easily accessed by core needle biopsy [55]. In addition, HER2 expression can vary during the course of the disease [56] and even between tumor lesions in the same patient [57]. To overcome these problems, a novel non-invasive technique such as HER2 PET imaging is required to determine HER2 expression. So far, full-length HER2 monoclonal antibodies have been labeled with 124I, 86Y, 76Br, 89Zr, and 64Cu for HER2 PET imaging [58, 59]. The smaller HER2 targeting antibody fragments, proteins, and peptides have been labeled with 18F, 68Ga, 64Cu, 124I, and 76Br for HER2 PET imaging [58]. With use of 64Cu-trastuzumab PET imaging technique, for example, primary tumor lesions larger than 2 cm in diameter and metastatic brain lesions larger than 1 cm in diameter could be visualized [59, 60]. The typical 64Cu-DOTA-trastuzumab PET images within HER2-positive breast cancer patients are shown in Fig. 4. In human subjects, an autoradiogram of a frozen section prepared from the removed tumor specimen revealed high accumulation in the area where HER2-positive cells were

123

Fig. 3 Normal uptake of FLT-PET/CT. An MIP image of whole body FLT-PET/CT in a 64-year-old woman. FLT has been developed to visualize the activity of DNA synthesis that is increased in tumor cells compared to normal cells. Physiological high uptakes of FLT were also shown in normal organs of liver, bone marrow and the urinary tract

seen by IHC (Fig. 5), confirming the HER2 specificity of Cu-DOTA-trastuzumab PET imaging. Another HER2 PET imaging technique, 89Zrtrastuzumab PET imaging, also have succeeded to visualize

64

Breast Cancer Fig. 4 64Cu-DOTAtrastuzumab PET images of HER2-positive breast cancer and metastatic brain lesion. a Primary lesion of HER2positive breast cancer in a 69-year-old woman was visualized by whole body PET/ CT with use of 64Cu-DOTAtrastuzumab. Compared to the normal breast tissue, higher uptake was found within the tumor. Upper panel axial PET/ CT fusion image; lower panel magnified PET/CT fusion image (94). b Metastatic brain lesion in a 54-year-old woman having HER2-positive breast cancer was also visualized by 64CuDOTA-trastuzumab PET/CT (upper panel). A significant uptake was found in the areas corresponding to the brain metastatic lesion that was detected by MRI (lower panel)

HER2 positive tumor in a human. Due to the relatively longer half-life of Zr-89, 89Zn-trastuzumab provides clearer images; however, it induces higher radiation exposure [61]. In contrast, the shorter half-life of 64Cu induces lower radiation exposure but provides images with non-specific activity in the blood [59, 62]. Recently, several novel HER2 inhibitors have been developed and approved, including lapatinib, pertuzumab, and trastuzumab emtansine (T-DM1). Monitoring changes in HER2 expression at tumor sites may help physicians to determine which kind of HER2 inhibitors should be used during the different phases of treatment or if other nonHER2 inhibitors should be used instead. Preclinical results with HER2 imaging are abundant and promising; however, clinical data are still limited. The findings with clinical HER2 PET imaging may support the further development and exploration of the potential of this technique.

Other targets for PET imaging of breast cancer Triple-negative breast cancer, a type of breast cancer that is immunohistochemically negative for ER, PR, and HER2, typically has a poor prognosis [63–65]. Much research is

ongoing to identify the biological processes and targets that drive the behaviors of triple-negative breast cancer. Molecular imaging of these targets may aid target identification, drug development, and in predicting and evaluating response to therapy. For example, EGFR overexpression is seen in 57 % of triple-negative breast cancers. Several tracers have been developed for EGFR imaging, including radiolabeled EGFR tyrosine kinase inhibitors, epidermal growth factor, and EGFR antibodies [66]. The EGFR-directed monoclonal antibody cetuximab has been successfully radiolabeled with 64Cu at our institute [67].

