Skip to main content

SYSTEMATIC REVIEW article

Front. Oncol., 26 June 2020
Sec. Women's Cancer

The Diagnostic Performance of Minimally Invasive Biopsy in Predicting Breast Pathological Complete Response After Neoadjuvant Systemic Therapy in Breast Cancer: A Meta-Analysis

\nYan LiYan LiYidong ZhouYidong ZhouFeng MaoFeng MaoYan LinYan LinXiaohui ZhangXiaohui ZhangSongjie ShenSongjie ShenQiang Sun
Qiang Sun*
  • Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China

Background: Neoadjuvant systemic therapy (NST) is commonly used in patients with early stage breast cancer before definitive surgery. The standard diagnostic approach for pathologic complete response (pCR) of the breast is breast surgery and pathologic examination. In recent years, several trials investigated the predictive value of image-guided minimally invasive biopsy (MIB) for breast pCR after NST. This study conducted a meta-analysis to evaluate the diagnostic accuracy of MIB.

Materials and Methods: We identified relevant research reports in online databases through February 2020. The Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) tool was used to evaluate the quality of included trials. We extracted relevant data and constructed a 2 × 2 contingency table to analyze the predictive accuracy of MIB for breast pCR. Subgroup analyses and meta-regressions were also performed to investigate potential causes of heterogeneity.

Results: Nine trials (with 1,030 breast cancer patients) were included in this meta-analysis. The pooled sensitivity and specificity of MIB were 0.72 [95% confidence interval (CI): 0.61–0.81] and 0.99 (95% CI: 0.89–1.00), respectively. By combining relevant data, there were no significant differences in sensitivity or specificity among different molecular subtypes of breast cancer (P > 0.05). Subgroup analyses and meta-regressions implied that trials with responses not limited to clinical complete response (cCR) had a significantly higher accuracy of MIB than those with only cCR (RDOR: 7.65; 95% CI: 1.05–55.46; P = 0.046).

Conclusion: Current image-guided MIB methods are not accurate enough in terms of predicting breast pCR after NST. It is of utmost clinical importance to standardize the MIB procedure and incorporate other factors into the evaluation in order to improve the accuracy to an acceptable level.

Introduction

Neoadjuvant systemic therapy (NST) is used in approximately 30% of patients with early stage breast cancer before definitive surgery (1). Pathologic complete response (pCR) is an ideal response to NST, indicating the absence of residual cancer in a surgical specimen although it has different definitions (2, 3). In recent years, with the improvement of neoadjuvant chemotherapy and targeted therapy, the pCR rates of breast cancer have increased dramatically. For triple-negative (TN) and human epidermal growth factor receptor 2-positive (HER2+) subtypes, pCR rates of up to 60–70% can be achieved with the administration of carboplatin regimens and dual HER2 blockage (4, 5).

Achievement of pCR after NST is associated with less recurrence and favorable survival of breast cancer, and recent studies show that escalation of adjuvant systemic therapy could have additional benefits for patients with residual disease (non-pCR) (2, 3, 6, 7). Currently, the standard diagnostic approach for pCR of the breast is breast surgery and pathologic examination of the specimen. As one of the main options of breast surgery, breast-conserving surgery after NST is universally performed, the oncologic safety of which has been confirmed by a series of studies (8, 9). For patients with no residual cancer in the breast (breast pCR), it is reasonable to consider an omission of breast surgery (10, 11). From this point of view, it is of great clinical significance to explore a less invasive method to predict breast pCR after NST.

Some studies focus on the accuracy and reliability of non-invasive imaging methods to predict breast pCR, but the results are far from satisfactory. Neither ultrasound nor mammography is reliable with false negative rates (FNR) ranging from 9 to 70% (1214). Similarly, magnetic resonance imaging (MRI) demonstrated an FNR up to 30–50% in predicting breast residual tumor after NST (1517). Imaging alone is not accurate enough to replace the pathologic examination of a surgical specimen.

In recent years, several trials have investigated the predictive value of image-guided minimally invasive biopsy (MIB) for breast pCR after NST, and these approaches include core needle biopsy (CNB), vacuum-assisted biopsy (VAB), and fine-needle aspiration (FNA) (1827). With different biopsy procedures, these trials demonstrate the diverse accuracy of MIB in identifying breast pCR.

In the present study, meta-analysis is performed to assess the diagnostic accuracy of MIB in predicting breast pCR after NST. We also perform subgroup analyses and meta-regressions to find which factors are associated with the predictive capability.

