Back to Journals » Breast Cancer: Targets and Therapy » Volume 17
Promising Response to Neoadjuvant Chemotherapy Plus Immunotherapy in Metaplastic Breast Carcinoma
Authors Tan Q , Li N, Wang Y, Du T, Liang G, Zhao Z, Tang J, Wu H
Received 10 January 2025
Accepted for publication 13 May 2025
Published 23 May 2025 Volume 2025:17 Pages 447—454
DOI https://doi.org/10.2147/BCTT.S512790
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Pooja Advani
Qingjian Tan,* Na Li,* Yan Wang, Tian Du, Gehao Liang, Zixuan Zhao, Jun Tang, Hao Wu
Department of Breast Cancer, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Jun Tang, Email [email protected] Hao Wu, Email [email protected]
Purpose: Metaplastic breast carcinoma (MpBC) is a rare and aggressive subtype of breast cancer that often shows poor response to conventional neoadjuvant chemotherapy (NAC). This study aimed to evaluate the efficacy of combining NAC with immune checkpoint inhibitors (ICIs) in MpBC patients.
Methods: We conducted a retrospective analysis of MpBC patients treated with NAC, with or without the addition of immunotherapy, at Sun Yat-sen university Cancer center between 2017 and 2024. We assessed clinical and pathological response to NAC in MpBC patients.
Results: 40 MpBC patients treated with NAC were identified, 33 patients treated with NAC alone, 7 patients treated with NAC and immunotherapy, 4 (10%) patients achieved pCR. Among the 33 patients treated with NAC alone, only 2 (6%) achieved pCR. In contrast, 7 patients received additional immunotherapy, and 3 started immunotherapy at the initiation of NAC, with 2 of these (67%) achieving pCR. Patients who received immunotherapy after disease progression on NAC showed varying degrees of tumor response, from stable disease (SD) to partial response (PR).
Conclusion: We observed a promising response on addition of immunotherapy to NAC among patients with MpBC, suggesting that immunotherapy may have great potential in the treatment of metaplastic breast carcinoma.
Keywords: metaplastic breast carcinoma, neoadjuvant chemotherapy, pathological complete response, immunotherapy
Introduction
Metaplastic breast carcinoma (MpBC) is a rare and aggressive subtype of breast cancer, accounting for less than 1% of all invasive breast malignancies.1–3 It comprises various histological subtypes, including squamous cell carcinoma, sarcomatoid carcinoma, and chondroid carcinoma, and predominantly affects relatively younger patients. Most MpBC cases are classified as triple-negative breast cancer (TNBC), which is associated with poorer clinical outcomes.4 Ong et al reported that the 5-year overall survival rate for stage I–III MpBC patients is 72.5%, significantly lower than the 87.5% seen in non-MpBC breast cancers (p < 0.001).2 Another retrospective analysis from the US National Cancer Database indicated a 5-year overall survival rate of 63.1% for MpBC patients, further emphasizing the lower survival rates compared to other breast cancer subtypes.5
While neoadjuvant chemotherapy (NAC) is the standard approach for high-risk early-stage TNBC, MpBC has shown a significantly lower response to conventional NAC.6–15 Studies have demonstrated that the pathological complete response (pCR) rate for MpBC remains low, typically between 10–15%,8,9,11–15 which is considerably lower than that of other breast cancer subtypes. This limited response to NAC highlights the need for alternative therapeutic strategies to improve outcomes in MpBC patients.
Immune checkpoint inhibitors, particularly PD-1/PD-L1 inhibitors, have demonstrated efficacy in several highly immunogenic cancers, including non-small cell lung cancer, melanoma, and TNBC.16–18 Given MpBC’s poor response to standard chemotherapy and its immunogenic potential, there is growing interest in exploring the role of immunotherapy in this subtype.
The objective of this study is to evaluate the impact of combining NAC with immune checkpoint inhibitors in MpBC patients, with a focus on improving pCR rates and treatment outcomes.
Materials and Methods
This study was conducted in accordance with the principles of the Declaration of Helsinki and was approved by the Institutional Review Board (IRB) of Sun Yat-sen University Cancer Center. The permit number was B2024-672-01.Given that our research is a retrospective study utilizing a subset of patient medical records, it meets the following criteria: the risk to participants is minimal, and patient confidentiality will be fully protected. The inclusion of the relevant patient population is rare, and obtaining informed consent in this context would not be practical. Furthermore, previous studies had already obtained written consent from participants for the use of their medical records in additional research endeavors. In compliance with ethical guidelines, we submitted a request to the Ethics Committee of Sun Yat-sen University Cancer Center for a waiver of informed consent, which was subsequently approved.