Future direction of molecular imaging with PET Molecular imaging of breast cancer is not commonly used in daily clinical practice; however, its applications are expected and promised for improving breast cancer management. We have reported results made with sensitive PET molecular imaging techniques but these techniques still have limitations, including feasible capacity and spatial resolution, which impede early tumor detection. The resolutions of PEM and the latest generation of PET/CT

123

Breast Cancer Fig. 5 HER2 specificity of 64 Cu-DOTA-trastuzumab. a HER2-positive breast cancer in a 55-year-old woman; b metastatic brain tumor in a 61-year-old woman having HER2-positive breast cancer. Frozen section specimens removed from the lesions showing high uptake on 64CuDOTA-trastuzumab PET/CT images (upper panels) were used for IHC (middle panels) and autoradiogram (lower panels). High accumulations of autoradiogram (red arrows) were observed in the area where HER2-positive cells (black arrows) were seen by IHC, confirming the HER2 specificity of 64Cu-DOTA-trastuzumab PET imaging in human subjects

and PET/MR scanners have improved and may lead to improved anatomical interpretation of the images. In addition to the innovations in imaging modalities, the indirect combination of PET imaging and other novel modalities such as optical imaging could serve other purposes. The molecular target and its tracer for the novel modality still have to be validated for clinical use. In a combined setting, more sensitive PET imaging can be used for the target and tracer validation. Thus, molecular imaging with PET could prove to be a powerful tool for novel tracer discovery and development. Another potential application of molecular PET imaging could lie in clinical trials. In combination with pharmacokinetic and pharmacodynamic studies, molecular imaging studies in a clinical trial would provide a more powerful approach to explore appropriate ways of using molecularly targeted drugs. Moreover, during preclinical phase or phase 0 trials, molecular imaging with

123

PET can provide proof of concept and further the development of new therapeutic agents. Acknowledgments The authors thank Dr. Masami Kawamoto for providing appropriate PEM images. The authors thank Mr. Takayuki Namma and the staff of Sumitomo Heavy Industries for supporting 64 Cu-DOTA-trastuzumab/cetuximab production, and Mr. Akira Hirayama and the staff of GE Healthcare for optimizing PET/CT scan. We also thank Ms. Riako Onoe for secretarial support during this study. Conflict of interest of interest.

The authors declare that they have no conflict

References 1. Mankoff DA. A definition of molecular imaging. J Nucl Med. 2007;48:18N–21N.

Breast Cancer 2. Barrett T, Brechbiel M, Bernardo M, Choyke PL. MRI of tumor angiogenesis. J Magn Reson Imaging. 2007;26:235–49. 3. Padhani AR, Leach MO. Antivascular cancer treatments: functional assessments by dynamic contrast-enhanced magnetic resonance imaging. Abdom Imaging. 2005;30:324–41. 4. Haddadin IS, McIntosh A, Meisamy S, Corum C, Styczynski Snyder AL, Powell NJ, et al. Metabolite quantification and highfield MRS in breast cancer. NMR Biomed. 2009;22:65–76. 5. Meisamy S, Bolan PJ, Baker EH, Bliss RL, Gulbahce E, Everson LI, et al. Neoadjuvant chemotherapy of locally advanced breast cancer: predicting response with in vivo (1)H MR spectroscopy— a pilot study at 4 T. Radiology. 2004;233:424–31. 6. Lehman CD, Smith RA. The role of MRI in breast cancer screening. J Natl Compr Cancer Netw. 2009;7:1109–15. 7. Mankoff DA, Eary JF, Link JM, Muzi M, Rajendran JG, Spence AM, et al. Tumor-specific positron emission tomography imaging in patients: [18F] fluorodeoxyglucose and beyond. Clin Cancer Res. 2007;13:3460–9. 8. Rosen EL, Eubank WB, Mankoff DA. FDG PET, PET/CT, and breast cancer imaging. Radiographics. 2007;27:S215–29. 9. Partridge SC, Vanantwerp RK, Doot RK, Chai X, Kurland BF, Eby PR, et al. Association between serial dynamic contrast-enhanced MRI and dynamic 18F-FDG PET measures in patients undergoing neoadjuvant chemotherapy for locally advanced breast cancer. J Magn Reson Imaging. 2010;32:1124–31. 10. Benard F, Turcotte E. Imaging in breast cancer: single-photon computed tomography and positron-emission tomography. Breast Cancer Res. 2005;7:153–62. 11. Brem RF, Fishman M, Rapelyea JA. Detection of ductal carcinoma in situ with mammography, breast specific gamma imaging, and magnetic resonance imaging: a comparative study. Acad Radiol. 2007;14:945–50. 12. Brem RF, Floerke AC, Rapelyea JA, Teal C, Kelly T, Mathur V. Breast-specific gamma imaging as an adjunct imaging modality for the diagnosis of breast cancer. Radiology. 2008;247:651–7. 13. Tromberg BJ, Pogue BW, Paulsen KD, Yodh AG, Boas DA, Cerussi AE. Assessing the future of diffuse optical imaging technologies for breast cancer management. Med Phys. 2008;35:2443–51. 14. Tromberg BJ, Cerussi A, Shah N, Compton M, Durkin A, Hsiang D, et al. Imaging in breast cancer: diffuse optics in breast cancer: detecting tumors in pre-menopausal women and monitoring neoadjuvant chemotherapy. Breast Cancer Res. 2005;7:279–85. 15. Sorace AG, Saini R, Mahoney M, Hoyt K. Molecular ultrasound imaging using a targeted contrast agent for assessing early tumor response to antiangiogenic therapy. J Ultrasound Med. 2012;31:1543–50. 16. Eisenbrey JR, Forsberg F. Contrast-enhanced ultrasound for molecular imaging of angiogenesis. Eur J Nucl Med Mol Imaging. 2010;37:S138–46. 17. Kenny L, Coombes RC, Vigushin DM, Al-Nahhas A, Shousha S, Aboagye EO. Imaging early changes in proliferation at 1 week post chemotherapy: a pilot study in breast cancer patients with 30 deoxy-30 -[18F]- fluorothymidine positron emission tomography. Eur J Nucl Med Mol Imaging. 2007;34:1339–47. 18. Kenny LM, Contractor KB, Stebbing J, Al-Nahhas A, Palmieri C, Shousha S, et al. Altered tissue 30 -deoxy-30 -[18F]fluorothymidine pharmacokinetics in human breast cancer following capecitabine treatment detected by positron emission tomography. Clin Cancer Res. 2009;15:6649–57. 19. Phelps ME, Hoffman EJ, Mullani NA, Ter-Pogossian MM. Application of annihilation coincidence detection to transaxial reconstruction tomography. J Nucl Med. 1975;16:210–24. 20. Groheux D, Espie´ M, Giacchetti S, Hindie´ E. Performance of FDG PET/CT in the clinical management of breast cancer. Radiology. 2013;266:388–405.

