Access the full text.
Sign up today, get DeepDyve free for 14 days.
S. Nik-Zainal, P. Loo, D. Wedge, L. Alexandrov, C. Greenman, K. Lau, K. Raine, David Jones, J. Marshall, Manasa Ramakrishna, A. Shlien, S. Cooke, Jonathan Hinton, A. Menzies, L. Stebbings, Catherine Leroy, Mingming Jia, Richard Rance, L. Mudie, Stephen Gamble, P. Stephens, Stuart McLaren, P. Tarpey, E. Papaemmanuil, H. Davies, I. Varela, D. McBride, G. Bignell, Kenric Leung, Adam Butler, J. Teague, Sancha Martin, G. Jönsson, O. Mariani, S. Boyault, P. Miron, Aquila Fatima, A. Langerød, S. Aparicio, A. Tutt, A. Sieuwerts, Å. Borg, G. Thomas, A. Salomon, A. Richardson, A. Børresen-Dale, P. Futreal, Michael Stratton, P. Campbell (2012)
The Life History of 21 Breast CancersCell, 149
M. Ellis, L. Ding, D. Shen, Jingqin Luo, V. Suman, J. Wallis, B. Tine, J. Hoog, Reece Goiffon, Theodore Goldstein, S. Ng, Li Lin, R. Crowder, J. Snider, K. Ballman, J. Weber, Ken Chen, D. Koboldt, C. Kandoth, W. Schierding, Joshua McMichael, Christopher Miller, Charles Lu, C. Harris, M. McLellan, M. Wendl, K. Deschryver, D. Allred, L. Esserman, G. Unzeitig, J. Margenthaler, G. Babiera, P. Marcom, J. Guenther, M. Leitch, K. Hunt, J. Olson, Y. Tao, C. Maher, L. Fulton, R. Fulton, M. Harrison, Benjamin Oberkfell, Feiyu Du, R. Demeter, T. Vickery, A. Elhammali, H. Piwnica-Worms, Sandra McDonald, M. Watson, D. Dooling, D. Ota, Li-Wei Chang, R. Bose, T. Ley, D. Piwnica-Worms, Joshua Stuart, R. Wilson, E. Mardis (2012)
Whole Genome Analysis Informs Breast Cancer Response to Aromatase InhibitionNature, 486
N. Wolmark, Jiping Wang, E. Mamounas, J. Bryant, B. Fisher (2001)
Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18.Journal of the National Cancer Institute. Monographs, 30
E. Amir, M. Clemons, C. Purdie, N. Miller, P. Quinlan, W. Geddie, R. Coleman, O. Freedman, L. Jordan, A. Thompson (2012)
Tissue confirmation of disease recurrence in breast cancer patients: pooled analysis of multi-centre, multi-disciplinary prospective studies.Cancer treatment reviews, 38 6
V. Amoroso, D. Generali, T. Buchholz, M. Cristofanilli, R. Pedersini, G. Curigliano, M. Daidone, S. Cosimo, M. Dowsett, S. Fox, A. Harris, A. Makris, L. Vassalli, A. Ravelli, M. Cappelletti, C. Hatzis, C. Hudis, P. Pedrazzoli, A. Sapino, V. Semiglazov, G. Minckwitz, E. Simoncini, Michael Jacobs, P. Barry, T. Kühn, S. Darby, K. Hermelink, F. Symmans, A. Gennari, G. Schiavon, L. Dogliotti, A. Berruti, A. Bottini (2011)
International Expert Consensus on Primary Systemic Therapy in the Management of Early Breast Cancer: Highlights of the Fifth Symposium on Primary Systemic Therapy in the Management of Operable Breast Cancer, Cremona, Italy (2013).Journal of the National Cancer Institute. Monographs, 2015 51
A. Ring, A. Webb, S. Ashley, W. Allum, S. Ebbs, G. Gui, N. Sacks, G. Walsh, I. Smith (2003)
Is surgery necessary after complete clinical remission following neoadjuvant chemotherapy for early breast cancer?Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 21 24
Kenneth Hess, K. Anderson, W. Symmans, V. Valero, N. Ibrahim, Jaime Mejia, D. Booser, Richard Theriault, A. Buzdar, Peter Dempsey, R. Rouzier, N. Sneige, Jeffrey Ross, T. Vidaurre, Henry Gómez, G. Hortobagyi, L. Pusztai (2006)
Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide in breast cancer.Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 24 26
M. Dowsett, I. Smith, S. Ebbs, J. Dixon, A. Skene, R. A’Hern, J. Salter, S. Detre, M. Hills, G. Walsh (2007)
Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer.Journal of the National Cancer Institute, 99 2
N. Navin, A. Krasnitz, L. Rodgers, K. Cook, J. Meth, J. Kendall, M. Riggs, Y. Eberling, J. Troge, V. Grubor, D. Levy, P. Lundin, S. Månér, A. Zetterberg, J. Hicks, M. Wigler (2010)
Inferring tumor progression from genomic heterogeneity.Genome research, 20 1
D. Gagliato, A. González-Angulo, X. Lei, S. Giordano, R. Theriault, V. Valero, G. Hortobagyi, M. Gregor (2013)
Impact of delaying initiation of adjuvant chemotherapy in breast cancer patients.Journal of Clinical Oncology, 31
Sarah Aitken, J. Thomas, S. Langdon, D. Harrison, D. Faratian (2010)
Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases.Annals of oncology : official journal of the European Society for Medical Oncology, 21 6
L. Lindström, E. Karlsson, U. Wilking, U. Johansson, J. Hartman, E. Lidbrink, T. Hatschek, L. Skoog, J. Bergh (2012)
Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression.Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 30 21
Jenny Chang, A. Makris, M. Gutiérrez, S. Hilsenbeck, J. Hackett, Jennie Jeong, Mei-lan Liu, J. Baker, K. Clark-Langone, F. Baehner, K. Sexton, S. Mohsin, T. Gray, L. Álvarez, G. Chamness, C. Osborne, S. Shak (2008)
Gene expression patterns in formalin-fixed, paraffin-embedded core biopsies predict docetaxel chemosensitivity in breast cancer patientsBreast Cancer Research and Treatment, 108
L. Gianni, M. Zambetti, K. Clark, J. Baker, M. Cronin, J. Wu, G. Mariani, Jaime Rodriguez, M. Carcangiu, D. Watson, P. Valagussa, R. Rouzier, W. Symmans, J. Ross, G. Hortobagyi, L. Pusztai, S. Shak (2004)
Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer.Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 23 29
G. Curigliano, V. Bagnardi, G. Viale, L. Fumagalli, N. Rotmensz, G. Aurilio, M. Locatelli, G. Pruneri, S. Giudici, M. Bellomi, P. Vigna, L. Monfardini, F. Orsi, F. Nolè, E. Munzone, A. Goldhirsch (2011)
Should liver metastases of breast cancer be biopsied to improve treatment choice?Annals of oncology : official journal of the European Society for Medical Oncology, 22 10
M. Marinovich, F. Sardanelli, S. Ciatto, E. Mamounas, Meagan Brennan, P. Macaskill, L. Irwig, G. Minckwitz, N. Houssami (2012)
Early prediction of pathologic response to neoadjuvant therapy in breast cancer: systematic review of the accuracy of MRI.Breast, 21 5
E. Mittendorf, T. Buchholz, S. Tucker, F. Meric-Bernstam, H. Kuerer, A. González-Angulo, I. Bedrosian, G. Babiera, K. Hoffman, M. Yi, M. Ross, G. Hortobagyi, K. Hunt (2013)
Impact of Chemotherapy Sequencing on Local-Regional Failure Risk in Breast Cancer Patients Undergoing Breast-Conserving TherapyAnnals of Surgery, 257
N. Houssami, P. Macaskill, G. Minckwitz, M. Marinovich, E. Mamounas (2012)
Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy.European journal of cancer, 48 18
A. Caudle, T. Yu, S. Tucker, I. Bedrosian, J. Litton, A. González-Angulo, K. Hoffman, F. Meric-Bernstam, K. Hunt, T. Buchholz, E. Mittendorf (2012)
Local-regional control according to surrogate markers of breast cancer subtypes and response to neoadjuvant chemotherapy in breast cancer patients undergoing breast conserving therapyBreast Cancer Research : BCR, 14
W. Symmans, F. Peintinger, C. Hatzis, Radhika Rajan, H. Kuerer, V. Valero, L. Assad, A. Poniecka, B. Hennessy, Marjorie Green, A. Buzdar, S. Singletary, G. Hortobagyi, L. Pusztai (2007)
Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy.Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 25 28
P. Ellis, S. Ashley, G. Walsh, S. Ebbs, M. Baum, N. Sacks, I. Smith (1997)
0-38. Identification of clinical factors predicting outcome following primary chemotherapy for operable breast cancerThe Breast, 6
T. Barrett, D. Bowden, D. Greenberg, C. Brown, G. Wishart, P. Britton (2009)
Radiological staging in breast cancer: which asymptomatic patients to image and howBritish Journal of Cancer, 101
T. Yap, M. Gerlinger, M. Gerlinger, P. Futreal, L. Pusztai, C. Swanton, C. Swanton (2012)
Intratumor Heterogeneity: Seeing the Wood for the TreesScience Translational Medicine, 4
L. Ding, M. Ellis, Shunqiang Li, D. Larson, Ken Chen, J. Wallis, C. Harris, M. McLellan, R. Fulton, L. Fulton, R. Abbott, J. Hoog, D. Dooling, D. Koboldt, Heather Schmidt, Joelle Kalicki, Qunyuan Zhang, Lei Chen, Ling Lin, M. Wendl, Joshua McMichael, V. Magrini, L. Cook, S. McGrath, T. Vickery, Elizabeth Appelbaum, K. Deschryver, S. Davies, Therese Guintoli, Li Lin, R. Crowder, Y. Tao, J. Snider, Scott Smith, Adam Dukes, Gabriel Sanderson, C. Pohl, K. Delehaunty, C. Fronick, Kimberley Pape, Jerry Reed, J. Robinson, J. Hodges, W. Schierding, N. Dees, D. Shen, D. Locke, Madeline Wiechert, James Eldred, Joshua Peck, Benjamin Oberkfell, Justin Lolofie, Feiyu Du, Amy Hawkins, M. O'Laughlin, Kelly Bernard, M. Cunningham, Glendoria Elliott, M. Mason, Dominic Thompson, J. Ivanovich, P. Goodfellow, C. Perou, G. Weinstock, R. Aft, M. Watson, T. Ley, R. Wilson, E. Mardis (2010)
Genome Remodeling in a Basal-like Breast Cancer Metastasis and XenograftNature, 464
J. Boughey, F. Peintinger, F. Meric-Bernstam, Allison Perry, K. Hunt, G. Babiera, S. Singletary, I. Bedrosian, A. Lucci, A. Buzdar, L. Pusztai, H. Kuerer (2006)
Impact of Preoperative Versus Postoperative Chemotherapy on the Extent and Number of Surgical Procedures in Patients Treated in Randomized Clinical Trials for Breast CancerAnnals of Surgery, 244
N. Turner, J. Reis-Filho (2012)
Genetic heterogeneity and cancer drug resistance.The Lancet. Oncology, 13 4
D. Yardley, N. Peacock, C. Hendricks, S. Huh, S. Ketchum, C. Chao, C. Yoshizawa, Iii Burris, J. Hainsworth (2011)
P5-13-09: Correlation of Oncotype DX Recurrence Scores with Pathologic Response Following Neoadjuvant Ixabepilone and Cyclophosphamide in Patients with HER2−Negative Breast Cancer: A Sarah Cannon Research Institute Phase II Trial.Cancer Research, 71
N. Navin, J. Kendall, J. Troge, Peter Andrews, L. Rodgers, J. McIndoo, K. Cook, A. Stepansky, D. Levy, D. Esposito, L. Muthuswamy, A. Krasnitz, W. McCombie, J. Hicks, M. Wigler (2011)
Tumour evolution inferred by single-cell sequencingNature, 472
I. Komenaka, M. Hibbard, Chiu-Hsieh Hsu, B. Low, J. Salganick, Marcia Bouton, C. Jha (2011)
Preoperative chemotherapy for operable breast cancer improves surgical outcomes in the community hospital setting.The oncologist, 16 6
T. Kuehn, I. Bauerfeind, T. Fehm, B. Fleige, M. Hausschild, G. Helms, A. Lebeau, C. Liedtke, G. Minckwitz, V. Nekljudova, S. Schmatloch, P. Schrenk, A. Staebler, M. Untch (2013)
Sentinel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study.The Lancet. Oncology, 14 7
Wennuan Liu, Sari Laitinen, Sofia Khan, M. Vihinen, J. Kowalski, Guoqiang Yu, Li Chen, C. Ewing, M. Eisenberger, M. Carducci, W. Nelson, S. Yegnasubramanian, Jun Luo, Yue Wang, Jianfeng Xu, W. Isaacs, T. Visakorpi, Steven Bova (2009)