Materials and Methods

Search Strategy and Study Inclusion

This review was conducted according to the guidelines stipulated in Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) for diagnostic test accuracy (28). We searched electronic databases, including PubMed, EMBASE, and the Cochrane library, and the latest search was performed on February 25, 2020. In addition, we searched conference presentations and abstracts, such as San Antonio Breast Cancer Symposium (SABCS), American Society of Clinical Oncology (ASCO) meetings, and the European Society for Medical Oncology (ESMO) meetings held within the last 15 years. To identify relevant studies, the following terms were employed as queries: “breast cancer,” “neoadjuvant,” “biopsy,” and “pathologic complete response (OR pCR)” as well as MeSH terms “breast neoplasms” and “neoadjuvant therapy.” We included clinical trials using the following criteria: (1) The investigation involved patients with early stage invasive breast cancer who received NST; (2) after completion of NST, the patients received MIB (e.g., CNB, VAB, and FNA) of the breast, followed by standard breast surgery (lumpectomy or mastectomy); (3) the trials report histopathologic results of both index tests (MIB) and reference standard tests (surgery) and provide measures of test accuracy (e.g., sensitivity, specificity, and false negative rate), which allowed construction of a 2 × 2 contingency table with absolute numbers of true positive (TP), true negative (TN), false positive (FP), and false negative (FN) results. Studies meeting the following criteria were excluded: (1) reviews or case reports, (2) studies focusing on non-invasive tests (e.g., imaging examinations), (3) studies focusing on axillary evaluation rather than breast, and (4) studies without sufficient data even after attempting to contact the corresponding authors.

Data Extraction and Quality Evaluation

Data extraction and the evaluation of the quality of the studies were independently performed by two reviewers (YL and YZ). In cases of discrepancies, consensus was reached by them. The following data were extracted: first author, country of origin, update year, study design, sample size, patient characteristics, procedures of MIB, breast pCR rate, test accuracy measures, and complications of biopsy. For investigations with more than one report, data were gathered from the most recent findings.

Residual tumor was defined as “positive,” and the absence of residual tumor was defined as “negative” in both surgical and MIB specimens. For purposes of data synthesis, we extracted raw cell numbers for TP, TN, FP, and FN. If the numbers were not available, we employed the Revman calculator to generate these data based on the detailed information of test accuracy measures.

Eligible studies were evaluated for quality using the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) tool.

Statistical Analysis

Inter-study heterogeneity was evaluated using the Cochran's Q statistics and I2-test. Heterogeneity was considered significant as either P < 0.05 or I2 > 50%. In the absence of statistically significant heterogeneity, we calculated the pooled effect using a fixed-effects model; however, with significant heterogeneity, we employed the Mantel–Haenszel random effects model.

The measures of interest for pooled analyses included sensitivity, specificity, false negative rate (FNR = 1-sensitivity), negative likelihood ratio (NLR), positive likelihood ratio (PLR), and diagnostic odds ratio (DOR). Sensitivity and specificity of each study were used to plot the summary receiver operator characteristic (SROC) curve with the area under the curve (AUC) to indicate test accuracy.

For those studies reporting detailed information of specific molecular subtypes of breast cancer, we performed pooled analyses according to different molecular subtypes (TNBC, HER2+, and HR+HER2−). We also conducted subgroup analyses and meta-regressions according to region, study design, tumor response to NST, and sample size in evaluating potential causes of heterogeneity. The relative DOR (RDOR) was used to assess the heterogeneity of subgroups.

The analysis was performed using Stata 12.0 software (Stata Corporation, College Station, TX, USA), RevMan 5.3 software (the Cochrane Information Management System), and Meta-Disc (XI Cochrane Colloquium; Barcelona, Spain). P < 0.05 (two-sided) was considered to be statistically significant.

Results

Characteristics of the Studies

A total of nine clinical trials fulfilled the eligibility criteria. Figure 1 shows the search and selection process used in this study. Altogether, the nine trials described a total of 1,030 patients. Among the trials included, five had published articles with full text, and the other four were reported only as abstracts at annual conferences; five were single-center studies, and four were multicenter studies; eight were prospective, and only one was retrospective; six of them were carried out in Europe, two in America, and one in Asia (Table 1).

FIGURE 1
www.frontiersin.org

Figure 1. Flow diagram of the systematic search and selection process of studies.

TABLE 1
www.frontiersin.org

Table 1. Study characteristics.

With regard to the response to NST of included patients, three of the trials only included patients with clinical complete response (cCR) by imaging and physical examination, and six of them included patients with responses not limited to cCR (plus other responses, e.g., clinical partial response, “cPR”) (Table 1).