Patient Cohort
Eligible participants in the study had a definitive diagnosis of MpBC, confirmed through pathological examination of either postsurgical specimens or core biopsy samples, with no evidence of distant metastasis. All participants received chemotherapy before surgery. There were no restrictions on sex or age for patient eligibility in this study. From 2017 to 2024, we identified 51 patients with MpBC who received NAC followed by surgery in our institutional database. Eleven patients were excluded from the analysis; a summary of the patient screening process is shown in Figure 1. Finally, a total of 40 patients were deemed eligible for inclusion in the study. Data were extracted from the patients’ medical charts, including age at diagnosis, tumor stage (tumor size and local lymph node involvement), presence of ductal carcinoma components, Ki-67 proliferative index, hormonal receptor status, HER2 status, and types of systemic treatment, radiotherapy, and surgery.
Pathologic Evaluation
Clinical response was assessed through both clinical examination and radiologic evaluation using RECIST criteria.19 Pathologic complete response (pCR) is defined as the absence of invasive components in both the primary tumor and lymph nodes, regardless of any residual ductal carcinoma in situ (ypT0/pTis ypN0). Assessment of estrogen receptor, progesterone receptor, and HER2 status was conducted according to ASCO/CAP guidelines.20,21
Result
Clinicopathological Characteristics
The clinicopathologic characteristics of patients prior to NAC, along with their treatment plans, are summarized in Table 1. Among the 40 female patients, the median age at diagnosis was 44 years (range: 27–70 years), and the median tumor size was 4.6 cm (range: 1.8–16 cm). Prior to receiving NAC, 68% (n=27) of the patients were diagnosed with triple-negative breast cancer (TNBC), and 85% (n=34) had clinical T2 or T3 stage tumors, with 65% (n=26) classified as clinical stage III. The median Ki-67 proliferation index was 54% (range: 5–90%), with 97% (n=39) of patients showing a Ki-67 index of ≥20%.
![]() |
Table 1 Clinicopathological Characteristics and Treatment |
Overall Efficacy of NAC and Immunotherapy
Out of the 40 patients, 33 received NAC alone, while 7 received NAC combined with immunotherapy. Among the 33 patients who underwent NAC alone, only 2 (6%) achieved a pCR (Figure 2). In contrast, of the 7 patients who received combined immunotherapy, 3 had immunotherapy initiated at the beginning of treatment, and 2 of these (67%) achieved pCR. The remaining 4 patients experienced disease progression during NAC but exhibited varying degrees of response after the addition of immunotherapy, ranging from stable disease (SD) to partial response (PR) (Figure 3). The detailed regimens for the 7 patients are presented in Table 2.
![]() |
Table 2 Tumor Size; Patient Age; and Neoadjuvant Treatment Regimens |
Surgical and Pathological Evaluation
All patients underwent surgery following chemotherapy. Of these, 37 patients (92.5%) received a mastectomy with axillary lymph node dissection (ALND), and 3 successfully underwent breast-conserving surgery (BCS). Out of the 40 patients, 4 (10%) achieved pCR after NAC, including 3 patients with clinical stage II and 1 patient with clinical stage III. Additionally, 1 patient achieved pCR in the breast tissue but had residual metastatic disease in the axillary lymph nodes.
Overall pCR Performance
In total, 4 patients (10%) from the entire cohort achieved pCR, consistent with the low pCR rates reported in other studies on MpBC. Achieving pCR was associated with earlier clinical stages, suggesting that combining NAC with immunotherapy may be more effective in patients with early-stage disease.
Discussion
Metaplastic breast carcinoma (MpBC) is a rare and highly heterogeneous subtype of breast cancer, often presenting with larger tumors and a poorer response to standard therapies, including neoadjuvant chemotherapy (NAC).22–24 Consistent with previous studies,8,9,11–15 our findings confirm the low pathological complete response (pCR) rate in MpBC when treated with NAC alone. In our cohort, only 6% of patients achieved pCR following NAC without immunotherapy, highlighting the limited efficacy of chemotherapy in this subtype.