Copy Number Analysis Indicates Monoclonal Origin of Lethal Metastatic Prostate CancerNature medicine, 15
R. Rouzier, C. Perou, W. Symmans, N. Ibrahim, M. Cristofanilli, K. Anderson, K. Hess, J. Stec, J. Stec, M. Ayers, P. Wagner, P. Morandi, Chang Fan, Islam Rabiul, J. Ross, G. Hortobagyi, L. Pusztai (2005)
Breast Cancer Molecular Subtypes Respond Differently to Preoperative ChemotherapyClinical Cancer Research, 11
A. Lowery, M. Kell, R. Glynn, M. Kerin, K. Sweeney (2012)
Locoregional recurrence after breast cancer surgery: a systematic review by receptor phenotypeBreast Cancer Research and Treatment, 133
Ana Lourenco, M. Mainiero, E. Lazarus, D. Giri, B. Schepps (2007)
Stereotactic breast biopsy: comparison of histologic underestimation rates with 11- and 9-gauge vacuum-assisted breast biopsy.AJR. American journal of roentgenology, 189 5
C. Curtis, Sohrab Shah, S. Chin, G. Turashvili, O. Rueda, M. Dunning, D. Speed, A. Lynch, S. Samarajiwa, Yinyin Yuan, S. Gräf, G. Ha, Gholamreza Haffari, A. Bashashati, R. Russell, S. McKinney, A. Langerød, A. Green, E. Provenzano, G. Wishart, S. Pinder, P. Watson, F. Markowetz, L. Murphy, I. Ellis, A. Purushotham, A. Børresen-Dale, J. Brenton, S. Tavaré, C. Caldas, S. Aparicio (2012)
The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroupsNature, 486
I. Komenaka, Chiu-Hsieh Hsu, M. Martínez, Marcia Bouton, B. Low, J. Salganick, J. Nodora, M. Hibbard, C. Jha (2011)
Preoperative chemotherapy for operable breast cancer is associated with better compliance with adjuvant therapy in matched stage II and IIIA patients.The oncologist, 16 6
D. Mauri, N. Pavlidis, J. Ioannidis (2005)
Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis.Journal of the National Cancer Institute, 97 3
S. Yachida, Siân Jones, I. Bozic, T. Antal, R. Leary, Baojin Fu, M. Kamiyama, R. Hruban, J. Eshleman, M. Nowak, V. Velculescu, K. Kinzler, B. Vogelstein, C. Iacobuzio-Donahue (2010)
Distant Metastasis Occurs Late during the Genetic Evolution of Pancreatic CancerNature, 467
Y. Chia, M. Ellis, Cynthia Ma (2010)
Neoadjuvant endocrine therapy in primary breast cancer: indications and use as a research toolBritish Journal of Cancer, 103
Abstract Despite the ever-changing breast surgeon’s technical role, the surgeon forms an indispensible link between imaging, diagnostics, pathology, and the medical oncologist. Biomarkers of prognosis, prediction of response, and resistance to treatments, including imaging, tissue and circulating markers apply to the primary diagnostic and treatment settings as well as scenarios which include disease recurrence, both in the early and advanced settings. Whether it is via the diagnostic clinic referred by the primary care physician or via a breast screening service, primary early breast cancer is referred for initial treatment and/or diagnosis and currently remains the domain of the surgical oncologist. The surgeon is privileged by this unique “window of opportunity” to consider the biological aspects of the diagnosis and guide the patient appropriately toward initial therapy, only one of which is primary surgery. Options of neoadjuvant endocrine, cytotoxic, or targeted therapy as either standard of care or else in the clinical trial context should be considered to optimize treatment in all patients. Precision therapy in medicine is a novel concept encompassing personalized medicine for all available treatment modalities including surgery, radiotherapy as well as systemic therapies. Precision chemotherapy was used to describe agents like the antibody–drug conjugate T-DM1, where trastuzumab selectively delivers a very effective cytotoxic agent directly to HER2-positive tumor cells, thereby sparing normal cells. Precision as a concept in treatment should also imply selecting the patient population most suitable for a particular treatment which is then personalized depending on the individual’s pharmacogenetic and tumor characteristics to maximize effectiveness (1). Surgery should be similarly tailored to match the patient’s preferences, the tumor’s biology, and predicted response to systemic treatment. Traditionally surgery has been the mainstay of breast cancer treatment. However, despite increasing realization of limitations of such an approach at the exclusion of other treatments, surgeons have commanded the domain of initial diagnosis and definitive treatment decisions. Until recently, surgeons played a key role in utilizing neoadjuvant endocrine therapy (initially tamoxifen and now aromatase inhibitors) in the postmenopausal patient unsuitable for surgery due to advanced disease or significant comorbidity precluding surgery (2). Increasingly, in a multidisciplinary care environment (ie, the breast unit) it behooves the surgeon to consider neoadjuvant systemic therapy (cytotoxic, endocrine, and targeted) as standard practice in selected circumstances and particularly in a research setting to examine the effect of novel treatments on a given tumor biology. Diagnosis A full history including a detailed family history, menopausal status, and current exogenous hormonal treatments (contraception, and hormone replacement) and examination is followed by “triple-assessment” using mammography, ultrasound, and ultrasound-guided core biopsy in most instances. While in the primary systemic therapy (PST) population, the majority of cancers will be symptomatic this is not exclusively the case. Care must be taken in the situation where a patient is referred from a screening unit to assess tumor characteristics and perform appropriate staging—even if repeat biopsy is sometimes necessary. The initial diagnostic biopsy should where possible include performance of estrogen receptor (ER), progesterone receptor (PR), HER2 immunohistochemistry (IHC) (+/− in situ hybridization) and Ki-67 in an accredited facility. The reliable determination of receptor phenotype as an approximation of intrinsic subtype is a prerequisite to decision making in PST. Further biopsies as part of standard operating procedure or research protocols should be considered for at least DNA sequencing and RNA expression analysis. Such tissue sampling is facilitated by using a vacuum-assisted device (increased tissue volume obtained) which can obtain multiple samples in a single pass and should be 14G or larger (3). In selected cases (eg, invasive lobular carcinoma [ILC]) magnetic resonance imaging should be considered to more accurately estimate true extent of disease. The surgeon’s decision to advise PST depends on several patient, tumor, and treatment factors and is facilitated in a breast unit setting with a multidisciplinary team discussion before initial treatment where an integrated decision with the medical oncologist takes place. Staging investigations such as computed tomography +/− bone scan should be reserved for patients with clinical stage III disease (either cN2 or cT3, cN1, or for any cT4) where occult metastases are detected in up to 14% of patients (4). Biological Evaluation—Selecting Patients for Standard Care Neoadjuvant chemotherapy was compared directly with adjuvant chemotherapy in the National Surgical Adjuvant Breast and Bowel Project (NSABP)-B18 trial and shown to give patients equivalent outcomes, both for disease-free survival and overall survival (5). In addition, patients aged under 50 years demonstrated a trend toward increased survival (disease-free survival and overall survival) for the neoadjuvant group (6) suggesting a subgroup who might benefit from the PST approach. It may well be that for certain breast cancer subtypes (not examined in B18) delay in chemotherapy initiation may have contributed. De Melo Gagliato et al. (7) showed that for HER2+ and triple-negative tumors such a delay (>60 days) results in poorer overall survival. It is well accepted that the pathological complete response (pCR) rates attained depend on receptor phenotype as well as the treatment regime (8). Subtype-specific pCR rate was: 8.3% in hormone receptor (HR)+/HER2−, 18.7% in HR+/HER2+, 31.1% in triple negative, and 38.9% in HER2+/HR− (9). One of the guiding principles of patient selection is to use the same pathological selection criteria as for adjuvant chemotherapy. It is interesting to reflect that recurrence score (OncotypeDx), validated in NSABP-B14, B-20, and SWOG-8814 has been shown to predict response in patients with HR+, HER2− disease to paclitaxel and doxorubicin (10), to docetaxel (11), and to ixabepilone and cyclophosphamide (12) in the PST setting. Tumor size is clearly a determinant. Down-sizing, which may convert the need for a mastectomy into breast conserving therapy is one of the clear indications for considering PST. For cT2/3 tumors it has been shown that a reduction in tumor volume excised (13) and re-excisions can be achieved (14). For cT1 tumors (<2cm) this aspect is not as clear as there has not been shown to be a benefit in terms of breast volume excised (13). In fact, the tumor bed must still be removed given that current imaging cannot reliably predict pCR (15). Patients to be considered are those with high-grade tumors, HER2+ or triple negative as well as those with cN1/N2 disease or fine needle aspiration (FNA)-positive nodal metastasis despite small primary tumor size. Surgery following PST is still recommended for all patients given higher local recurrence rates when radiotherapy was given after omitting surgery despite a clinical complete response (16,17). A radio-opaque or ultrasound-detectable marker should be deployed early in the treatment course, especially in cases where biology predicts high rates of pCR. It remains to be seen whether definitive surgery can be avoided for those with clinical complete response and a biopsy-proven pCR. The significance of PST however are far wider-reaching than mere cosmetic outcome. The “window of opportunity” for in vivo chemosensitivity testing of the efficacy of treatment on the primary (and nodal disease) and knowledge of therapy resistance can provide much valuable information, in addition to being prognostic for the patient and facilitating translational research. The question of down-staging axillary nodal disease is now a focus of ongoing study (18) and importance for minimizing surgery-associated