The procedures of MIB of all of the included studies were guided by imaging methods (ultrasound, mammography, or both). Three of the studies used CNB, three of them used VAB, two used CNB and/or VAB, and one used a combination of FNA and VAB. Standard breast surgeries (lumpectomy or mastectomy) were performed following the biopsy (Table 1).

Histopathological evaluation of both MIB and surgical specimen was performed to see whether residual tumors existed. In all of the nine included trials, breast pCR was defined as the absence of cancer (both invasive and in situ, “ypT0”).

The quality assessment of included studies by the QUADAS-2 tool is shown in Figure 2. One study has a high risk of bias for patient selection due to a lack of a clear description of the inclusion criteria. Risks of bias for the index test and reference test were primarily caused by a lack of reported blinding when performing pathologic examinations.

FIGURE 2
www.frontiersin.org

Figure 2. Evaluation of risk of bias by QUADAS-2 tool. (A) Risk of bias graph and (B) risk of bias summary.

Pooled pCR Rate According to Histopathologic Examinations of Surgical Specimens

The pCR rates confirmed by histopathologic examinations of surgical specimens of the included nine studies ranged from 10.0 to 67.5%. A random-effects model was employed due to the significant heterogeneity (P < 0.01, I2 = 85.5%). The pooled pCR rate was 49.0% with a 95% CI of 40.0–57.1% (Figure 3).

FIGURE 3
www.frontiersin.org

Figure 3. Forest plot of meta-analysis on breast pCR rate of surgical specimen.

Overall Diagnostic Accuracy of MIB in Predicting pCR

Sensitivities of MIB of the included studies ranged from 50.0 to 95.0%, and specificities ranged from 85.0 to 100.0%. We selected the random effects model based on significant heterogeneity in both sensitivity and specificity (sensitivity: P < 0.01, I2 = 83.5%; specificity: P < 0.01, I2 = 80.5%). The pooled sensitivity was 0.72 with a 95% CI of 0.61–0.81. The pooled specificity was 0.99 with a 95% CI of 0.89–1.00 (Figure 4). The pooled PLR was 11.61 (5.74–23.50), the pooled NLR was 0.32 (0.23–0.44), and the pooled DOR was 38.99 (19.38–78.45).

FIGURE 4
www.frontiersin.org

Figure 4. Forest plot of meta-analysis on pooled sensitivity and specificity of MIB.

The AUC of the SROC provides a global summary of the diagnostic performance of included studies. The AUC was 0.90 (0.87–0.93) as shown in Figure 5. An AUC score of 1.0 indicates a perfect diagnostic performance.

FIGURE 5
www.frontiersin.org

Figure 5. Summary receiver operator characteristic (SROC) curve of diagnostic performance of MIB.

Pooled Analyses According to Molecular Subtypes

Several studies described detailed information about the test accuracy of MIB in specific molecular subtypes of breast cancer (19, 2224, 26). Table 2 shows a pooled analysis based on these data. For TNBC subtype, the pooled sensitivity was 0.47 (0.31–0.64), the pooled specificity was 0.97 (0.90–1.00), and the pooled DOR was 25.26 (6.07–105.20). For the HER2+ subtype, the pooled sensitivity was 0.59 (0.41–0.76), the pooled specificity was 0.94 (0.87–0.98), and the pooled DOR was 20.74 (3.97–108.51). For the HR+HER2− subtype, the pooled sensitivity was 0.60 (0.47–0.72), the pooled specificity was 0.94 (0.87–1.00), and the pooled DOR was 27.27 (4.84–153.76). There were no significant differences in sensitivity, specificity, or DOR among these subtypes (P > 0.05).

TABLE 2
www.frontiersin.org

Table 2. Pooled analyses by molecular subtypes of breast cancer.

Subgroup Analysis and Meta-Regression

We also performed subgroup analysis and meta-regression according to region, study design, number of centers, response to NST, and sample size. In terms of response to NST, the studies with responses not limited to cCR had a significantly higher accuracy than those with only cCR (RDOR: 7.65; 95% CI, 1.05–55.46; P = 0.046). However, meta-regression analysis showed that other factors had no significant influence on the diagnostic performance of MIB (P > 0.05; Table 3).

TABLE 3
www.frontiersin.org

Table 3. Subgroup analysis and meta-regression.

Complications of MIB

Only two of the studies described complications after MIB. Basik et al. reported that 7.1% of the patients had post-procedure complications, with hematoma being the most common. However, Kuerer et al. reported a complication rate of 20.0% with bleeding as the most common, followed by hematoma [Table 1; (19, 23)].

Publication Bias Assessment

Publication bias was not analyzed because of the limited number of included studies (29).