However, the addition of immune checkpoint inhibitors (ICIs) to NAC in our study led to a notable improvement in pCR rates, particularly in patients with early-stage disease. Among the 7 patients who received immunotherapy, 3 had immunotherapy initiated at the beginning of treatment, and 2 of 3 (67%) achieved pCR. This suggests that combining NAC with immunotherapy may enhance the tumor’s response, aligning with findings from studies such as the KEYNOTE-522 trial, which demonstrated improved pCR rates and OS in TNBC with the addition of pembrolizumab.25–27
MpBC shares several characteristics with TNBC, including a lack of hormone receptor expression and HER-2 negativity, which may explain its potential responsiveness to immunotherapy. Studies have shown high levels of PD-L1 expression28–30 and increased tumor-infiltrating lymphocytes (TILs)31 in MpBC, most research suggests that elevated PD-L1 expression correlates with higher pathological complete response (pCR) rates and improved prognosis.32 Additionally, the presence of TILs, particularly CD8+ lymphocytes, has been linked to better responses to immunotherapy in various cancers, including TNBC,33 further supporting the use of ICIs in this subtype. Our results suggest that patients with earlier clinical stages (stage II) may benefit the most from this approach, as evidenced by the higher pCR rates observed in this group.
Patients with MpBC often present with advanced tumor stages and larger tumor sizes at diagnosis. In our study, the average maximum tumor diameter at presentation was 5.3 cm, and most patients were not eligible for BCS. Only 3 out of 40 patients (7.5%) opted for BCS. Among the 7 patients who received immunotherapy, one successfully underwent BCS with negative margins and achieved a pathological complete response (pCR). Previous studies have reported the feasibility and safety of BCS in patients with clinical T3–T4 breast cancer following neoadjuvant chemotherapy, challenging the traditional preference for mastectomy in this subgroup. Given the promising response to immunotherapy observed in our study, it raises the question of whether more patients could become eligible for BCS while maintaining oncologic safety and achieving better cosmetic outcomes. However, further research is needed to explore this possibility.
The observed pCR rate of 28.6% (2/7) with neoadjuvant immunotherapy in MpBC surpasses historical chemotherapy rates (5–10%),9 yet remains lower than HER2+ breast cancer (50–70% with anti-HER2 therapy34 and non-metaplastic TNBC (60–70% in KEYNOTE-522.18 While direct cross-trial comparisons are confounded by biological heterogeneity, these contrasts emphasize the need for subtype-tailored strategies. Our findings provide the first benchmark for immunotherapy efficacy in MpBC, distinct from conventional TNBC paradigms.
The clinical significance of these findings is substantial. Achieving pCR has been associated with improved long-term outcomes, including better survival rates.27,35,36 Thus, incorporating immunotherapy into the neoadjuvant setting for MpBC could represent a promising therapeutic strategy, particularly for early-stage patients.
Our study is a retrospective analysis. Due to the rarity of patients with metaplastic breast carcinoma (MBC) and the diagnostic challenges associated with preoperative needle biopsy, the number of cases in our study is relatively small, making it difficult to standardize chemotherapy regimens. Further prospective studies are warranted to confirm these findings and explore the full potential of immunotherapy in MpBC. While the KEYNOTE-522 study has demonstrated the efficacy of immunotherapy in TNBC, not all metaplastic carcinomas are TNBC, and this subgroup has not been specifically analyzed. Our study provides preliminary evidence supporting the use of ICIs in combination with NAC as a viable treatment option for improving outcomes in this rare and aggressive breast cancer subtype.
Conclusion
In conclusion, our real-world, single-center data provide detailed insights into the treatments received by nonmetastatic MpBC patients. We found that combining neoadjuvant chemotherapy (NAC) with immunotherapy may improve the pCR rate and may lead to better treatment outcomes, particularly in patients at relatively early stages.
Data Sharing Statement
The datasets generated during and analysed during the current study are not publicly available due to ethical concerns, the data used in this study cannot be made publicly available, but are available from the corresponding author on reasonable request.
Ethical Approval
This study received approval from the Institutional Review Board of Sun Yat-sen University Cancer Center.
Author Contributions
All authors made a significant contribution to the work reported, whether in the conception, study design, execution, acquisition of data, analysis, and interpretation, or in all these areas, took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This research was funded by the National Natural Science Foundation of China (Grant Nos. 82203463 and 82203569, Hao Wu) and the Department of Science and Technology Basic and Applied Basic Research Special Fund in Guangzhou (Grant No. SL2022A04J01651).