21. Brindle KM. New approaches for imaging tumour responses to treatment. Nat Rev Cancer. 2008;8:94–107. 22. Quon A, Gambhir SS. FDG-PET and beyond: molecular breast cancer imaging. J Clin Oncol. 2005;23:1664–73. 23. Neves AA, Brindle KM. Assessing responses to cancer therapy using molecular imaging. Biochim Biophys Acta. 2006;1766:242–61. 24. Yoo J, Dence CS, Sharp TL, Katzenellenbogen JA, Welch MJ. Synthesis of an estrogen receptor beta-selective radioligand: 5-[18F]fluoro-(2R,3S)-2,3- bis(4-hydroxyphenyl)pentanenitrile and comparison of in vivo distribution with 16alpha-[18F]fluoro17beta-estradiol. J Med Chem. 2005;48:6366–78. 25. Fletcher JW, Djulbegovic B, Soares HP, Siegel BA, Lowe VJ, Lyman GH, et al. Recommendations on the use of 18F-FDG PET in oncology. J Nucl Med. 2008;49:480–508. 26. Endo K, Oriuchi N, Higuchi T, Iida Y, Hanaoka H, Miyakubo M, et al. PET and PET/CT using 18F-FDG in the diagnosis and management of cancer patients. Int J Clin Oncol. 2006;11:286–96. 27. Hayashi M, Murakami K, Oyama T, Domeki Y, Hagiwara S, Katsumata D, et al. PET/CT supports breast cancer diagnosis and treatment. Breast Cancer. 2008;15:224–30. 28. Pace L, Nicolai E, Luongo A, Aiello M, Catalano OA, Soricelli A, et al. Comparison of whole-body PET/CT and PET/MRI in breast cancer patients: lesion detection and quantitation of 18Fdeoxyglucose uptake in lesions and in normal organ tissues. Eur J Radiol. 2014;83:289–96. 29. Buck A, Schirrmeister H, Kuhn T, Shen C, Kalker T, Kotzerke J, et al. FDG uptake in breast cancer: correlation with biological and clinical prognostic parameters. Eur J Nucl Med Mol Imaging. 2002;29:1317–23. 30. Kubota K, Furumoto S, Iwata R, Fukuda H, Kawamura K, Ishiwata K. Comparison of 18F-fluoromethylcholine and 2-deoxy-Dglucose in the distribution of tumor and inflammation. Ann Nucl Med. 2006;20:527–33. 31. Samson DJ, Flamm CR, Pisano ED, Aronson N. Should FDG PET be used to decide whether a patient with an abnormal mammogram or breast finding at physical examination should undergo biopsy? Acad Radiol. 2002;9:773–83. 32. Kalinyak JE, Berg WA, Schilling K, Madsen KS, Narayanan D, Tartar M. Breast cancer detection using high-resolution breast PET compared to whole-body PET or PET/CT. Eur J Nucl Med Mol Imaging. 2014;41:260–75. 33. Tafra L, Cheng Z, Uddo J, Lobrano MB, Stein W, Berg WA, et al. Pilot clinical trial of 18F-fluorodeoxyglucose positron-emission mammography in the surgical management of breast cancer. Am J Surg. 2005;190:628–32. 34. Smyczek-Gargya B, Fersis N, Dittmann H, Vogel U, Reischl G, Machulla HJ, et al. PET with [18F]fluorothymidine for imaging of primary breast cancer: a pilot study. Eur J Nucl Med Mol Imaging. 2004;31:720–4. 35. Pio BS, Park CK, Pietras R, Hsueh WA, Satyamurthy N, Pegram MD, et al. Usefulness of 3-[F-18]fluoro-3-deoxythymidine with positron emission tomography in predicting breast cancer response to therapy. Mol Imaging Biol. 2006;8:36–42. 36. Salskov A, Tammisetti VS, Grierson J, Vesselle H. FLT: measuring tumor cell proliferation in vivo with positron emission tomography and 3-deoxy-3-[18F]fluorothymidine. Semin Nucl Med. 2007;37:429–39. 37. Spataro V, Price K, Goldhirsch A, Cavalli F, Simoncini E, Castiglione M, et al. Sequential estrogen receptor determinations from primary breast cancer and at relapse: prognostic and therapeutic relevance. The International Breast Cancer Study Group (formerly Ludwig Group). Ann Oncol. 1992;3:733–40. 38. Kuukasjarvi T, Kononen J, Helin H, Holli K, Isola J. Loss of estrogen receptor in recurrent breast cancer is associated with

