Narrative review on the evolving role of HER2/neu targeting in uterine serous cancers
Review Article | Data-Driven Clinical Practice and Policy Making

Narrative review on the evolving role of HER2/neu targeting in uterine serous cancers

Aarthi S. Jayraj1 ORCID logo, Salamatu Abdul-Aziz1, Anisa Mburu2 ORCID logo, Avinash Upadhyay3 ORCID logo, Nilanchali Singh4 ORCID logo, Prafull Ghatage5 ORCID logo

1Department of Gynaecologic Oncology, James Cook University Hospital, South Tees NHS Foundation Trust, Middlesbrough, UK; 2Department of Gynaecologic Oncology, Aga Khan Hospital, Mombasa, Kenya; 3Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India; 4Department of Gynaecologic Oncology, All India Institute of Medical Sciences, New Delhi, India; 5Department of Gynecological Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada

Contributions: (I) Conception and design: AS Jayraj, P Ghatage; (II) Administrative support: AS Jayraj, S Abdul-Aziz, P Ghatage; (III) Provision of study materials or patients: AS Jayraj, S Abdul-Aziz; (IV) Collection and assembly of data: AS Jayraj, S Abdul-Aziz, A Mburu; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Prafull Ghatage, FRCSC. Department of Gynecological Oncology, Tom Baker Cancer Centre, 1331-29th Street NW, Calgary, Alberta T2N 4N2, Canada. Email: Prafull.Ghatage@albertahealthservices.ca.

Background and Objective: Serous endometrial cancers (ECs) are an aggressive histotype of ECs which are disproportionately responsible for 40% of cancer-specific mortality rates despite constituting only 5–10% of all uterine cancers in incidence. In recent times, it has become increasingly evident that about 20–40% of uterine serous cancers (USCs) have molecular alterations in ERBB2 pathway with human epidermal growth factor receptor 2 (HER2/neu) amplification or overexpression. We summarise the evidence on genetic and molecular alterations in HER2/neu pathway in USC with a focus on testing criteria, targeting agents and resistance mechanisms.

Methods: We conducted a database search of PubMed/Medline up to 28th February 2023 for articles published in the English language using pre-defined search terms. One hundred and seventy-one relevant articles were subsequently reviewed for eligibility and inclusion in the review.

Key Content and Findings: The Cancer Genome Atlas (TCGA) classification is a significant development in the molecular profiling of ECs with a positive impact on the treatment of these tumors including USCs. Testing criteria for HER2/neu in USC with immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) has evolved in more than a decade with progress made towards EC specific testing guidelines. The findings of a recent phase III study have led to the development of practice changing guidelines towards improving patient outcomes.

Conclusions: Molecular aberration in the HER2/neu pathway contributes to the aggressive behaviour of USC. Considering the clinical benefit conferred by HER2/neu targeted therapy, HER2/neu testing is recommended for all cases of serous EC in advanced and recurrent settings. Trastuzumab in combination with platinum and taxanes based chemotherapy is the recommended treatment option for patients with advanced or recurrent serous cancers who test positive to HER2/neu. Clinical trials on targeted therapy are ongoing and future research should focus on selection of patients who will derive the most benefit from such therapy.

Keywords: Human epidermal growth factor receptor 2 (HER2/neu); targeted therapy; uterine serous cancer (USC); trastuzumab


Submitted Apr 04, 2023. Accepted for publication Sep 15, 2023. Published online Nov 09, 2023.

doi: 10.21037/atm-23-1465


Introduction

Background

Endometrial cancer (EC) is one of the most common cancers affecting women worldwide. In the year 2020, there were over 417,000 new cases of EC and 97,000 deaths globally (1). Uterine serous cancer (USC) is an aggressive histological subtype of EC which accounts for 39% of all deaths due to EC despite constituting only 10% of all ECs in incidence (2). Historically, EC was categorised in to two groups: type I which are predominantly low-grade endometroid tumors and type II consisting of USC, clear cell carcinoma, carcinosarcoma and grade 3 endometroid tumors (3). Similar to endometroid EC, USC usually presents with postmenopausal bleeding but often arises from an endometrial polyp on a background of atrophic endometrium (4). In apparently early-stage disease, there is a tendency for lympho-vascular space invasion (LVSI), lymph node involvement and intra-peritoneal spread despite minimal or no myometrial invasion. All stage 5-year overall survival (OS) of USC is much lower than endometroid EC at 52% vs. 83% respectively (5). Therefore, in contrast to endometroid EC which has an excellent prognosis at early stages of the disease, USC is a high-grade tumor with high recurrence rates and poorer prognosis (6,7).

USC has been associated with increasing age, women of black ethnicity, history of breast cancer and tamoxifen use (8). Contrary to previous data, obesity is reported to be a risk factor for USC although it appears to be hormone independent (8,9). Population studies on EC have demonstrated racial disparities in the incidence, management and outcomes of women affected by the disease. Data suggest that black women are at increased risk of non-endometroid tumors including USC, present at advanced stages and have lower OS rates in comparison with white women (10,11).

Objective

In the recent years, the HER2/neu pathway has emerged as a commonly affected molecular aberration in USCs that contributes to their aggressive nature. Biomarker testing guidelines have not yet been defined for such tumors and the efficacy of HER2/neu targeting agents have been uncertain. The main objective of this article is to review the evidence regarding testing and reporting of HER2/neu amplification/overexpression in USCs and define the role of HER2/neu targeting agents in these cases. We focus on the methods of testing and interpreting HER2/neu overexpression/amplification in USCs. A summary of various preclinical and clinical trials available for HER2/neu targeting agents in USCs, the efficacy of such therapeutic targeting, emerging therapies and future directions have been outlined. We present this article in accordance with the Narrative Review reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-23-1465/rc).


Methods

PubMed/Medline database was searched for articles on HER2/neu testing and targeting in USCs from inception to 28th February 2023. The search terms used were “HER2”, “HER2/neu”, “Her2neu”, “uterine serous cancer”, “uterine papillary serous cancer”, “serous endometrial cancer”, “trastuzumab”, “pertuzumab”, “T-DM1” and “trastuzumab emtansine” (Table 1). Backward and forward reference searching was done on retrieved articles to include any articles which answer the study objectives. Results and references of interim study reports were updated after the date of the initial search when full results were published. The retrieved articles were analysed and summarized in the review (Figure 1).

Table 1

Summary of search strategy

Items Specification
Date of search 28th February 2023
Databases and other sources searched PubMed/Medline
Key search terms used “HER2”, “HER2/neu”, “Her2neu”, “uterine serous cancer”, “uterine papillary serous cancer”, “serous endometrial cancer”, “trastuzumab”, “pertuzumab”, “T-DM1” and “trastuzumab emtansine”
Timeframe Inception to February 2023
Inclusion and exclusion criteria Inclusion: original and review articles in English language with a focus on HER2/neu testing and HER2/neu targeted therapy in USC
Exclusion: studies where main topic is not focussed on USC
Selection process Retrieved articles were reviewed independently for eligibility for inclusion by two reviewers (A.S.J. and S.A.A.). Differences in opinion were resolved by third author (P.G.)

HER2/neu, human epidermal growth factor receptor 2; T-DM1, trastuzumab emtansine; USC, uterine serous cancer.

Figure 1 Literature search strategy. HER2, human epidermal growth factor receptor 2; T-DM1, trastuzumab emtansine.