arm morbidity but is beyond the scope of this paper. Modification of Surgery Based on Response and Phenotype There is evidence that phenotype based on receptor status affects risk of loco-regional recurrence in the adjuvant setting (19) and at least in retrospective comparisons is no different in the PST setting (20). For example, triple-negative receptor status for a patient with a poor response or progressive disease in the PST setting should lead to a more aggressive surgical approach to margin clearance as the risk of loco-regional recurrence is higher (21). Intraoperative ultrasound and/or multiparametric magnetic resonance imaging with single or multiple marker placement can be used to guide surgical excision more precisely in the PST setting. Careful mapping of any visible residual tumor, which is often irregular in distribution, should be undertaken. Where there is doubt about conservability, full use of oncoplastic techniques to maximize aesthetic outcome and margins should be made. This may involve “re-staging” the local extent of disease, particularly in the assessment of microcalcification. Close liaison with the radiologist and rebiopsy of microcalcification to determine the extent or presence of ductal carcinoma in situ (DCIS) is essential for surgical planning. Despite pCR definitions not including the presence of DCIS as its presence does not influence prognosis, it will affect surgical margins and therefore the approach to resection. Research Setting The ideal schema for the study of novel therapeutic agents or combinations was agreed at the previous consensus (22) and involves tissue sample collection ideally at the time of initial diagnostic biopsy for lesions which are suspicious on clinical and/or imaging criteria. Extra biopsies should be taken for both expression analysis and genomic analysis (eg, next generation sequencing) and epigenetic studies (DNA methylation) at this time. Studies will increasingly demand a genetic screen or selection to enrich the study population likely to respond to a given treatment. Blood for germline (genomic) DNA should also be collected at this time. Tissue biomarkers predictive of early response or resistance (such as Ki67) taken at 2 weeks or in some cases even earlier following commencement of treatment are an established need (23). Biomarkers should to be tailored to the specifics of the (on target) mechanism of action of agents being trialed (eg, cleaved caspase 3 for proapoptotic agents). For many studies this will be of an exploratory nature. Finally, the surgical specimen should have the residual cancer burden characterized and quantified using a validated index such as that of Symmans et al. (24). Tissue sampling and handling is critical and representative biopsies at the time of surgery to maximize quality and ensure adequate quantity for both DNA and RNA analysis should be ensured. Intratumoral Heterogeneity We propose a model based on the premise that intratumoral heterogeneity (ITH) is responsible for treatment resistance in a significant proportion of primary tumors (25). Partial response to PST may be due to the preexistence of a subclonal population within the primary which is selectively favored by the successful treatment of a competing subpopulation. Ten subtypes in 2000 primary breast cancer samples based on an integrated analysis of whole genomic copy number aberration and gene expression were described (26) and accounted for interpatient variation in breast cancer type. However, breast cancer heterogeneity has also been recognized to exist within patients when the molecular phenotype of the primary cancer is compared with metastatic deposits at sites remote to the breast (27). Several studies comparing receptor expression between paired biopsies have shown an altered phenotype at metastatic sites including axillary nodes, with either a reduction or loss of ER expression in up to 35% of tumors, and gain of HER2 in up to 10% of ER-positive breast cancers (28–30). By the time of clinical diagnosis, a primary breast cancer may comprise at least three or four detectable significant subclonal populations of cancer cells (31–34). The clinical significance of such underlying genetic ITH, is that it is postulated to be the main source for heterogeneity across different (metastatic) sites and to be a major mechanism of acquired resistance to treatment (25,35). While it was thought that treatment might lead to clonal selection of a resistant cell population which then metastasizes, it is more consistent with current evidence that ITH at the primary site, present before treatment, arising during cancer evolution, already exists (36,37). Particular subclones metastasize, survive, and develop into manifest recurrence at distant sites. The primary cancer site may act as a clonal “source” from which different subclones originate and disseminate to form heterogeneous metastases. Changes in clonal composition could be monitored during treatment to predict and anticipate development of drug resistance (38) and allow early modification of treatments before clinical progression occurs. Next generation sequencing