123

Breast Cancer

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

poor response to endocrine therapy. J Clin Oncol. 1996;14:2584–9. Brunn RB, Kamby C. Immunohistochemical detection of estrogen receptors in paraffin sections from primary and metastatic breast cancer. Pathol Res Pract. 1989;185:856–9. Normanno N, Di Maio M, De Maio E, De Luca A, de Matteis A, Giordano A, et al. Mechanisms of endocrine resistance and novel therapeutic strategies in breast cancer. Endocr Relat Cancer. 2005;12:721–47. Dehdashti F, Mortimer JE, Siegel BA, Griffeth LK, Bonasera TJ, Fusselman MJ, et al. Positron tomographic assessment of estrogen receptors in breast cancer: comparison with FDG-PET and in vitro receptor assays. J Nucl Med. 1995;36:1766–74. Mintun MA, Welch MJ, Siegel BA, Mathias CJ, Brodack JW, McGuire AH, et al. Breast cancer: PET imaging of estrogen receptors. Radiology. 1988;169:45–8. Peterson LM, Mankoff DA, Lawton T, Yagle K, Schubert EK, Stekhova S, et al. Quantitative imaging of estrogen receptor expression in breast cancer with PET and 18F-fluoroestradiol. J Nucl Med. 2008;49:367–74. Mortimer JE, Dehdashti F, Siegel BA, Katzenellenbogen JA, Fracasso P, Welch MJ. Positron emission tomography with 2-[18F]fluoro-2-deoxy-D-glucose and 16alpha-[18F]fluoro-17beta-estradiol in breast cancer: correlation with estrogen receptor status and response to systemic therapy. Clin Cancer Res. 1996;2:933–9. Linden HM, Stekhova SA, Link JM, Gralow JR, Livingston RB, Ellis GK, et al. Quantitative fluoroestradiol positron emission tomography imaging predicts response to endocrine treatment in breast cancer. J Clin Oncol. 2006;24:2793–9. Gemignani ML, Patil S, Seshan VE, Sampson M, Humm JL, Lewis JS, et al. Feasibility and predictability of perioperative PET and estrogen receptor ligand in patients with invasive breast cancer. J Nucl Med. 2013;54:1697–702. Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 2007;26:6469–87. Hayes DF, Thor AD, Dressler LG, Weaver D, Edgerton S, Cowan D, et al. HER2 and response to paclitaxel in node-positive breast cancer. N Engl J Med. 2007;357:1496–506. Buzdar AU, Ibrahim NK, Francis D, Boose DJ, Thomas ES, Theriault RL, et al. Significantly higher pathologic complete remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin chemotherapy: results of a randomized trial in human epidermal growth factor receptor 2–positive operable breast cancer. J Clin Oncol. 2005;23:3676–85. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72. Romond EH, Perez EA, Bryant J, Sumab VJ, Geyer CE Jr, Davidson NE, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–84. 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;344:783–92. Tripathy D, Slamon DJ, Cobleigh M, Arnord A, Saleh M, Mortimer JE, et al. Safety of treatment of metastatic breast cancer with trastuzumab beyond disease progression. J Clin Oncol. 2004;22:1063–70. Sauter G, Lee J, Bartlett JM, Slamon DJ, Press MF. Guidelines for human epidermal growth factor receptor 2 testing: biologic and methodologic considerations. J Clin Oncol. 2009;27:1323–33.