Discussion

Molecular profile and classification

USCs demonstrate the typical histological features of serous differentiation, papillary architecture, nuclear atypia and pleomorphism, slit-like spaces, scanty cytoplasm and psammoma bodies in one-third of cases (12,13). Historically, the Bokhman’s binary system was used to classify ECs into type I and type II tumors with distinct clinical, pathological and prognostic indicators. The Cancer Genome Atlas (TCGA) classification system categorises EC into four groups: (I) POLE ultra-mutated EC; (II) microsatellite instability hypermutated EC due to dysfunctional DNA mismatch repair proteins; (III) copy number low/microsatellite stable EC which are mostly endometroid (49%); (IV) copy number high with TP53 mutations in >90% and include USC (14). In the study by the TCGA Network, 50 of the 53 (94%) serous tumors analysed were classified under the cluster 4 (copy number high) group and had significantly recurring amplifications of HER2/neu oncogene. This group was also associated with significantly worse progression-free survival (PFS) compared to the endometrioid clusters and was suggested as a potential therapeutic target (15). Several other studies have shown a strong correlation between TP53 mutation and HER2/neu expression and/or amplification in ECs (16,17). Other gene mutations identified in USC include PIK3CA, PPP2R1A, ERBB2 (also known as HER2 and HER2/neu) and CHD4 (12,15,18).

HER2/neu receptor in USC

HER2/neu receptor is a 185-kDa, 1,225-aminoacid transmembrane glycoprotein tyrosine kinase receptor belonging to the epidermal growth factor receptor (EGFR) family encoded on the ErbB-2 gene found on the long arm of chromosome 17 and is also known as Erb-b2, ERBB2, c-erbB-2 or p185HER2. It consists of an extracellular domain which binds with other members of the EGFR family, a membrane spanning hydrophobic transmembrane domain and an intracellular domain responsible for the kinase action (19). HER2/neu receptor is an orphan receptor as it does not have a direct ligand. Instead, it binds with the extracellular domain to other EGFR family ligands activated by EGF ligand interaction. Such homo- or hetero-dimerization leads to activation of the intracellular tyrosine kinase function by autophosphorylation which in turn leads to activation of downstream signaling molecules and results in growth and proliferation. The various downstream pathways that are activated by HER2/neu receptor activation includes Ras/Raf/mitogen-activated protein (MAP) kinase, the PI3K/Akt, and the phospholipase Cγ (PLCγ)/protein kinase C (PKC) pathways. In malignant cells, activation of these mitogenic cell signaling pathways leads to proliferation, invasion, angiogenesis, migration and cell survival (20). HER2/neu mediated tumorigenesis can occur due to overexpression of the proto-oncogene coding for HER2/neu receptor or by overactivation of the receptor, though the former appears to be the most common case in clinical studies (19).

Non oncogenic activity of the HER2/neu receptors is important for the development of organs such as skin, breast, gastrointestinal, reproductive and urinary tracts (21). HER2/neu receptor overexpression has been described in tumorigenesis of a number of cancers including breast, gastric, ovary and uterine serous carcinoma (22,23). In USC, overexpression and/or amplification of HER2/neu by immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) respectively ranges from 16–48% (24-28). HER2/neu positive tumors were associated with advanced disease, myometrial invasion (24,25), lymphovascular space invasion (LVSI) (25,26), higher recurrence rates (24,26), as well as poorer overall and disease-free survival (24-28). In essence, the presence of the HER2/neu oncogene confers a more biologically aggressive disease with poorer prognosis.

There also appears to be a trend towards higher expression and amplification of HER2/neu in black women. Retrospective single centre studies by Santin et al. and Morrison et al. reported HER2/neu amplification in black women in the range of 18–67% (27,29). Similarly, overexpression of HER2/neu in black women ranges from 33–90% in comparison to 13–48% in white women (29,30). These findings have also been correlated with poorer survival in both studies. Further analysis of intensity of IHC staining revealed that heavy (3+) IHC staining in keeping with HER2/neu overexpression was increased in black women and appears to be the strongest prognostic factor for shorter survival (30).


HER2/neu testing and reporting in USC

HER2/neu testing in patients with USC is of paramount importance, as therapeutic decisions and prognostication depends on the accuracy of testing. The criteria for testing in USC were derived from pre-existing standardised protocols for testing in breast and gastric cancers (31-33). HER2/neu protein overexpression on IHC and amplification by FISH or chromogenic in situ hybridization (CISH) have been described in the literature to define HER2/neu positivity in tumor (34-36) (Table 2).

Table 2

HER2/neu amplification and overexpression in uterine papillary serous carcinoma

Study Sample size HER2/neu overexpression by IHC HER2/neu amplification by FISH Concordance between IHC and FISH
Test kit Scoring criteria 2+ 3+ Probe Criteria Incidence
Villella et al., 2006 (37) 17 USC DAKO Herceptest (positive if >5% crisp membranous staining of tumor cells) Negative: 0; focally positive: 1+; strongly positive: 3+ Not described 29.4% (5/17) PathVysion HER2 probe HER2/neu:CEP17 >2.5 36.4% (4/11) 27.3% (3/11)
Morrison et al., 2006 (29) 58 USC, 425 non-serous Ventana Pathway HER2 Negative: 0/1+; positive: 2+/3+ 43.1% (25/58) PathVysion HER2 DNA probe HER2/neu:CEP17 ≥2.2 29.3% (17/58) Not reported
Grushko et al., 2008 (GOG 177) (38) 38 USC and 196 non-serous EC DAKO Herceptest Negative: 0/1+; positive: 2+/3+ 34.2% (13/38) 26.3% (10/38) Path Vision DNA probe Modified ASCO/CAP breast [2007] 21.4% (6/28) Overall: 60%
Xu et al., 2010 (39) 75 pure serous carcinomas + additional 13 pure serous analysed HercepTest Scoring criteria not described 2.7% (2/75), 30.8% (4/13) 9.3% (7/75), 23.1% (3/13) PathVysion DNA probe Modified ASCO/CAP breast [2007] 9/75, 4/13
Fleming et al., 2010 (40) 286 advanced or recurrent EC (only FISH positive tumors included), 11 USC, 22 non-serous DAKO Herceptest Negative: 0/1+; positive: 2+/3+ 61.1% (22/36) 38.9% (14/36) PathVysion HER2 DNA Probe Kit Modified ASCO/CAP breast [2007] 11.5% (33/286) Positive correlation; Spearman’s correlation coefficient =0.354 (95% CI: 0.17–0.51)
Todeschini et al., 2011 (41) 10 USC DAKO Herceptest kit Positive: 3+ 10.0% (1/10) 50.0% (5/10) PathVysion HER2 DNA Probe Kit Modified ASCO/CAP breast [2007] 50% Not reported
Buza et al., 2013 (31) 85 USC & 23 mixed EC with a serous component DAKO Hercept testing Manufacturer vs. modified 2007 ASCO/CAP; positive: 3+; equivocal: 2+; negative: 0/1 FDA scoring criteria: 14.8%; ASCO/CAP scoring criteria: 21.3% FDA scoring criteria: 27.8%; ASCO/CAP scoring criteria: 23.2% PathVysion HER2 DNA Probe Kit Modified ASCO/CAP breast [2007] 16.7% (18/108) 75% when using the FDA criteria and 81% when using the 2007 ASCO/CAP criteria; after excluding 2+ IHC: 78% using the FDA criteria and 86% using the 2007 ASCO/CAP criteria
Buza & Hui, 2013 (42) 17 USC with heterogenous HER2 expression DAKO Herceptest Modified 2007 ASCO/CAP; positive: 3+/2+; negative: 0/1+ 8/17 9/17 PathVysion HER2 DNA Probe Kit Modified ASCO/CAP breast [2007] 47.1% (8/17) including 2+ initial reflex FISH testing; 82% on repeat testing showing diffuse and cluster amplification Not reported
Fader et al., 2018 (43) 58 advanced or recurrent USC Modified 2007 ASCO/CAP; positive: 3+/2+ (confirmed with FISH) Not reported Not reported Modified ASCO/CAP breast [2007] Not reported Not reported
Erickson et al., 2020 (26) 169 stage 1 USC DAKO HercepTest or Ventana PATHWAY anti-HER2 (4B5) Modified 2007 ASCO/CAP; negative: 0/1+; equivocal: 2+ (confirmed with FISH); positive: 3+ 24.9% (42/169) 16.6% (28/169) PathVysion HER2 DNA Probe Kit and Dako IQFISH kit ASCO/CAP breast [2013] 38.1% (16/42) Not reported
Tymon-Rosario et al., 2021 (44) 12 USC cell lines Thermo Fisher Scientific c-erbB-2 antibody Modified 2007 ASCO/CAP; positive: 3+; equivocal 2+; negative: 0/1+ 16.7% (2/12) 25.0% (3/12) PathVysion HER2 DNA Probe Kit Modified ASCO/CAP breast [2007] (39) 25.0% (3/12) Not reported
Buza & Hui, 2022 (33) 66 USC, 13 mixed endometrial (94 tumor specimens) Abcam EP3 Clone Modified 2007 ASCO/CAP; positive: 3+; equivocal 2+ reflex HER2 FISH; negative: 0/1+ 90.4% (85/94) 3.2% (3/94) PathVysion HER2 DNA Probe Kit Four criteria used: (I) modified ASCO/CAP breast [2007]; (II) ASCO/CAP breast [2013]; (III) ASCO/CAP gastric [2016]; (IV) ASCO/CAP breast [2018] 38.3% (36/94) based in modified ASCO/CAP [2007]; 39.4% (37/94) based on the other 3 guidelines Not reported