Discussion

In current clinical practice, breast surgery is an important part of standard multimodal treatment for breast cancer patients after NST, and it is also the primary approach to evaluating the pathologic response to NST. Without pathologic examination of surgical specimens, a definitive pathologic diagnosis cannot be obtained. Breast-conserving surgery, followed by radiotherapy and adjuvant systemic therapy, has being confirmed safe in terms of cancer recurrence and overall survival (30). The smaller the tumor size after NST, the less tissue needs to be excised as long as there is no residual tumor in the margins (8). Thus, for patients with breast pCR after NST, complete avoidance of breast surgery is a reasonable option (11, 31). Therefore, it is extremely important to explore a non-surgical approach that can replace breast surgery in predicting pCR for breast cancer patients after NST.

Multiple studies evaluated the reliability of imaging approaches, including ultrasound, mammography, and MRI, in predicting breast pCR after NST. However, the accuracy of these techniques was far from satisfactory (1217). Some early studies reported that using radiotherapy alone (without breast surgery) as the local treatment approach for breast cancer patients with cCR on imaging will lead to much higher rates of relapses (3133). In recent years, there has been increasing interest in investigating the diagnostic performance of minimally invasive approaches.

To evaluate the diagnostic accuracy of image-guided MIB to identify residual cancer in the breast after NST, FNR (1-sensitivity) is a measure of paramount importance. “False negative” means that patients with the residual disease are diagnosed as “pCR,” which might lead to the incorrect omission of surgery and de-escalation of systemic therapy. On the contrary, “false positive” is of less importance because all of the residual tumors might be removed by MIB, and it is possible that the tumor was present only in the MIB specimen but not in the surgical specimen. In addition, if the MIB made a “positive” diagnosis, subsequent breast surgery is mandatory to remove potential residual disease. With regard to the maximum acceptable FNR, which will not translate into significantly worse survival outcomes, most of the included trials deduced 10% from the study design of sentinel lymph node trials (34, 35). An FNR <10% was considered acceptable although there is a lack of evidence.

This meta-analysis showed that the pooled sensitivity of image-guided MIB for the diagnosis of residual disease in the breast was 0.72, which means that the FNR was as high as 28%. An FNR of 28% in this pooled analysis is obviously far from accurate, which means that a large proportion of patients with residual disease would be diagnosed as “pCR.” Thus, breast surgery might be incorrectly omitted. Furthermore, escalation of adjuvant systemic therapy would not be administered to the patients diagnosed as “pCR.” Based on these consequences, it is reasonable to believe that missed residual tumor in the breast will eventually lead to insufficient treatment and, in the long run, lead to more relapses and worse survival outcomes. However, there is a lack of evidence about the recurrence and survival outcome of breast cancer patients with MIB-confirmed “pCR,” who forgo breast surgery accordingly. A clinical trial at MD Anderson is in the accrual phase, and the aim is to evaluate the survival consequence of eliminating breast cancer surgery in patients with VAB-confirmed “pCR” (36).

In terms of molecular subtypes of breast cancer, by combining relevant data, our pooled analysis showed that sensitivities, specificities, FNRs, and DORs had no significant differences among different subtypes. Different molecular subtypes of breast cancer showed different patterns of tumor shrinkage to NST: TNBC and HER2+ subtypes mostly exhibit concentric shrinkage, and luminal types mostly exhibit “honeycomb-like” shrinkage featured as scattered tiny foci of tumor and diffuse cell loss (37). This heterogeneous fashion of “honeycomb-like” pattern may increase the likelihood of wrong sampling of MIB, leading to low diagnostic accuracy. One of the trials included only TNBC and HER2+ subtypes and obtained a FNR of 5% (23). However, other trials reported contradictory results (19, 24). Future studies are needed to clarify the effects of molecular subtype on the accuracy of MIB.

This study also implied that trials with only response of cCR to NST had worse diagnostic accuracy than trials with responses not limited to cCR. Breast tumor evaluated as cCR by imaging methods has no clearly visible lesions. In this circumstance, MIB is always guided by clip markers placed in the tumor location prior to NST. Nevertheless, the location of clip markers is not necessarily where the tumor used to be, especially as the local tissue has changed dramatically after NST. As a result, this guidance method inevitably increases the likelihood of sampling error. For the tumor that is visible on the imaging or even palpable, it is more likely that the biopsy catches the tumor tissue. The uncertainty of clip-guided biopsy may partly explain the finding of this study, and it is helpful for further discussing the most appropriate patient group to have MIB.