123

55. Lebeau A, Turzynski A, Braun S, Behrhof W, Fleige B, Schmitt WD, et al. Reliability of human epidermal growth factor receptor 2 immunohistochemistry in breast core needle biopsies. J Clin Oncol. 2010;28:3264–70. 56. Xiao C, Gong Y, Han EY, Gonzalez-Angulo AM, Sneige N. Stability of HER2-positive status in breast carcinoma: a comparison between primary and paired metastatic tumors with regard to the possible impact of intervening trastuzumab treatment. Ann Oncol. 2011;22:1547–53. 57. Houssami N, Macaskill P, Balleine RL, Bilous M, Pegram MD. HER2 discordance between primary breast cancer and its paired metastasis: tumor biology or test artifact? Insights through metaanalysis. Breast Cancer Res Treat. 2011;129:659–74. 58. Dijkers EC, de Vries EG, Kosterink JG, Brouwers AH, Lub-de Hooge MN. Immunoscintigraphy as potential tool in the clinical evaluation of HER2/neu targeted therapy. Curr Pharm Des. 2008;14:3348–62. 59. Tamura K, Kurihara H, Yonemori K, Tsuda H, Suzuki J, Kono Y, et al. 64Cu-DOTA-Trastuzumab PET Imaging in Patients with HER2-Positive Breast Cancer. J Nucl Med. 2013;54:1869–75. 60. Kurihara H, Hamada A, Yoshida M, Shimma S, Hashimoto J, Yonemori K, et al. 64Cu-DOTA-trastuzumab PET imaging and HER2-specificity of brain metastases in HER2-positive breast cancer patients. EJNMMI Res. 2015, (in print). 61. Dijkers EC, Oude Munnink TH, Kosterink JG, Brouwers AH, Jager PL, de Jong JR, et al. Biodistribution of 89Zr-trastuzumab and PET imaging of HER2-positive lesions in patients with metastatic breast cancer. Clin Pharmacol Ther. 2010;87:586–92. 62. Mortimer JE, Bading JR, Colcher DM, Conti PS, Frankel PH, Carroll MI, et al. Functional imaging of human epidermal growth factor receptor 2-positive metastatic breast cancer using (64)CuDOTA-trastuzumab PET. J Nucl Med. 2014;55:23–9. 63. Schneider BP, Winer EP, Foulkes WD, Garber J, Perou CM, Richardson A, et al. Triple-negative breast cancer: risk factors to potential targets. Clin Cancer Res. 2008;14:8010–8. 64. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer. 2007;109:1721–8. 65. Mersin H, Yildirim E, Berberoglu U, Gulben K. The prognostic importance of triple negative breast carcinoma. Breast. 2008;17:341–6. 66. Cai W, Niu G, Chen X. Multimodality imaging of the HERkinase axis in cancer. Eur J Nucl Med Mol Imaging. 2008;35:186–208. 67. Honda N, Kurihara H, Takahashi K, Tazawa S, Tamura K, Zochi R, et al. Radiosynthesis of 64Cu-labelled cetuximab for clinical use. J Nucl Med. 2013;54:s1193. 68. Inoue T, Kim EE, Wallace S, Yang DJ, Wong FC, Bassa P, et al. Positron emission tomography using [18F]fluorotamoxifen to evaluate therapeutic responses in patients with breast cancer: preliminary study. Cancer Biother Radiopharm. 1996;11:235–45. 69. Verhagen A, Luurtsema G, Pesser JW, de Groot TJ, Wouda S, Oosterhuis JW, et al. Preclinical evaluation of a positron emitting progestin ([18F]fluoro-16a-methyl-19-norprogesterone) for imaging progesterone receptor positive tumours with positron emission tomography. Cancer Lett. 1991;59:125–32. 70. Dehdashti F, McGuire AH, Van Brocklin HF, Siegel BA, Andriole DO, Griffeth LK, et al. Assessment of 21-[18F]fluoro-16aethyl-19-norprogesterone as a positron-emitting radiopharmaceutical for the detection of progestin receptors in human breast carcinomas. J Nucl Med. 1991;32:1532–7. 71. Lee JH, Zhou HB, Dence CS, Carlson KE, Welch MJ, Katzenellenbogen JA. Development of [F-18]fluorine substituted