Scoring criteria for overexpression on IHC: manufacturer’s scoring criteria (FDA approved): (I) Vertana pathway: 0, no staining or staining without membranous pattern; 1+, incomplete membranous staining or complete membranous staining in <10% of tumor cells; 2+, complete moderate intensity membranous staining in >10% of tumor cells; 3+, complete strong intensity membranous staining in >10% of tumor cells; (II) Dako Hercept: 0, undetectable staining or membrane staining in <10% of the tumor cells; 1+, faint and incomplete membrane staining in >10% of the tumor cells; 2+, weak to moderate complete membrane staining in >10% of the tumor cells; 3+, strong complete membrane staining observed in >10% of the tumor cells; ASCO/CAP [2007] scoring (45): 0, no staining is observed in invasive tumor cells; 1+, weak, incomplete membrane staining in any proportion of invasive tumor cells or weak, complete membrane staining in o10% of cells; 2+, complete membrane staining that is non-uniform or weak but with obvious circumferential distribution in at least 10% of cells or intense complete membrane staining in ≤30% of tumor cells; 3+, uniform intense membrane staining in >30% of invasive tumor cells. Scoring criteria for amplification with FISH (33): (I) modified ASCO/CAP breast [2007]: HER2/neu:CEP17 ≥2.2 OR HER2/neu copy number >6.0/nucleus; (II) ASCO/CAP breast [2013]: HER2/neu:CEP17 ≥2.0 OR HER2/neu:CEP17 <2.0 AND HER2/neu copy number ≥6.0/nucleus; (III) ASCO/CAP gastric [2016]: HER2/neu:CEP17 ≥2.0 OR HER2/neu:CEP17 <2.0 AND HER2/neu IHC is 2+ AND CEP17 ≥3.0/nucleus AND HER2/neu copy number >6.0/nucleus; (IV) ASCO/CAP breast [2018]: HER2/neu:CEP17 ratio ≥2.0 AND average HER2/neu copy number ≥4.0/ nucleus (Gr 1); OR Gr 2 with IHC 3+, Gr 3 with IHC 2+ OR 3+, Gr 4 with IHC 3+. HER2/neu, human epidermal growth factor receptor 2; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; USC, uterine serous cancer; ASCO/CAP, American Society of Clinical Oncology/College of American Pathologists; EC, endometrial cancer; CI, confidence interval; FDA, Food and Drug Administration.

HER2/neu overexpression

Correlation between HER2/neu overexpression, IHC scoring systems and clinical performance have not been satisfactorily elucidated in women with serous ECs. Data on HER2/neu expression from breast cancer cells cannot be extrapolated for USC as it has been widely recognised now that the staining pattern and expression are different for different organs (46). The expression and amplification of HER2/neu differs between tumors of different organs so does the pattern of staining observed in breast, gastric and USC (33). HER2/neu positive breast tumors usually demonstrate a relatively uniform and circumferential staining on IHC while lateral or basolateral, i.e., ‘U-shaped’ staining is seen more frequently in gastric and USC (31). In addition, tumors with basolateral/lateral membranous staining as seen in gastric and serous ECs demonstrated a glandular or pseudo-glandular pattern. Recognition of these differences is important when applying existing guidelines for breast and gastric cancer to HER2/neu testing in USC and highlights the need for testing guidelines specific to EC. Previous studies evaluating HER2/neu expression in endometrial tumors have used the IHC scoring definition of 3+ if >10% cells in the tumor have shown strong complete membrane staining to define HER2/neu positive cells (24,30,38,40,47). Recent studies have found that using a scoring criterion of >30% cells showing strong complete membrane staining to define HER2/neu positive tumors to be more concordant with clinical behaviour (43). Another issue to consider is whether to consider 2+ IHC scoring tumors as HER2/neu positive or equivocal. Though the initial GOG trials have considered such tumors to be HER2/neu positive (38,40), the eligibility as HER2/neu positive tumor in the recent trial by Fader et al. necessitates all 2+ IHC scoring tumors to be confirmed with FISH for gene amplification. The criteria employed defined a specific group with HER2/neu positive tumors who derived significant clinical benefit from targeted therapy as demonstrated even in the subsequent OS results (43,48).

HER2/neu amplification

Amplification by FISH is expressed as the ratio of HER2/neu to chromosome enumeration probe 17 (HER2/neu:CEP17) and HER2/neu copy number signal per cell (49). The International Society of Gynecological Pathologists (ISGyP) published practical recommendations allowing tumors with an IHC score of 2+ with FISH HER2/neu:CEP17 ratio ≥2.0 or <2.0 with average HER2/neu copy number ≥6/nucleus to be designated as HER2/neu positive tumors, in addition to those scoring 3+ on IHC. While definite testing guidelines are yet to be established, these criteria could form the basis for future guideline development for EC (46).

Challenges in HER2/neu testing for USC

Concordance between the two testing modalities has also been an important subject area of previous studies. Reported concordance rates between IHC and FISH/CISH in USC differ significantly with estimated rates between 32% and 100% (35,36). The highest concordance was observed in tissues staining 3+ on IHC (35). It is important to note however that testing methods and scoring criteria do vary between studies and could have contributed to the differences reported. Buza et al. showed that the concordance between HER2/neu overexpression and amplification was 75% when Food and Drug Administration (FDA) criteria for breast cancer was used for IHC scoring while the concordance increased to 81% when American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) scoring system for breast cancer was used (31). Another challenge encountered with HER2/neu testing in USC is intra-tumoral heterogeneity which is described as the presence of at least 2 degrees of difference in staining in at least 5% of tumor cells or amplification of HER2/neu within the same tissue sample (50). Intra-tumoral heterogeneity ranging from 31–97% in HER2/neu expression has been reported with IHC (31,32,36). In small samples, this could be misleading as results may not be entirely representative of the whole tumor and potentially risks excluding some patients from targeted therapy. Therefore, testing multiple sections or larger area of tissue is recommended to reduce the occurrence of false results (31,32,42).

Novel methods of testing HER2/neu

Next-generation sequencing (NGS) is a novel technology used in the identification of gene mutations in cancers and provides a molecular basis for targeted therapy. NGS isolates nucleic acid (DNA or RNA) from the tumor sample, generates sequencing libraries prior to amplification of DNA and subsequently analyses the data (51). A comparative study of targeted NGS with IHC and FISH for the ErbB2 gene in USC showed that NGS was comparable to IHC and FISH in detecting ErbB2 gene amplification with 100% concordance (52). Although the results of this study were positive, it had a small sample size and did not demonstrate the superiority of NGS over current testing modalities.