techniques may provide detailed analysis of a single or multiple tumor core biopsies performed at different times (eg, before, during, and after treatment) on any number of patients, particularly those with rapidly acquired and clinically recognized treatment resistance—manifest by residual disease. Biopsy with a view to DNA sequencing and resolution of cellular (subclonal) populations will lead to an understanding of not only the primary tumor characteristics at diagnosis, but also the effect of drug treatments on the dominant populations and the proportional distribution of subclonal populations which the effects of drug treatments may alter. Genetic ITH has been described in human breast cancer through investigation of ex-vivo cancer specimens, cell lines and mouse xenografts (31). In a study of ITH in 20 breast cancers, almost half (n = 9) were found to be monogenomic (defined as a homogenous population of cells with similar genomic profiles throughout the tumor), the remainder polygenomic (32). In the latter group, the anatomical distribution of the clonal subpopulations was either segregated (localized to one region) or intermixed throughout the tumor. It can be speculated that the latter may account for the “scattered” response seen with PST irrespective of receptor phenotype. For some tumors a single biopsy may be representative of the genetic make-up of the entire lesion whereas for the “segregated” polygenetic tumors, multiple biopsies from different regions may be required. Currently used indices such as tumor grade, ER, PR, HER2, and Ki-67 and their distribution within a tumor do not necessarily correspond to the presence and distribution of genetic heterogeneity (32). We suggest that tissue collection taken before treatment and at the time of surgery undergo analysis to compare possibly heterogeneous populations of cells and their relative abundance, by various techniques including fluorescent-activated cell sorting and next generation sequencing with deep exome sequencing. This applies when there is a lack of a complete response to treatment. The nature of the residual tumor and the possible emergence of new subclones is of particular interest. Involved axillary nodes could also be sampled, particularly in the situation where there is a differential treatment response between the primary breast tumor and the axillary nodal metastases. Resampling and sequencing the residual cancer burden for clonal characterization may eventually lead to rational trialing of additional therapies by switching or combining treatments with those already given. Instead of surgery at the time of either completion of a predetermined number of chemotherapy cycles or after stable disease/progressive disease has been detected, rebiopsy of the residual or resistant tumor to guide further treatment might be the next stage in the evolution of the neoadjuvant testing paradigm (Figure 1). Figure 1. View largeDownload slide A generalized, hypothetical schema for neoadjuvant testing of multiple novel agents. Drug A is a standard therapy (eg, endocrine therapy) and Drug B is a novel agent tested in combination with A. Both drugs have a lead-in period (alone) for 2 weeks to permit biomarker study. They are then combined and the four arms are randomized for this purpose. The three combination arms can then be recombined after the initial biomarker analysis. Rebiopsy at the end of the regime is undertaken instead of definitive surgery. If residual disease is present, the patient can be further randomized to test one or more agents (or against a standard agent). Tissue biopsy may include study of tumor heterogeneity and blood is taken at multiple time points for circulating markers (circulating tumor cells and plasma nucleic acids). pCR = pathological complete response; RAN = randomization. Figure 1. View largeDownload slide A generalized, hypothetical schema for neoadjuvant testing of multiple novel agents. Drug A is a standard therapy (eg, endocrine therapy) and Drug B is a novel agent tested in combination with A. Both drugs have a lead-in period (alone) for 2 weeks to permit biomarker study. They are then combined and the four arms are randomized for this purpose. The three combination arms can then be recombined after the initial biomarker analysis. Rebiopsy at the end of the regime is undertaken instead of definitive surgery. If residual disease is present, the patient can be further randomized to test one or more agents (or against a standard agent). Tissue biopsy may include study of tumor heterogeneity and blood is taken at multiple time points for circulating markers (circulating tumor cells and plasma nucleic acids). pCR = pathological complete response; RAN = randomization. Tissue Collection for Research Modern biopsy devices, particularly those capable of single-pass multiple biopsy acquisitions with vacuum assistance to minimize patient discomfort and maximize accuracy of biopsy placement (and avoidance of multiple passes which require relocalization for each one) should be utilized in this setting. Such devices may increase patient compliance and reduce risk compared with