Disclosure
The authors have no conflicts of interest relevant to this article. This paper has been uploaded to ResearchGate as a preprint: https://www.researchgate.net/publication/386386000_Promising_Response_to_Neoadjuvant_Chemotherapy_Plus_Immunotherapy_in_Metaplastic_Breast_Carcinoma
References
1. Moreno AC, Lin YH, Bedrosian I, Shen Y, Babiera GV, Shaitelman SF. Outcomes after treatment of metaplastic versus other breast cancer subtypes. J Cancer. 2020;11(6):1341–1350. doi:10.7150/jca.40817
2. Ong CT, Campbell BM, Thomas SM, et al. Metaplastic breast cancer treatment and outcomes in 2500 patients: a retrospective analysis of a national oncology database. Ann Surg Oncol. 2018;25(8):2249–2260. doi:10.1245/s10434-018-6533-3
3. Polamraju P, Haque W, Cao K, et al. Comparison of outcomes between metaplastic and triple-negative breast cancer patients. Breast. 2020;49:8–16. doi:10.1016/j.breast.2019.10.003
4. Ullah A, Khan J, Yasinzai AQK, et al. Metaplastic breast carcinoma in U.S. population: racial disparities, survival benefit of adjuvant chemoradiation and future personalized treatment with genomic landscape. Cancers. 2023;15(11):2954. doi:10.3390/cancers15112954
5. Elimimian EB, Samuel TA, Liang H, Elson L, Bilani N, Nahleh ZA. Clinical and demographic factors, treatment patterns, and overall survival associated with rare triple-negative breast carcinomas in the US. JAMA Network Open. 2021;4(4):e214123. doi:10.1001/jamanetworkopen.2021.4123
6. Hashmi AA, Aijaz S, Mahboob R, et al. Clinicopathologic features of invasive metaplastic and micropapillary breast carcinoma: comparison with invasive ductal carcinoma of breast. BMC Res Notes. 2018;11(1):531. doi:10.1186/s13104-018-3623-z
7. Tseng WH, Martinez SR. Metaplastic breast cancer: to radiate or not to radiate? Ann Surg Oncol. 2011;18(1):94–103. doi:10.1245/s10434-010-1198-6
8. Ladipo OL, Ren Y, Caddell KB, Sampathkumar A, Almond CA, Fayanju OM. Does treatment sequence affect outcomes in patients with metaplastic breast cancer? Am J Surg. 2021;221(4):701–705. doi:10.1016/j.amjsurg.2021.01.007
9. Haque W, Verma V, Schwartz MR, et al. Neoadjuvant chemotherapy for metaplastic breast cancer: response rates, management, and outcomes. Clin Breast Cancer. 2022;22(5):e691–e699. doi:10.1016/j.clbc.2022.01.006
10. Wong W, Brogi E, Reis-Filho JS, et al. Poor response to neoadjuvant chemotherapy in metaplastic breast carcinoma. Npj Breast Cancer. 2021;7(1):96. doi:10.1038/s41523-021-00302-z
11. Cimino-Mathews A, Verma S, Figueroa-Magalhaes MC, et al. A clinicopathologic analysis of 45 patients with metaplastic breast carcinoma. Am J Clin Pathol. 2016;145(3):365–372. doi:10.1093/ajcp/aqv097
12. Han M, Salamat A, Zhu L, et al. Metaplastic breast carcinoma: a clinical-pathologic study of 97 cases with subset analysis of response to neoadjuvant chemotherapy. Mod Pathol. 2019;32(6):807–816. doi:10.1038/s41379-019-0208-x
13. Corso G, Frassoni S, Girardi A, et al. Metaplastic breast cancer: prognostic and therapeutic considerations. J Surg Oncol. 2021;123(1):61–70. doi:10.1002/jso.26248
14. Tadros AB, Sevilimedu V, Giri DD, Zabor EC, Morrow M, Plitas G. Survival outcomes for metaplastic breast cancer differ by histologic subtype. Ann Surg Oncol. 2021;28(8):4245–4253. doi:10.1245/s10434-020-09430-5
15. Al-Hilli Z, Choong G, Keeney MG, et al. Metaplastic breast cancer has a poor response to neoadjuvant systemic therapy. Breast Cancer Res Treat. 2019;176(3):709–716. doi:10.1007/s10549-019-05264-2
16. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. New Engl J Med. 2015;372(26):2521–2532. doi:10.1056/NEJMoa1503093
17. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. New Engl J Med. 2015;373(19):1803–1813. doi:10.1056/NEJMoa1510665
18. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for early triple-negative breast cancer. New Engl J Med. 2020;382(9):810–821. doi:10.1056/NEJMoa1910549
19. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45(2):228–247. doi:10.1016/j.ejca.2008.10.026
20. Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and progesterone receptor testing in breast cancer: ASCO/CAP guideline update. J clin oncol. 2020;38(12):1346–1366. doi:10.1200/JCO.19.02309
21. Wolff AC, Hammond MEH, Allison KH, et al. Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline focused update. Arch Pathol Lab Med. 2018;142(11):1364–1382. doi:10.5858/arpa.2018-0902-SA
22. Wu K, Yang Q, Liu Y, Wu A, Yang Z. Meta-analysis on the association between pathologic complete response and triple-negative breast cancer after neoadjuvant chemotherapy. World J Surg Oncol. 2014;12:95. doi:10.1186/1477-7819-12-95
23. Thomas HR, Hu B, Boyraz B, et al. Metaplastic breast cancer: a review. Crit rev oncol/hematol. 2023;182:103924. doi:10.1016/j.critrevonc.2023.103924
24. Pezzi CM, Patel-Parekh L, Cole K, Franko J, Klimberg VS, Bland K. Characteristics and treatment of metaplastic breast cancer: analysis of 892 cases from the national cancer data base. Ann Surg Oncol. 2007;14(1):166–173. doi:10.1245/s10434-006-9124-7
25. Cetin B, Gumusay O. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(26):e108. doi:10.1056/NEJMc2006684
26. Schmid P, Cortes J, Dent R, et al. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386(6):556–567. doi:10.1056/NEJMoa2112651
27. Schmid P, Cortes J, Dent R, et al. Overall survival with pembrolizumab in early-stage triple-negative breast cancer. N Engl J Med. 2024;391(21):1981–1991. doi:10.1056/NEJMoa2409932
28. Salisbury T, Abozina A, Zhang C, et al. Histological subtype is associated with PD-L1 expression and CD8+ T-cell infiltrates in triple-negative breast carcinoma. Ann Diagn Pathol. 2022;57:151901. doi:10.1016/j.anndiagpath.2022.151901
29. Grabenstetter A, Jungbluth AA, Frosina D, et al. PD-L1 expression in metaplastic breast carcinoma using the PD-L1 SP142 assay and concordance among PD-L1 immunohistochemical assays. Am J Surg Pathol. 2021;45(9):1274–1281. doi:10.1097/PAS.0000000000001760
30. Joneja U, Vranic S, Swensen J, et al. Comprehensive profiling of metaplastic breast carcinomas reveals frequent overexpression of programmed death-ligand 1. J Clin Pathol. 2017;70(3):255–259. doi:10.1136/jclinpath-2016-203874
31. Lien HC, Lee YH, Chen IC, et al. Tumor-infiltrating lymphocyte abundance and programmed death-ligand 1 expression in metaplastic breast carcinoma: implications for distinct immune microenvironments in different metaplastic components. Virchows Archiv. 2021;478(4):669–678. doi:10.1007/s00428-020-02954-x
32. Kumar S, Chatterjee M, Ghosh P, Ganguly KK, Basu M, Ghosh MK. Targeting PD-1/PD-L1 in cancer immunotherapy: an effective strategy for treatment of triple-negative breast cancer (TNBC) patients. Genes Dis. 2023;10(4):1318–1350.
33. Evangelou Z, Papoudou-Bai A, Karpathiou G, et al. PD-L1 expression and tumor-infiltrating lymphocytes in breast cancer: clinicopathological analysis in women younger than 40 years old. vivo. 2020;34(2):639–647. doi:10.21873/invivo.11818
34. Takada M, Toi M. Neoadjuvant treatment for HER2-positive breast cancer. Chin clin oncol. 2020;9(3):32. doi:10.21037/cco-20-123
35. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–172. doi:10.1016/S0140-6736(13)62422-8
36. Shepherd JH, Ballman K, Polley MC, et al. CALGB 40603 (Alliance): long-term outcomes and genomic correlates of response and survival after neoadjuvant chemotherapy with or without carboplatin and bevacizumab in triple-negative breast cancer. J Clin Oncol. 2022;40(12):1323–1334. doi:10.1200/JCO.21.01506
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

A Case of Complete Remission in Proficient Mismatch Repair (pMMR) Advanced Colon Cancer Treated with Sintilimab and XELOX
Zhu J, Li G, Zhang Z, Wang Y
ImmunoTargets and Therapy 2023, 12:17-23
Published Date: 19 February 2023
Predictive Value of Pretreatment Neutrophil to Albumin Ratio in Response to Neoadjuvant Chemotherapy of Breast Cancer
Deng YX, Zhao YJ, Nong QH, Qiu HM, Guo QL, Hu H
Breast Cancer: Targets and Therapy 2024, 16:393-402
Published Date: 23 July 2024