In breast cancer, alternative HER2/neu testing techniques like immunodetection using Phosphor Integrate Dot fluorescent nanoparticles (53), novel gene protein (54) and the HER2DX clinical response prediction assays (55) have been reported. Hou et al. used the novel gene protein assay on double equivocal cases (IHC and FISH) to determine both gene copy number and protein expression simultaneously and also identified HER2/neu heterogeneity in breast cancer cells (54). This is a promising finding but requires further clinical testing in other tumors including EC. Larger studies are required to confirm the results and cost effectiveness of adopting alternative testing techniques including targeted NGS in clinical practice in comparison with IHC and FISH.


Targeting HER2/neu in uterine serous papillary cancers

Various pharmacologic agents have been developed to inhibit the activity of HER2/neu receptor in breast cancer cell lines with improvement in survival outcomes. Given the molecular similarities of USC with breast cancer, these agents are being actively investigated in EC in preclinical and clinical settings with considerable success.

They can be grouped as follows:

Agents with demonstrable activity in clinical studies:

  • Trastuzumab.

Agents with demonstrable activity in preclinical studies:

  • Targeting extracellular domain of HER2/neu receptors: pertuzumab;
  • Antibody drug conjugates (ADCs): trastuzumab emtansine (T-DM1), DHES0815A, SYD985 (Byondis B.V., Nijmegen, the Netherlands);
  • Targeting intracellular domain of HER2/neu receptors: afatinib, neratinib, dacomitinib, lapatinib;
  • Targeting the downstream pathway of HER2/neu receptor activation such as PIK3CA/Akt/mTOR signaling pathways: GDC-0980 (Genentech Inc., South San Francisco, CA, USA), taselisib.

The only drug tested and used in current clinical practice is trastuzumab, while the other HER2/neu targeting agents were tested in preclinical trials on cell lines. Though promising, the clinical efficacy of these drugs is yet to be established in real world settings.

Trastuzumab

The most important and well-studied of these drugs is trastuzumab (Herceptin, Genentech, San Francisco, CA, USA/Hoffman-Roche, Switzerland). Trastuzumab is a humanized murine IgG1 immunoglobulin that targets the extracellular domain IV of HER2/neu and interferes with dimerization leading to suppression of downstream signaling pathways (56). It also acts by reducing HER2/neu activity by inducing receptor degradation through the ubiquitin proteosome pathway (57). Binding of trastuzumab to HER2/neu receptor also activates immune recognition of tumor cells via antibody dependent cellular cytotoxicity (ADCC) (56). As early as 2002, Santin et al. proposed that trastuzumab can be a potential therapeutic option targeting HER2/neu overexpressing USC. They made this proposition on the basis of their finding that cell lines from patients with chemotherapy and radiotherapy resistant USC were highly sensitive to trastuzumab mediated ADCC (58). In 2006, Jewell et al. reported the successful use of trastuzumab in achieving a partial response on two subsequent recurrences in a patient with HER2/neu overexpressing stage IIIA endometrioid EC (59). Villella et al. reported two patients with HER2/neu positive advanced USC (stage IIIC & IVB) who achieved stable disease and complete response with trastuzumab, respectively (37).

The GOG 181B trial was a phase II trial conducted to evaluate the efficacy of single agent trastuzumab in HER2/neu positive advanced/recurrent endometrial carcinoma. The trial initially recruited patients who were considered HER2/neu positive based on IHC results alone (2+/3+ overexpressing tumors) from 2000 to November 2002. But when 23 patients (Sample A) included during this time (Period A) showed no response, the trial was modified to include patients who were FISH positive for HER2/neu amplification between 2004 and 2007 (Period B). Sample B included patients who showed HER2/neu amplification in Period B plus patients who were FISH positive amongst Sample A. Despite these amendments, the study did not show major objective tumor responses amongst its participants. Tumor response and survival was not associated with HER2/neu amplification or overexpression. The authors concluded that trastuzumab has no activity in HER2/neu positive EC, though the study closed before completion due to poor accrual (40).

A phase II randomized controlled trial by Fader et al. evaluated the addition of trastuzumab to conventional chemotherapy in 58 evaluable patients with advanced and recurrent USC. The experimental arm received paclitaxel and carboplatin for 6 cycles with trastuzumab followed by trastuzumab maintenance till disease progression or toxicity. The trastuzumab arm experienced significant increase in median PFS [12.9 vs. 8.0 months, hazard ratio (HR) =0.46, P=0.005] and OS (29.6 vs. 24.4 months, HR =0.58, P=0.046) compared to the control arm. The greatest benefit was observed in the subset of women who received trastuzumab as primary treatment (OS: not reached vs. 24.4 months, HR =0.49, P=0.041; PFS: 17.7 vs. 9.3 months, HR =0.44, P=0.015) (43,48). As a result of these encouraging results, National Comprehensive Cancer Network (NCCN) guidelines have incorporated trastuzumab to conventional chemotherapy as primary treatment for advanced stage USC and as a first-line option for recurrent USC (60). A modelling study based on a theoretical cohort of 4,000 women also showed that addition of trastuzumab to conventional chemotherapy is cost-effective in advanced and recurrent USC until the cost of treatment for 6 months crosses $38,505 (61). Trastuzumab has good tolerability and an acceptable toxicity profile when given in patients with USC. The most common side effects are gastrointestinal of which almost 95% are low grade. No significant toxicity was observed between experimental arm including trastuzumab and control arms, even in cardiac toxicity (62). Table 3 shows summary of clinical trials evaluating USC treated with the HER2/neu targeting agent trastuzumab.

Table 3

Summary of clinical trials evaluating USC treated with HER2/neu targeting agents in literature

Author, year Sample size Age (years) Stage Disease site HER2/neu testing HER2/neu testing on primary tumor/recurrence HER2/neu targeting at diagnosis/recurrence Drug used Dose Cycles/months of HER2/neu targeted therapy Response PFS (months) OS (months) Comments
Villella et al., 2006 (37) 2 IIIC 3+ overexpression & amplification >10 copies Primary tumor Trastuzumab alone 3 months SD → PD 3
IVA 3+ overexpression & amplification >10 copies Primary tumor Trastuzumab alone 6 months CR → PD 6
Santin et al., 2008 (63) 1 63 IIIC RPLN, vaginal cuff 2+ overexpression & amplification (c-erbB2 gene: reference gene, 3.10) Primary tumor Recurrence Trastuzumab alone LD: 4 mg/kg, MD: 4 mg/kg every 2 weeks 7 months PR 7 CA-125 levels dropped from 144 at start of therapy to 41 U/mL and nodal disease disappeared
Vandenput et al., 2009 (64) 1 71 IVB Vagina, lungs 3+ overexpression & amplification Primary and recurrent tumor Recurrence Trastuzumab alone ×4 cycles trastuzumab + paclitaxel ×11 cycles LD: 8 mg/kg, MD: 6 mg/kg 3 weekly; combined with weekly paclitaxel: LD 4 mg/kg, MD 3 mg/kg weekly Alone: 4 cycles; with paclitaxel: 11 cycles PD
Fleming et al., 2010 (40) 33 HER2/neu-positive tumors out of which 11 were USC Advanced: 7, recurrent: 26 Sample A: 2+/3+ IHC overexpression (n=23) Primary and recurrent tumor Diagnosis for advanced tumors/recurrence for recurrent tumors Trastuzumab alone 4 mg/kg in week 1, then 2 mg/kg weekly until disease progression Mean: 2 cycles SD: 12; PD: 18; indeterminate: 3 1.84 7.85 Testing strategy changed to mandate FISH testing in later part of trial and hence analyses as sample A and sample B; trial closed early due to poor accrual; trastuzumab showed no discernible activity in HER2/ne-positive endometrial cancers; serous and clear cell tumors more likely to show HER2/neu amplification
Sample B: amplification (HER2/neu:CEP17 ratio >2.0) +/− 2+/3+ IHC overexpression (n=18 including 8 patients from sample A) Primary and recurrent tumor 1.81 6.80
Fader et al., 2018, 2020 (43,48) Control arm: 28; experimental arm: 30 Median: 69; control arm: 67; experimental arm: 73 Advanced: stage III, 23; stage IV, 18; recurrent, 17 3+ IHC overexpression & 2+ IHC overexpression confirmed by FISH amplification Primary and recurrent tumor Diagnosis for advanced tumors/recurrence for recurrent tumors Control arm: TP ×6 cycles; experimental arm: TP ×6 cycles + trastuzumab till toxicity or progression T: 175 mg/m2; P: AUC, 5; trastuzumab: 1st cycle, 8 mg/kg; subsequent cycles, 6 mg/kg Median: 15 (range, 5–53) Control arm: 8.0; experimental arm: 12.9 (P=0.005; HR =0.46) Control arm: 24.4; experimental arm: 29.6 (P=0.046; HR =0.58) Greatest benefit of trastuzumab seen in stage III/IV disease (OS: 24.4 vs. NR, P=0.041, HR =0.49)
Pelligra et al., 2020 (65) 2 69 IVB Omentum 3+ IHC overexpression & amplified Primary Diagnosis Trastuzumab TP + trastuzumab: 6 cycles; trastuzumab alone: 11 cycles PD HER2/neu status on biopsy at progression was 1+; received 9 cycles of TP, ixabepilone + bevacizumab, abraxane + bevacizumab + oral cyclophosphamide, doxorubicin and bevacizumab, BSC
76 IVB Omentum 2+ IHC score with HER2/neu:CEP17 ratio of 3.69 to 1.8 Primary Recurrence Trastuzumab Trastuzumab: 31 cycles PD HER2/neu status on biopsy at progression was 0 and FISH was negative