It is important to notice that the procedures of MIB of the included trials varied a lot, and that is one of the main origins of heterogeneity. Many aspects of the MIB procedure can determine the diagnostic performance, including guiding imaging methods (ultrasound, mammography, and MRI), biopsy apparatus (CNB or VAB), size of needle, number of cores, and number and location of clip markers. Lee et al. reported that if more than five samples were obtained, the FNR would be around 10% (27). Heil et al. reported that using a 7G needle dramatically increases the accuracy of VAB (18). However, these included trials did not provide enough data for a pooled subgroup analysis in terms of the MIB procedure. It is of utmost importance for future studies to propose a standardized MIB procedure, which can achieve the highest predictive accuracy.

It is reported that there are other factors that are associated with pCR rates after NST for breast cancer. These factors include tumor-infiltrating lymphocytes and several specific biomarkers (3840). Moreover, prior studies have already confirmed the predicting value of some clinical/pathological characteristics, including molecular subtypes, imaging response, and chemotherapy regimens (16, 17, 41). In the exploratory analysis of one of the included trials, Kuerer et al. reported that combining imaging appearances and MIB results can achieve an even higher predicting accuracy for pCR (23). With more and more research data accumulated, it is possible to incorporate multiple potentially relevant factors into a predictive model, which would dramatically improve the accuracy of confirming residual tumors after NST. In the construction of a predictive model with multiple parameters, artificial intelligence can play an important role (4244). Only when a reliable non-surgical tool to rule out residual disease has been developed can omission of breast surgery be clinically feasible and safe.

The main benefit of omitting breast surgery is avoiding potential complications, better aesthetic appearance, and higher quality of life. Considering the current unaccepted accuracy of the MIB method and high possibility of missing residual disease, the benefit of surgery omission should not be achieved at the cost of oncologic safety. Furthermore, with the development of oncoplastic surgery, surgical complications, and the appearance deficits have increased significantly (45, 46). Breast surgery cannot be replaced in excellent responders of NST until the accuracy of MIB has improved to a reliable level.

There are some limitations to the present meta-analysis. First, the participants of included trials had various clinical and pathological characteristics, and these different characteristics, including age, molecular subtype, response to NST, and chemotherapy regimens, will inevitably contribute to heterogeneity. We performed meta-regressions to investigate the effects of some of these factors on pCR prediction. Second, our pooled analysis included trials with diverse MIB procedures, thereby causing bias. However, as mentioned above, there was not enough information about biopsy procedures from the included trials to make further subgroup analyses. Third, we extracted data from conference abstracts of some of the included trials, and thus, the information was not complete and may increase the difficulty of data extraction and quality assessment.

In conclusion, current image-guided MIB methods are not accurate enough in terms of predicting breast pCR after NST for breast cancer patients. The predicting accuracy is not significantly different among different molecular subtypes of breast cancer. Including patients with only cCR to NST may lead to worse prediction accuracy. It is of utmost clinical importance to standardize the MIB procedure and incorporate other factors into the evaluation in order to reduce the FNR to an acceptable level.

Data Availability Statement

All datasets presented in this study are included in the article/supplementary files.

Author Contributions

YLi, YZ, FM, and QS: conception and design. YLi and QS: administrative support. YLi, YZ, FM, YLin, XZ, and SS: data analysis and interpretation. All of the authors: manuscript writing and final approval of manuscript.

Funding

This work was supported by the Fundamental Research Funds for the Central Universities (3332019023) and by the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019XK320016).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Ruth Shen, Gillian Li, and Gilbert Li for their enlightening advice.

References

1. Murphy BL, Day CN, Hoskin TL, Habermann EB, Boughey JC. Neoadjuvant chemotherapy use in breast cancer is greatest in excellent responders: triple-negative and HER2+ subtypes. Ann Surg Oncol. (2018) 25:2241–8. doi: 10.1245/s10434-018-6531-5

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Berruti A, Amoroso V, Gallo F, Bertaglia V, Simoncini E, Pedersini R, et al. Pathologic complete response as a potential surrogate for the clinical outcome in patients with breast cancer after neoadjuvant therapy: a meta-regression of 29 randomized prospective studies. J Clin Oncol. (2014) 32:3883–91. doi: 10.1200/JCO.2014.55.2836

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. (2014) 384:164–72. doi: 10.1016/S0140-6736(13)62422-8

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Baselga J, Bradbury I, Eidtmann H, Di Cosimo S, de Azambuja E, Aura C, et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. (2012) 379:633–40. doi: 10.1016/S0140-6736(11)61847-3

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Castrellon AB, Pidhorecky I, Valero V, Raez LE. The role of carboplatin in the neoadjuvant chemotherapy treatment of triple negative breast cancer. Oncol Rev. (2017) 11:324. doi: 10.4081/oncol.2017.324