Breast Cancer

72.

73.

74.

75.

76.

tanaproget as a progesterone receptor imaging agent for positron emission tomography. Bioconjug Chem. 2010;21:1096–104. Smith-Jones PM, Solit DB, Akhurst T, Afroze F, Rosen N, Larson SM. Imaging the pharmacodynamics of HER2 degradation in response to Hsp90 inhibitors. Nat Biotechnol. 2004;22:701–6. Makris NE, Boellaard R, van Lingen A, Lammertsma AA, van Dongen GA, Verheul HM, et al. PET/CT-derived whole-body and bone marrow dosimetry of 89Zr-cetuximab. J Nucl Med. 2015;56:249–54. Bhattacharyya S, Kurdziel K, Wei L, Riffle L, Kaur G, Hill GC, et al. Zirconium-89 labeled panitumumab: a potential immunoPET probe for HER1-expressing carcinomas. Nucl Med Biol. 2013;40:451–7. Wang H, Yu J, Yang G, Song X, Sun X, Zhao S, et al. Assessment of 11C-labeled-4-N-(3-bromoanilino)-6, 7-dimethoxyquinazoline as a positron emission tomography agent to monitor epidermal growth factor receptor expression. Cancer Sci. 2007;98:1413–6. Heskamp S, Van Laarhoven HWM, Molkenboer-Kuenen JD, Franssen GM, Versleijen-Jonkers YM, Oyen WJ, et al.

77.

78.

79.

80.

ImmunoSPECT and immunoPET of IGF-1R expression with the radiolabeled antibody R1507 in a triple-negative breast cancer model. J Nucl Med. 2010;51:1565–72. Wang H, Cai W, Chen K, Li ZB, Kashefi A, He L, et al. A new PET tracer specific for vascular endothelial growth factor receptor 2. Eur J Nucl Med Mol Imaging. 2007;34:2001–10. Gaykema SB, Brouwers AH, Lub-de Hooge MN, Pleijhuis RG, Timmer-Bosscha H, Pot L, et al. 89Zr-bevacizumab PET imaging in primary breast cancer. J Nucl Med. 2013;54:1014–8. Chen X, Park R, Tohme M, Shahinian AH, Bading JR, Conti PS. MicroPET and autoradiographic imaging of breast cancer av-Integrin Expression Using 18F- and 64Cu-Labeled RGD Peptide. Bioconjug Chem. 2004;15:41–9. Beer AJ, Niemeyer M, Carlsen J, Sarbia M, Nahrig J, Watzlowik P, et al. Patterns of avb3 expression in primary and metastatic human breast cancer as shown by 18F-galacto-RGD PET. J Nucl Med. 2008;49:255–9.

123

Molecular imaging using PET for breast cancer.

Molecular imaging can visualize the biological processes at the molecular and cellular levels in vivo using certain tracers for specific molecular tar...
6MB Sizes 1 Downloads 18 Views