USC, uterine serous cancer; HER2/neu, human epidermal growth factor receptor 2; PFS, progression-free survival; OS, overall survival; SD, stable disease; PD, progressive disease; CR, complete response; RPLN, retroperitoneal lymph node; LD, loading dose; MD, maintenance dose; PR, partial response; CA-125, cancer antigen 125; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; T, paclitaxel; P, carboplatin; AUC, area under curve; HR, hazard ratio; NR, not reported; BSC, best supportive care.

In stage I serous ECs, conventional staging surgery and chemotherapy seems inadequate as the observed recurrence rate is 25.4% despite 71% of patients receiving some form of chemoradiation. Almost 26% of this cohort show HER2/neu positivity and are associated with poorer overall [adjusted R (aHR) =2.00, 95% confidence interval (CI): 1.04–3.88, P=0.039] and PFS (aHR =3.50, 95% CI: 1.84–6.67, P<0.001) compared to HER2/neu negative tumors (26). Clinical trials targeting HER2/neu in this early-stage USCs of patients are lacking (66).

Pertuzumab

Pertuzumab is another humanized monoclonal antibody that targets extracellular domain IV of HER2/neu receptor, which is a different epitope than that targeted by trastuzumab. This binding of pertuzumab to HER2/neu receptor inhibits heterodimerization of HER2 and HER3 which is considered the most potent cellular signaling heterodimer of the EGFR family (67). Pertuzumab works predominantly by stimulating immune activation by ADCC. Pertuzumab has been shown to have efficacy similar to trastuzumab in HER2/neu overexpressing USC cells in vitro. Interestingly, it has also been shown that addition of pertuzumab to trastuzumab significantly increased the ADCC dependent cell killing in vitro in USC cells with low or no expression of HER2/neu receptors compared to either drug alone (68).

T-DM1

T-DM1 is an antibody-drug conjugate of trastuzumab linked to the anti-microtubule cytotoxic agent DM1 through a nonreducible thioether linkage. Each molecule of trastuzumab carries 3.5 molecules of DM1. When trastuzumab binds to HER2/neu receptors, the T-DM1 antibody-drug complex enters the cell via a receptor-mediated endocytosis. Upon proteolytic degradation, the DM1 molecules are released intracellularly and exert their antimicrotubular activities. English et al. showed that T-DM1 was more effective in vitro by inhibition of cell proliferation and induction of apoptosis in HER2/neu positive USC cell lines and in vivo by decreasing tumor formation and prolonging survival of severe combined immunodeficient (SCID) mice harbouring USC xenografts (69).

DHES0815A

DHES0815A is also an ADC in which the antibody against HER2/neu receptor named MHES0488A is bound to pyrrolo[2,1-c][1,4] benzodiazepine monoamide (PBD-MA) which is an alkylating agent at a drug antibody ratio (DAR) of 2. DHES0815A significantly decreases growth of HER2/neu overexpressing USC cell lines in an in vitro study (44).

SYD985

SYD985 is a new antibody-drug conjugate in which trastuzumab is linked to duocarmycin, a DNA alkylating agent having an average DAR of 2.8. In USC cell lines, SYD985 was shown to be 10 to 70 times more potent than T-DM1 and was also effective in USC cells with low to moderate HER2/neu expression, unlike T-DM1 which was only effective against USC with strong HER2/neu expression (70).

Afatinib

Afatinib (BIBW-2992) is an oral HER2/neu inhibiting antibody that works by targeting the intracellular tyrosine kinase domains of ErbB1, ErbB2 and ErbB4 and inhibits transphosphorylation of ErbB3. Afatinib was shown to be active in vitro and in vivo in chemotherapy resistant USC harbouring HER2/neu gene amplification (71).

Neratinib

Neratinib (HKI 272) is another tyrosine kinase inhibitor that binds to the adenosine triphosphate (ATP) pocket of ErbB1 and HER2/neu, thereby inhibiting the downstream signaling pathway. Schwab et al. showed that USC lines with HER2/neu amplification were more sensitive to growth inhibition by neratinib compared to non-amplified cell lines. Neratinib also showed in vitro activity by increasing OS and decreasing tumor growth (72). Olaparib and neratinib showed synergistic activity in USC with high HER2/neu expression. Olaparib treatment increased HER2/neu expression in USC cell lines while treatment with neratinib increased poly(ADP-ribose) polymerases (PARP) activity (73).

Dacomitinib

Dacomitinib is an oral small molecule inhibitor that acts by inhibiting the tyrosine kinase activity of HER1, HER2 and HER4. Dacomitinib has shown to significantly inhibit growth of USC lines harbouring HER2/neu amplification in a dose dependent manner (74).

Lapatinib

Lapatinib is a dual ErbB1/HER2/neu tyrosine inhibitor molecule which has shown antitumor activity in combination with trastuzumab in HER2/neu amplified USC which were impervious to trastuzumab alone in vitro and in vivo (75).

GDC-0980

GDC-0980 is a small molecule inhibitor which selectively inhibits class 1 PIK3 and mTORC1/mTORC2 kinase. PIK3/Akt/mTOR pathway is a signaling cascade that is located downstream to HER2/neu receptor activation. GDC-0980 was found to inhibit growth in USC harbouring HER2/neu amplifications. Among HER2/neu amplified USC lines, those lines that also harboured PIK3CA mutations were more sensitive to inhibition by GDC-0980 compared to PIK3CA wild type USC cell lines (76).

Taselisib

Taselisib, otherwise known as GDC-0032, is an oral small molecule inhibitor of PIK3CA. A strong differential growth inhibition was seen with taselisib in HER2/neu FISH positive tumors with PIK3CA mutation compared to those without HER2/neu amplification or PIK3CA mutation. In vivo survival benefits were observed in HER2/neu FISH positive/PIK3CA-mutated xenografts treated with taselisib compared to controls (77).

The pre-clinical evidence for HER2/neu targeting agents in USC cell lines is summarized in Table 4.