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. (2017) 376:2147–59. doi: 10.1056/NEJMoa1612645

PubMed Abstract | CrossRef Full Text | Google Scholar

7. von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. (2019) 380:617–28. doi: 10.1056/NEJMoa1814017

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Kummel S, Holtschmidt J, Loibl S. Surgical treatment of primary breast cancer in the neoadjuvant setting. Br J Surg. (2014) 101:912–24. doi: 10.1002/bjs.9545

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Teshome M, Kuerer HM. Breast conserving surgery and locoregional control after neoadjuvant chemotherapy. Eur J Surg Oncol. (2017) 43:865–74. doi: 10.1016/j.ejso.2017.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kuerer HM, Vrancken Peeters M, Rea DW, Basik M, De Los Santos J, Heil J. Nonoperative management for invasive breast cancer after neoadjuvant systemic therapy: conceptual basis and fundamental international feasibility clinical trials. Ann Surg Oncol. (2017) 24:2855–62. doi: 10.1245/s10434-017-5926-z

PubMed Abstract | CrossRef Full Text | Google Scholar

11. van la Parra RF, Kuerer HM. Selective elimination of breast cancer surgery in exceptional responders: historical perspective and current trials. Breast Cancer Res. (2016) 18:28. doi: 10.1186/s13058-016-0684-6

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Savaridas SL, Sim YT, Vinnicombe SJ, Purdie CA, Thompson AM, Evans A. Are baseline ultrasound and mammographic features associated with rates of pathological completes response in patients receiving neoadjuvant chemotherapy for breast cancer? Cancer Imaging. (2019) 19:67. doi: 10.1186/s40644-019-0251-3

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Schaefgen B, Mati M, Sinn HP, Golatta M, Stieber A, Rauch G, et al. Can routine imaging after neoadjuvant chemotherapy in breast cancer predict pathologic complete response? Ann Surg Oncol. (2016) 23:789–95. doi: 10.1245/s10434-015-4918-0

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Fowler AM, Mankoff DA, Joe BN. Imaging neoadjuvant therapy response in breast cancer. Radiology. (2017) 285:358–75. doi: 10.1148/radiol.2017170180

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Negrao EMS, Bitencourt AGV, de Souza JA, Marques EF. Accuracy of breast magnetic resonance imaging in evaluating the response to neoadjuvant chemotherapy: a study of 310 cases at a cancer center. Radiol Brasileira. (2019) 52:299–304. doi: 10.1590/0100-3984.2018.0149

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Li H, Yao L, Jin P, Hu L, Li X, Guo T, et al. MRI and PET/CT for evaluation of the pathological response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis. Breast. (2018) 40:106–15. doi: 10.1016/j.breast.2018.04.018

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Rauch GM, Adrada BE, Kuerer HM, van la Parra RF, Leung JW, Yang WT. Multimodality imaging for evaluating response to neoadjuvant chemotherapy in breast cancer. AJR Am J Roentgenol. (2017) 208:290–9. doi: 10.2214/AJR.16.17223

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Heil J, Pfob A, Sinn H-PP, Rauch G, Bach P, Schaefgen B, et al. Abstract GS5-03: diagnosing residual disease and pathologic complete response after neoadjuvant chemotherapy in breast cancer patients by image-guided vacuum-assisted breast biopsy: results of a prospective multicenter trial. Cancer Res. (2020) 80(4 Suppl): GS5-03. doi: 10.1158/1538-7445.SABCS19-GS5-03

CrossRef Full Text | Google Scholar

19. Basik M, Cecchini RS, Santos JFDL, Umphrey HR, Julian TB, Mamounas EP, et al. Abstract GS5-05: primary analysis of NRG-BR005, a phase II trial assessing accuracy of tumor bed biopsies in predicting pathologic complete response (pCR) in patients with clinical/radiological complete response after neoadjuvant chemotherapy (NCT) to explore the feasibility of breast-conserving treatment without surgery. Cancer Res. (2020) 80(4 Suppl):GS5-05. doi: 10.1158/1538-7445.SABCS19-GS5-05

CrossRef Full Text | Google Scholar

20. Peeters M-JTFDV, van Loevezijn A, van der Noordaa ME, van Duijnhoven FH, Loo CE, van Werkhoven E, et al. Abstract GS5-06: towards omitting breast surgery in patients with a pathologic complete response after neoadjuvant systemic treatment: interim analysis of the MICRA trial (minimally invasive complete response assessment). Cancer Res. (2020) 80(Suppl. 4): GS5-06. doi: 10.1158/1538-7445.SABCS19-GS5-06