devices such as the older spring-loaded single fire biopsy ones. Use of such devices at diagnosis may reassure patients regarding the need for further biopsies, particularly when consenting to clinical trials of PST. Patient Decision Making With the surgeon as the initial team member at or after diagnosis of breast cancer, patients will be guided by advice given, as they perceive the surgeon to be the pivotal member of the team. The surgeon who not only supports but also advocates chemotherapy as initial treatment instead of surgery might find greater patient acceptance of chemotherapy compared with the adjuvant setting (39). This is an opportunity to educate patients about the potential advantages of PST. These include conversion of mastectomy into breast conservation or a reduction in the extent of surgery in BCT resulting in enhanced oncoplastic outcome and aesthetic results. Furthermore, a neoadjuvant approach for patients who are still advised to undergo mastectomy (eg, germline carriers of BRCA1 or 2 mutations) may be subject to reduction in complications from immediate reconstruction if chemotherapy is completed before surgery and adjuvant radiotherapy is deemed unnecessary. Patient acceptance may in part be due to the clinical and radiological objective responses seen with PST which in the adjuvant setting is difficult for patients to comprehend. In fact, adjuvant systemic therapy is a “blind” procedure as it is given after the only opportunity to monitor treatment effectiveness has been eliminated. The effect on potential micrometastatic disease can still be emphasized in the PST setting especially when objective tumor response is seen. Furthermore if tumor progression or lack of objective response occurs, switching agents (eg, from the standard anthracycline-based to taxane-based regime) may provide some reassurance. The early truncation/cessation of a full regime which is seen to be ineffective may also reassure patients that they are not receiving excessive potentially solely toxic treatment. A surgeon endorsing such an approach to treatment and supporting the medical oncologist may give patients the reassurance to participate and comply more fully. Conclusion Despite the aim of minimizing the extent of surgery and its unwanted side-effects, surgeons need to adopt an increasingly active and interactive role in the PST setting. This includes patient selection, biological assessment of the tumor, monitoring response to standard regimes and newer agents in the clinical trial context as well as determining the most efficacious form of definitive surgery on completion of treatment. Closer engagement and regular collaboration with radiologists, pathologists, and medical oncologists will greatly facilitate this to optimize long-term patient outcomes. References 1. Hess KR Anderson K Symmans WF et al. Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide in breast cancer. J Clin Oncol . 2006; 24( 26): 4236– 4244. 2. Chia YH Ellis MJ Ma CX . Neoadjuvant endocrine therapy in primary breast cancer: indications and use as a research tool. Br J Cancer . 2010; 103( 6): 759– 764. 3. Lourenco AP Mainiero MB Lazarus E Giri D Schepps B . Stereotactic breast biopsy: comparison of histologic underestimation rates with 11- and 9-gauge vacuum-assisted breast biopsy. AJR Am J Roentgenol . 2007; 189( 5): W275– W279. 4. Barrett T Bowden DJ Greenberg DC Brown CH Wishart GC Britton PD . Radiological staging in breast cancer: which asymptomatic patients to image and how. Br J Cancer . 2009; 101( 9): 1522– 1528. 5. Mauri D Pavlidis N Ioannidis JP . Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis. J Natl Cancer Inst . 2005; 97( 3): 188– 194. 6. Wolmark N Wang J Mamounas E et al. Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18. J Natl Cancer Inst Monogr . 2001;( 30): 96– 102. 7. De Melo Gagliato D Gonzalez-Angulo AM Lei X et al. Impact of delaying initiation of adjuvant chemotherapy in breast cancer patients. J Clin Oncol . 2013; 31( suppl; abstr 1022). 8. Rouzier R Perou CM Symmans WF et al. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res . 2005; 11( 16): 5678– 5685. 9. Houssami N Macaskill P von Minckwitz G Marinovich ML Mamounas E . Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy. Eur J Cancer . 2012; 48( 18): 3342– 3354. 10. Gianni L Zambetti M Clark K et al. Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer. J Clin Oncol . 2005; 23( 29): 7265– 7277. 11. Chang JC Makris A Gutierrez MC et al. Gene expression patterns in formalin-fixed, paraffin-embedded core biopsies predict docetaxel chemosensitivity in breast cancer patients. Breast Cancer Res Treat . 2008; 108( 2): 233– 240. 12. Yardley DA Peacock NW Hendricks C et al. P5-13-09: Correlation of oncotype DX recurrence scores with pathologic response following neoadjuvant ixabepilone and cyclophosphamide in patients with HER2–negative breast cancer: A Sarah Cannon Research Institute Phase II Trial. Cancer Res . 2011; 71( 24 suppl:Abstract nr P5-13-09). doi: 10.1158/0008-5472.SABCS11-P5-13-09 13. Boughey JC Peintinger F Meric-Bernstam F et al. Impact of preoperative versus postoperative chemotherapy on the extent and number of surgical procedures in patients treated in randomized clinical trials for breast cancer. Ann Surg . 