Table 4

Summary of pre-clinical trials evaluating USC treated with HER2/neu targeting agents in literature

Author, year Sample In vitro/in vivo Drug In vitro comparison Tumor growth inhibition (μM), IC50 ± SEM In vivo comparison P value Comments
OS Tumor growth inhibition
El-Sahwi et al.,
2010 (68)
6 USC cell lines In vitro Pertuzumab and/or trastuzumab 3 USC cells with HER2/neu amplification and overexpression vs. 3 HER2/neu-negative USC cells Combination of pertuzumab and trastuzumab was more cytostatic than trastuzumab alone in all cell lines (P<0.005); combination of pertuzumab and trastuzumab was significantly more cytostatic against the high HER2/neu expressor cell lines when compared with pertuzumab used alone (P<0.01)
English et al., 2013 (76) 22 USC cell lines In vitro GDC-0980 9 USC cell lines with HER2/neu amplification vs. 13 cell lines without HER2/neu amplification 0.29±0.05 vs. 1.09±0.20 (P=0.02) FISH USC harboring PIK3CA mutations were significantly more sensitive to GDC-0980 exposure when compared with USC cell lines harboring wild-type PIK3CA (P=0.03)
Lopez et al., 2014 (77) 9 USC cell lines Both Taselisib 4 lines with HER2/neu amplification (FISH) (2 with PIK3CA mutations) vs. 5 lines without HER2/neu amplification 0.042±0.006 vs. 0.38±0.06 (P<0.0001) 10 mice with PIK3CA-mutated/HER2/neu-amplified tumor xenograft (taselisib vs. placebo) <0.0001 0.007
Schwab et al., 2014 (72) 24 USC cell lines (8 were HER2/neu-amplified) Both Neratinib 4 lines with HER2/neu amplification vs. 4 lines without HER2/neu amplification 0.011±0.0008 vs. 0.312±0.0456 (P<0.0001) 10 mice with HER2/neu-amplified tumor xenograft (neratinib vs. placebo) 0.0019 0.0027
Schwab et al., 2014 (71) 15 USC cell lines (5 were HER2/neu-amplified) Both Afatinib 4 lines with HER2/neu amplification vs. 4 lines without HER2/neu amplification 0.0056±0.0006 vs. 0.563±0.092 (P<0.0001) 10 mice with HER2/neu-amplified tumor xenograft (afatinib vs. placebo) 0.0017
Zhu et al., 2015 (74) 8 USC cell lines In vitro Dacomitinib (PF-00299804) 4 lines with HER2/neu amplification vs. 4 lines without HER2/neu amplification 0.02803±0.003355 vs. 1.498±0.2209 (P<0.0001)
English et al., 2014 (69) 15 USC cell lines Both T-DM1 vs. trastuzumab 5 lines with HER2/neu amplification or overexpression vs. 10 HER2/neu-negative cell lines 15 mice with HER2/neu-amplified tumor xenograft not harbouring a PIK3CA mutation (rituximab vs. trastuzumab vs. T-DM1) ≤0.0001 0.04 T-DM1 was considerably more effective than Trastuzumab in inhibiting cell proliferation and in causing apoptosis (P=0.004) of USC showing HER2/neu overexpression
Groeneweg et al., 2014 (75) 42 USC cell lines (10 were HER2/neu-amplified) Both Lapatinib + trastuzumab vs. trastuzumab alone 3 cell lines treated with trastuzumab alone or combination of trastuzumab and lapatinib Mice bearing xenograft randomized into groups of 5 or 6 (lapatinib vs. vehicle) 0.02 Dual anti-HER2/neu therapy with lapatinib led to improved inhibition of tumor growth in HER2/neu-amplified USC
Tymon-Rosario et al., 2021 (44) 12 USC cell lines Both DHES0815A vs. MHES0488A (unconjugated antibody) 3 lines with HER2/neu amplification vs. 1 line without HER2/neu amplification P<0.05 12 mice with xenograft model with 3+ HER2/neu expression (DHES0815A vs. MHES0488A) <0.01 <0.01
Yadav et al., 2022 (73) 6 USC cell lines Both Neratinib vs. olaparib vs. both 2 lines with HER2/neu amplification vs. 1 line without HER2/neu amplification 5–6 mice with xenograft model with 3+ HER2/neu expression per group (vehicle, olaparib, neratinib, combination) <0.05 (combination of the two inhibitors caused a stronger and durable growth inhibition in both USC xenografts) Neratinib was more potent than olaparib in suppression of in-vitro growth of HER2/neu 3+ cell lines while no difference was noted against HER2/neu 1+ tumors. combination of olaparib with neratinib synergistically improved tumor suppression compared to either single-agent in vitro

USC, uterine serous cancer; HER2/neu, human epidermal growth factor receptor 2; IC50, half maximal inhibitory concentration; SEM, standard error of the mean; OS, overall survival; FISH, fluorescence in situ hybridization.


Mechanisms of resistance to HER2/neu targeted therapy in USC and ways to overcome it

Solid tumors that express HER2/neu may not respond to HER2/neu targeting therapy either from the initiation of therapy (primary resistance) or after some time of response (secondary resistance). Several factors have been proposed to account for such resistance.

Presence of p95HER2 variant

P95HER2 is a variant of HER2/neu receptor which loses the extracellular binding site for trastuzumab while retaining the intracellular domain responsible for activation of downstream signaling pathway. As a result, these HER2/neu overexpressing tumors are resistant to the effects of trastuzumab. Growdon et al. showed that almost 53% of high-grade ECs (predominantly consisting of USC) showed p95HER2 expression. This was considerably higher than p95HER2 expression in a matched breast cancer cohort (78).

Upregulation of downstream pathways

Tumors that show increased activity/signaling of HER2/neu downstream pathways such as PIK3CA and mTOR pathways are resistant to HER2/neu targeting agents. Black et al. showed that HER2/neu amplified/PIK3CA-mutated tumors were more resistant to trastuzumab compared to HER2/neu amplified/PIK3CA wild type cell lines. Resistance of tumors to trastuzumab that were transfected with oncogenic PIK3CA mutations increased compared to baseline (79).

Signaling from other HER receptors

Tumors may escape HER2/neu receptor targeting by increasing activation of other HER receptors like HER3. HER2-HER3 receptor signaling is considered the most potent signal among signaling by EGFR family of receptors. HER3 overexpression may lead to heterodimerization with HER2/neu receptors which may in turn lead to activation of downstream signaling pathways leading to tumor proliferation and growth.

Tumor selection of resistant clones

Treatment of HER2/neu positive serous endometrial tumors may induce the proliferation of HER2/neu negative clones within the tumor as a way of resistance. In a case report on two patients with initial tumor cells shown to be overexpressing HER2/neu by IHC and ErbB2 amplification by FISH, biopsy at the time of disease progression on trastuzumab therapy demonstrated loss of HER2/neu overexpression in the resistant tumors (65).

Overcoming resistance in HER2/neu targeted therapy

Resistance to HER2/neu targeted therapy can be mitigated in various ways. Combination of HER2/neu targeted therapy to a drug that acts on the downstream pathways is the most common strategy employed in such cases. Lopez et al. showed that combination therapy with neratinib and taselisib is found to produce stronger and long-lasting growth inhibition of tumor cell lines compared to either treatment alone. They also showed that combination therapy is effective even when initiated after tumor progression compared to single targeted therapy with either agent alone in mice xenografts that are HER2/neu and PIK3CA overexpressed/amplified (80). Combination of a intracellular HER2/neu targeting agent with a drug that targets extracellular domain is also a way of overcoming resistance (75).


Ongoing trials

Several ongoing trials are evaluating the efficacy of HER2/neu targeting agents in serous ECs. Most are basket trials which require tumors of different organs to harbour HER2/neu overexpression and/or amplification to be included in the trial.