CrossRef Full Text | Google Scholar

21. Francis A, Herring K, Molyneux R, Jafri M, Trivedi S, Shaaban A, et al. Abstract P5-16-14: NOSTRA PRELIM: a non randomised pilot study designed to assess the ability of image guided core biopsies to detect residual disease in patients with early breast cancer who have received neoadjuvant chemotherapy to inform the design of a planned trial. Cancer Res. (2017) 77(Suppl. 4):P5-16-14. doi: 10.1158/1538-7445.SABCS16-P5-16-14

CrossRef Full Text | Google Scholar

22. Heil J, Schaefgen B, Sinn P, Richter H, Harcos A, Gomez C, et al. Can a pathological complete response of breast cancer after neoadjuvant chemotherapy be diagnosed by minimal invasive biopsy? Eur J Cancer. (2016) 69:142–50. doi: 10.1016/j.ejca.2016.09.034

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Kuerer HM, Rauch GM, Krishnamurthy S, Adrada BE, Caudle AS, DeSnyder SM, et al. A clinical feasibility trial for identification of exceptional responders in whom breast cancer surgery can be eliminated following neoadjuvant systemic therapy. Ann Surg. (2018) 267:946–51. doi: 10.1097/SLA.0000000000002313

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Heil J, Kummel S, Schaefgen B, Paepke S, Thomssen C, Rauch G, et al. Diagnosis of pathological complete response to neoadjuvant chemotherapy in breast cancer by minimal invasive biopsy techniques. Br J Cancer. (2015) 113:1565–70. doi: 10.1038/bjc.2015.381

PubMed Abstract | CrossRef Full Text | Google Scholar

25. van der Noordaa MEM, van Duijnhoven FH, Loo CE, van Werkhoven E, van de Vijver KK, Wiersma T, et al. Identifying pathologic complete response of the breast after neoadjuvant systemic therapy with ultrasound guided biopsy to eventually omit surgery: study design and feasibility of the MICRA trial (minimally invasive complete response assessment). Breast. (2018) 40:76–81. doi: 10.1016/j.breast.2018.04.015

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Tasoulis MK, Roche N, Rusby JE, Pope R, Allen S, Downey K, et al. Post neoadjuvant chemotherapy vacuum assisted biopsy in breast cancer: can it determine pathologic complete response before surgery? J Clin Oncol. (2018) 36(Suppl. 15):567. doi: 10.1200/JCO.2018.36.15_suppl.567

CrossRef Full Text | Google Scholar

27. Lee H-B, Kim S-Y, Kim KE, Park JH, Ju YW, Lee E, et al. Prediction of pathologic complete response by image-guided biopsy before surgery in breast cancer with complete clinical response to neoadjuvant chemotherapy: a prospective feasibility trial. J Clin Oncol. (2018) 36(Suppl. 15):566. doi: 10.1200/JCO.2018.36.15_suppl.566

CrossRef Full Text | Google Scholar

28. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. (2009) 339:b2700. doi: 10.1136/bmj.b2700

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Song F, Eastwood AJ, Gilbody S, Duley L, Sutton AJ. Publication and related biases. Health Technol Assess. (2000) 4:1–115. doi: 10.3310/hta4100

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Early Breast Cancer Trialists' Collaborative G. Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. (2018) 19:27–39. doi: 10.1016/S1470-2045(17)30777-5

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Ring A, Webb A, Ashley S, Allum WH, Ebbs S, Gui G, et al. Is surgery necessary after complete clinical remission following neoadjuvant chemotherapy for early breast cancer? J Clin Oncol. (2003) 21:4540–5. doi: 10.1200/JCO.2003.05.208

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Daveau C, Savignoni A, Abrous-Anane S, Pierga JY, Reyal F, Gautier C, et al. Is radiotherapy an option for early breast cancers with complete clinical response after neoadjuvant chemotherapy? Int J Radiat Oncol Biol Phys. (2011) 79:1452–9. doi: 10.1016/j.ijrobp.2010.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Ellis P, Smith I, Ashley S, Walsh G, Ebbs S, Baum M, et al. Clinical prognostic and predictive factors for primary chemotherapy in operable breast cancer. J Clin Oncol. (1998) 16:107–14. doi: 10.1200/JCO.1998.16.1.107

PubMed Abstract | CrossRef Full Text | Google Scholar

34. El Hage Chehade H, Headon H, El Tokhy O, Heeney J, Kasem A, Mokbel K. Is sentinel lymph node biopsy a viable alternative to complete axillary dissection following neoadjuvant chemotherapy in women with node-positive breast cancer at diagnosis? An updated meta-analysis involving 3,398 patients. Am J Surg. (2016) 212:969–81. doi: 10.1016/j.amjsurg.2016.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. (2010) 11:927–33. doi: 10.1016/S1470-2045(10)70207-2

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kuerer H. Eliminating Breast Cancer Surgery in Exceptional Responders with Neoadjuvant Systemic Therapy [ClinicalTrials.gov] (2017). Available online at: https://clinicaltrialsgov/ct2/show/NCT02945579 (accessed February 14, 2020).