2006; 244( 3): 464– 470. 14. Komenaka IK Hibbard ML Hsu CH et al. Preoperative chemotherapy for operable breast cancer improves surgical outcomes in the community hospital setting. Oncologist . 2011; 16( 6): 752– 759. 15. Marinovich ML Sardanelli F Ciatto S et al. Early prediction of pathologic response to neoadjuvant therapy in breast cancer: systematic review of the accuracy of MRI. Breast . 2012; 21( 5): 669– 677. 16. Ring A Webb A Ashley S et al. Is surgery necessary after complete clinical remission following neoadjuvant chemotherapy for early breast cancer? J Clin Oncol . 2003; 21( 24): 4540– 4545. 17. Ellis P Ashley S Walsh G et al. Identification of clinical factors predicting outcome following primary chemotherapy for operable breast cancer. The Breast . 1999; 8: 235. 18. Kuehn T Bauerfeind I Fehm T et al. Sentinel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study. Lancet Oncol . 2013; 14( 7): 609– 618. 19. Lowery AJ Kell MR Glynn RW et al. Locoregional recurrence after breast cancer surgery: a systematic review by receptor phenotype. Breast Cancer Res Treat . 2012; 133( 3): 831– 841. 20. Mittendorf EA Buchholz TA Tucker SL et al. Impact of chemotherapy sequencing on local-regional failure risk in breast cancer patients undergoing breast-conserving therapy. Ann Surg . 2013; 257( 2): 173– 179. 21. Caudle AS Yu TK Tucker SL et al. Local-regional control according to surrogate markers of breast cancer subtypes and response to neoadjuvant chemotherapy in breast cancer patients undergoing breast conserving therapy. Breast Cancer Res . 2012; 14( 3): R83. 22. Berruti A Generali D Kaufmann M et al. International expert consensus on primary systemic therapy in the management of early breast cancer: highlights of the Fourth Symposium on Primary Systemic Therapy in the Management of Operable Breast Cancer, Cremona, Italy (2010). J Natl Cancer Inst Monogr . 2011; 2011( 43): 147– 151. 23. Dowsett M Smith IE Ebbs SR et al. Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst . 2007; 99( 2): 167– 170. 24. Symmans WF Peintinger F Hatzis C et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol . 2007; 25( 28): 4414– 4422. 25. Turner NC Reis-Filho JS . Genetic heterogeneity and cancer drug resistance. Lancet Oncol . 2012; 13( 4): e178– e185. 26. Curtis C Shah SP Chin SF et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature . 2012; 486( 7403): 346– 352. 27. Aitken S Thomas J Langdon S et al. Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases. Ann Oncol . 2010; 21( 6): 1254– 1261. 28. Amir E Clemons M Purdie CA et al. Tissue confirmation of disease recurrence in breast cancer patients: pooled analysis of multi-centre, multi-disciplinary prospective studies. Cancer Treat Rev . 2012; 38( 6): 708– 714. 29. Curigliano G Bagnardi V Viale G et al. Should liver metastases of breast cancer be biopsied to improve treatment choice? Ann Oncol . 2011; 22( 10): 2227– 2233. 30. Lindström LS Karlsson E Wilking UM et al. Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol . 2012; 30( 21): 2601– 2608. 31. Ding L Ellis MJ Li S et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature . 2010; 464( 7291): 999– 1005. 32. Navin N Krasnitz A Rodgers L et al. Inferring tumor progression from genomic heterogeneity. Genome Res . 2010; 20( 1): 68– 80. 33. Navin N Kendall J Troge J et al. Tumour evolution inferred by single-cell sequencing. Nature . 2011; 472( 7341): 90– 94. 34. Nik-Zainal S Van Loo P Wedge DC et al. The life history of 21 breast cancers. Cell . 2012; 149( 5): 994– 1007. 35. Yap T Gerlinger M Futreal P et al. Intratumor heterogeneity: seeing the wood for the trees. Sci Transl Med . 2012; 4( 127): 127ps10. 36. Liu W Laitinen S Khan S et al. Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat Med . 2009; 15( 5): 559– 565. 37. Yachida S Jones S Bozic I et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature . 2010; 467( 7319): 1114– 1117. 38. Ellis MJ Ding L Shen D et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature . 2012; 486( 7403): 353– 360. 39. Komenaka IK Hsu CH Martinez ME et al. Preoperative chemotherapy for operable breast cancer is associated with better compliance with adjuvant therapy in matched stage II and IIIA patients. Oncologist . 2011; 16( 6): 742– 751. © The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.
JNCI Monographs – Oxford University Press
Published: Jun 10, 2015
Read and print from thousands of top scholarly journals.
Already have an account? Log in
Bookmark this article. You can see your Bookmarks on your DeepDyve Library.
To save an article, log in first, or sign up for a DeepDyve account if you don’t already have one.
Copy and paste the desired citation format or use the link below to download a file formatted for EndNote
Access the full text.
Sign up today, get DeepDyve free for 14 days.
All DeepDyve websites use cookies to improve your online experience. They were placed on your computer when you launched this website. You can change your cookie settings through your browser.