DESTINY-Breast01 was a two-part open labelled phase 2 trial to evaluate the efficacy of trastuzumab deruxtecan on metastatic breast cancer patients who were previously treated with T-DM1. Trastuzumab deruxtecan showed a overall response rate (ORR) of 60.9% and a median PFS of 16.4 months (81). DESTINY-PanTumor02 trial is an open label phase II multicentre basket trial that evaluated the efficacy of trastuzumab deruxtecan in 40 patients with advanced or metastatic EC in addition to other tumors including biliary tract, bladder, cervical, ovarian, pancreatic, or other tumors (excluding breast, gastric, colorectal, and non-small cell lung cancer). Only patients with tumors expressing 2+/3+ were included in the study. The highest ORR among all tumor groups was observed in ECs at 57.5%. In uterine tumors that showed strong positivity (3+) for HER2/neu expression, the ORR was 84.6% (82,83).

MyPathway is a non-randomised phase 2a basket trial evaluating targeted therapy in solid tumors with specific molecular alterations which are not currently approved in clinical practice. The combination of pertuzumab and trastuzumab was used in HER2/neu overexpressing or amplified tumors including EC. Early reports published show no objective response in seven cases of HER2/neu positive EC recruited but conclusive results are still awaited (84). NCT04585958 is a phase II trial looking at the maximum tolerated dose of trastuzumab deruxtecan when combined with olaparib in HER2/neu expressing tumors including ECs (85). A multicentre phase II/III trial involving 172 centers in the USA is currently evaluating the effect of adding Trastuzumab or a combination of trastuzumab and pertuzumab in uterine carcinosarcomas and serous carcinomas (86).


Future directions

HER2/neu testing and interpretation varies between studies as demonstrated in this review and was mostly extrapolated from the ASCO/CAP guidelines for HER2/neu testing in breast cancer. Despite these extrapolated recommendations, testing criteria/guidelines specific to EC should be established as was done for breast (ASCO/CAP), gastric (ASCO/CAP) and most recently colorectal cancer (HERACLES trial) (50). The criteria employed by Fader et al. provides a basis upon which further trials can be designed towards standardising testing practices and interpretation (43).

It is observed that in addition to USC, other non-serous endometrial tumors also demonstrate HER2/neu overexpression and amplification. Whether these histological subtypes might derive benefit from HER2/neu targeted therapy is a question that needs to be answered. In this respect, molecular analysis of patients enrolled in PORTEC-3 trial showed that there was a stronger correlation between HER2/neu amplification and TP53 mutation than there is with serous cancer (16). The STATICE trial which evaluated the effect of trastuzumab on carcinosarcoma recently concluded that targeted therapy with trastuzumab deruxtecan, an ADC in which trastuzumab is linked to a topoisomerase I inhibitor, confers some benefit even in non-serous tumors (87). NCCN guidelines recommend HER2/neu IHC testing in patients with carcinosarcomas and TP53 mutations, in addition to advanced and recurrent serous ECs (60).

HER2/neu targeting agents appear to be clinically more meaningful in the primary treatment of advanced serous uterine cancers compared to recurrent settings. Novel antibody-drug conjugates and suitable combinations with immune check-point inhibitors and PI3K/Akt/mTOR inhibitors should be evaluated to improve outcomes in advanced and recurrent cancer. Another avenue that warrants investigation is the use of these agents in neoadjuvant setting for cases that are unsuitable for surgical cytoreduction. Early-stage serous ECs overexpress HER2/neu in approximately one quarter of the cases and HER2/neu positive serous cancers are associated with a significant increase in recurrence and death compared to HER2/neu negative serous tumors. Beyond prognostication, the role of HER2/neu as an actionable target in these tumor remains to be elucidated in early-stage serous ECs.


Conclusions

USC is a biologically aggressive disease associated with a poor prognosis. Alteration of HER2/neu pathway is a commonly found molecular aberration that contributes to aggressive behaviour of these tumors. Considering its negative prognostic implications and clinical benefit conferred by HER2/neu targeted therapy, HER2/neu testing is recommended for all cases of serous EC in advanced and recurrent settings. Trastuzumab in combination with platinum and taxanes based chemotherapy is the recommended treatment option for patients with advanced or recurrent serous cancers who test positive to HER2/neu. Role of HER2/neu as a therapeutic target in early-stage serous cancers remains to be seen but appears promising given that greatest benefits have been in patients treated with HER2/neu targeting agents in the first-line therapy. Focus of future research should be directed towards refining biomarker testing and selection of patients who will derive the most benefit from such therapy.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-23-1465/rc