Google Scholar

37. Fukada I, Araki K, Kobayashi K, Shibayama T, Takahashi S, Gomi N, et al. Pattern of tumor shrinkage during neoadjuvant chemotherapy is associated with prognosis in low-grade luminal early breast cancer. Radiology. (2018) 286:49–57. doi: 10.1148/radiol.2017161548

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Bownes RJ, Turnbull AK, Martinez-Perez C, Cameron DA, Sims AH, Oikonomidou O. On-treatment biomarkers can improve prediction of response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. (2019) 21:73. doi: 10.1186/s13058-019-1159-3

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Datta J, Berk E, Xu S, Fitzpatrick E, Rosemblit C, Lowenfeld L, et al. Anti-HER2 CD4+ T-helper type 1 response is a novel immune correlate to pathologic response following neoadjuvant therapy in HER2-positive breast cancer. Breast Cancer Res. (2015) 17:71. doi: 10.1186/s13058-015-0584-1

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. (2018) 19:40–50. doi: 10.1016/S1470-2045(17)30904-X

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Jones RL, Lakhani SR, Ring AE, Ashley S, Walsh G, Smith IE. Pathological complete response and residual DCIS following neoadjuvant chemotherapy for breast carcinoma. Br J Cancer. (2006) 94:358–62. doi: 10.1038/sj.bjc.6602950

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Cain EH, Saha A, Harowicz MR, Marks JR, Marcom PK, Mazurowski MA. Multivariate machine learning models for prediction of pathologic response to neoadjuvant therapy in breast cancer using MRI features: a study using an independent validation set. Breast Cancer Res Treat. (2019) 173:455–63. doi: 10.1007/s10549-018-4990-9

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Cheng Q, Huang J, Liang J, Ma M, Ye K, Shi C, et al. The diagnostic performance of DCE-MRI in evaluating the pathological response to neoadjuvant chemotherapy in breast cancer: a meta-analysis. Front Oncol. (2020) 10:93. doi: 10.3389/fonc.2020.00093

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Lo Gullo R, Eskreis-Winkler S, Morris EA, Pinker K. Machine learning with multiparametric magnetic resonance imaging of the breast for early prediction of response to neoadjuvant chemotherapy. Breast. (2020) 49:115–22. doi: 10.1016/j.breast.2019.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Dahlback C, Ullmark JH, Rehn M, Ringberg A, Manjer J. Aesthetic result after breast-conserving therapy is associated with quality of life several years after treatment. Swedish women evaluated with BCCT.core BREAST-Q. Breast Cancer Res Treat. (2017) 164:679–87. doi: 10.1007/s10549-017-4306-5

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Papanikolaou IG, Dimitrakakis C, Zagouri F, Marinopoulos S, Giannos A, Zografos E, et al. Paving the way for changing perceptions in breast surgery: a systematic literature review focused on oncological and aesthetic outcomes of oncoplastic surgery for breast cancer. Breast Cancer. (2019) 26:416–27. doi: 10.1007/s12282-019-00968-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: breast cancer, neoadjuvant, minimal invasive biopsy, pathologic complete response, meta-analysis

Citation: Li Y, Zhou Y, Mao F, Lin Y, Zhang X, Shen S and Sun Q (2020) The Diagnostic Performance of Minimally Invasive Biopsy in Predicting Breast Pathological Complete Response After Neoadjuvant Systemic Therapy in Breast Cancer: A Meta-Analysis. Front. Oncol. 10:933. doi: 10.3389/fonc.2020.00933

Received: 21 March 2020; Accepted: 12 May 2020;
Published: 26 June 2020.

Edited by:

Noam Falbel Pondé, A.C.Camargo Cancer Center, Brazil

Reviewed by:

Miguel J. Gil Gil, Catalan Institute of Oncology, Spain
Simona Ruxandra Volovat, Grigore T. Popa University of Medicine and Pharmacy, Romania

Copyright © 2020 Li, Zhou, Mao, Lin, Zhang, Shen and Sun. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Qiang Sun, sunqiangpumch@126.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.