Peer Review File: Available at https://atm.amegroups.com/article/view/10.21037/atm-23-1465/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-23-1465/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Endometrial cancer statistics | World Cancer Research Fund International [Internet]. WCRF International. [cited 2023 Jul 13]. Available online: https://www.wcrf.org/cancer-trends/endometrial-cancer-statistics/
  2. Ferriss JS, Erickson BK, Shih IM, et al. Uterine serous carcinoma: key advances and novel treatment approaches. Int J Gynecol Cancer 2021;31:1165-74. [Crossref] [PubMed]
  3. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 1983;15:10-7. [Crossref] [PubMed]
  4. Pathology Outlines - Serous carcinoma [Internet]. [cited 2023 Jul 13]. Available online: https://www.pathologyoutlines.com/topic/uterusserous.html
  5. Creasman WT, Odicino F, Maisonneuve P, et al. Carcinoma of the corpus uteri. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int J Gynaecol Obstet 2006;95:S105-43. [PubMed]
  6. Cirisano FD Jr, Robboy SJ, Dodge RK, et al. Epidemiologic and surgicopathologic findings of papillary serous and clear cell endometrial cancers when compared to endometrioid carcinoma. Gynecol Oncol 1999;74:385-94. [Crossref] [PubMed]
  7. Hamilton CA, Cheung MK, Osann K, et al. Uterine papillary serous and clear cell carcinomas predict for poorer survival compared to grade 3 endometrioid corpus cancers. Br J Cancer 2006;94:642-6. [Crossref] [PubMed]
  8. Moore KN, Fader AN. Uterine papillary serous carcinoma. Clin Obstet Gynecol 2011;54:278-91. [Crossref] [PubMed]
  9. Fader AN, Boruta D, Olawaiye AB, et al. Uterine papillary serous carcinoma: epidemiology, pathogenesis and management. Curr Opin Obstet Gynecol 2010;22:21-9. [Crossref] [PubMed]
  10. Mukerji B, Baptiste C, Chen L, et al. Racial disparities in young women with endometrial cancer. Gynecol Oncol 2018;148:527-34. [Crossref] [PubMed]
  11. Bregar AJ, Alejandro Rauh-Hain J, Spencer R, et al. Disparities in receipt of care for high-grade endometrial cancer: A National Cancer Data Base analysis. Gynecol Oncol 2017;145:114-21. [Crossref] [PubMed]
  12. Bogani G, Ray-Coquard I, Concin N, et al. Uterine serous carcinoma. Gynecol Oncol 2021;162:226-34. [Crossref] [PubMed]
  13. Bartosch C, Manuel Lopes J, Oliva E. Endometrial carcinomas: a review emphasizing overlapping and distinctive morphological and immunohistochemical features. Adv Anat Pathol 2011;18:415-37. [Crossref] [PubMed]
  14. Di Donato V, Giannini A, Bogani G. Recent Advances in Endometrial Cancer Management. J Clin Med 2023;12:2241. [Crossref] [PubMed]
  15. Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497:67-73. [Crossref] [PubMed]
  16. Vermij L, Horeweg N, Leon-Castillo A, et al. HER2 Status in High-Risk Endometrial Cancers (PORTEC-3): Relationship with Histotype, Molecular Classification, and Clinical Outcomes. Cancers (Basel) 2020;13:44. [Crossref] [PubMed]
  17. Ross DS, Devereaux KA, Jin C, et al. Histopathologic features and molecular genetic landscape of HER2-amplified endometrial carcinomas. Mod Pathol 2022;35:962-71. [Crossref] [PubMed]
  18. Zhang L, Kwan SY, Wong KK, et al. Pathogenesis and Clinical Management of Uterine Serous Carcinoma. Cancers (Basel) 2020;12:686. [Crossref] [PubMed]
  19. Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007;26:6469-87. [Crossref] [PubMed]
  20. Sahin A, Zhang H. Invasive Breast Carcinoma. In: Pathobiology of Human Disease. In: McManus LM, Mitchell RN. editors. Pathobiology of Human Disease: A Dynamic Encyclopedia of Disease Mechanisms. Elsevier; 2014:934-51.
  21. Alrhmoun S, Sennikov S. The Role of Tumor-Associated Antigen HER2/neu in Tumor Development and the Different Approaches for Using It in Treatment: Many Choices and Future Directions. Cancers (Basel) 2022;14:6173. [Crossref] [PubMed]
  22. Ménard S, Casalini P, Campiglio M, et al. Role of HER2/neu in tumor progression and therapy. Cell Mol Life Sci 2004;61:2965-78. [PubMed]
  23. Iqbal N, Iqbal N. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol Biol Int 2014;2014:852748. [Crossref] [PubMed]
  24. Togami S, Sasajima Y, Oi T, et al. Clinicopathological and prognostic impact of human epidermal growth factor receptor type 2 (HER2) and hormone receptor expression in uterine papillary serous carcinoma. Cancer Sci 2012;103:926-32. [Crossref] [PubMed]
  25. Díaz-Montes TP, Ji H, Smith Sehdev AE, et al. Clinical significance of Her-2/neu overexpression in uterine serous carcinoma. Gynecol Oncol 2006;100:139-44. [Crossref] [PubMed]
  26. Erickson BK, Najjar O, Damast S, et al. Human epidermal growth factor 2 (HER2) in early stage uterine serous carcinoma: A multi-institutional cohort study. Gynecol Oncol 2020;159:17-22. [Crossref] [PubMed]
  27. Santin AD, Bellone S, Van Stedum S, et al. Amplification of c-erbB2 oncogene: a major prognostic indicator in uterine serous papillary carcinoma. Cancer 2005;104:1391-7. [Crossref] [PubMed]
  28. Odicino FE, Bignotti E, Rossi E, et al. HER-2/neu overexpression and amplification in uterine serous papillary carcinoma: comparative analysis of immunohistochemistry, real-time reverse transcription-polymerase chain reaction, and fluorescence in situ hybridization. Int J Gynecol Cancer 2008;18:14-21. [PubMed]
  29. Morrison C, Zanagnolo V, Ramirez N, et al. HER-2 is an independent prognostic factor in endometrial cancer: association with outcome in a large cohort of surgically staged patients. J Clin Oncol 2006;24:2376-85. [Crossref] [PubMed]
  30. Santin AD, Bellone S, Siegel ER, et al. Racial differences in the overexpression of epidermal growth factor type II receptor (HER2/neu): a major prognostic indicator in uterine serous papillary cancer. Am J Obstet Gynecol 2005;192:813-8. [Crossref] [PubMed]
  31. Buza N, English DP, Santin AD, et al. Toward standard HER2 testing of endometrial serous carcinoma: 4-year experience at a large academic center and recommendations for clinical practice. Mod Pathol 2013;26:1605-12. [Crossref] [PubMed]
  32. Banet N, Shahi M, Batista D, et al. HER-2 Amplification in Uterine Serous Carcinoma and Serous Endometrial Intraepithelial Carcinoma. Am J Surg Pathol 2021;45:708-15. [Crossref] [PubMed]
  33. Buza N, Hui P. Characteristics of HER2 Gene Amplification by Fluorescence In Situ Hybridization in Endometrial Serous Carcinoma. Arch Pathol Lab Med 2022;146:0.
  34. Singh P, Smith CL, Cheetham G, et al. Serous carcinoma of the uterus-determination of HER-2/neu status using immunohistochemistry, chromogenic in situ hybridization, and quantitative polymerase chain reaction techniques: its significance and clinical correlation. Int J Gynecol Cancer 2008;18:1344-51. [Crossref] [PubMed]
  35. Klc TR, Wu S, Wilhite AM, et al. HER2 in Uterine Serous Carcinoma: Testing platforms and implications for targeted therapy. Gynecol Oncol 2022;167:289-94. [Crossref] [PubMed]
  36. Mentrikoski MJ, Stoler MH. HER2 immunohistochemistry significantly overestimates HER2 amplification in uterine papillary serous carcinomas. Am J Surg Pathol 2014;38:844-51. [Crossref] [PubMed]
  37. Villella JA, Cohen S, Smith DH, et al. HER-2/neu overexpression in uterine papillary serous cancers and its possible therapeutic implications. Int J Gynecol Cancer 2006;16:1897-902. [PubMed]
  38. Grushko TA, Filiaci VL, Mundt AJ, et al. An exploratory analysis of HER-2 amplification and overexpression in advanced endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2008;108:3-9. [Crossref] [PubMed]
  39. Xu M, Schwartz P, Rutherford T, et al. HER-2/neu receptor gene status in endometrial carcinomas: a tissue microarray study. Histopathology 2010;56:269-73. [Crossref] [PubMed]
  40. Fleming GF, Sill MW, Darcy KM, et al. Phase II trial of trastuzumab in women with advanced or recurrent, HER2-positive endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2010;116:15-20. [Crossref] [PubMed]
  41. Todeschini P, Cocco E, Bellone S, et al. Her2/neu extracellular domain shedding in uterine serous carcinoma: implications for immunotherapy with trastuzumab. Br J Cancer 2011;105:1176-82. [Crossref] [PubMed]
  42. Buza N, Hui P. Marked heterogeneity of HER2/NEU gene amplification in endometrial serous carcinoma. Genes Chromosomes Cancer 2013;52:1178-86. [Crossref] [PubMed]
  43. Fader AN, Roque DM, Siegel E, et al. Randomized Phase II Trial of Carboplatin-Paclitaxel Versus Carboplatin-Paclitaxel-Trastuzumab in Uterine Serous Carcinomas That Overexpress Human Epidermal Growth Factor Receptor 2/neu. J Clin Oncol 2018;36:2044-51. [Crossref] [PubMed]
  44. Tymon-Rosario J, Bonazzoli E, Bellone S, et al. DHES0815A, a novel antibody-drug conjugate targeting HER2/neu, is highly active against uterine serous carcinomas in vitro and in vivo. Gynecol Oncol 2021;163:334-41. [Crossref] [PubMed]
  45. Wolff AC, Hammond ME, Schwartz JN, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 2007;131:18-43. [Crossref] [PubMed]
  46. Buza N. HER2 Testing and Reporting in Endometrial Serous Carcinoma: Practical Recommendations for HER2 Immunohistochemistry and Fluorescent In Situ Hybridization: Proceedings of the ISGyP Companion Society Session at the 2020 USCAP Annual Meeting. Int J Gynecol Pathol 2021;40:17-23. [Crossref] [PubMed]
  47. Slomovitz BM, Broaddus RR, Burke TW, et al. Her-2/ neu Overexpression and Amplification in Uterine Papillary Serous Carcinoma. J Clin Oncol 2004;22:3126-32. [Crossref] [PubMed]
  48. Fader AN, Roque DM, Siegel E, et al. Randomized Phase II Trial of Carboplatin-Paclitaxel Compared with Carboplatin-Paclitaxel-Trastuzumab in Advanced (Stage III-IV) or Recurrent Uterine Serous Carcinomas that Overexpress Her2/Neu (NCT01367002): Updated Overall Survival Analysis. Clin Cancer Res 2020;26:3928-35. [Crossref] [PubMed]
  49. Wolff AC, Hammond ME, Hicks DG, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 2013;31:3997-4013. [Crossref] [PubMed]
  50. Buza N. HER2 Testing in Endometrial Serous Carcinoma: Time for Standardized Pathology Practice to Meet the Clinical Demand. Arch Pathol Lab Med 2021;145:687-91. [Crossref] [PubMed]
  51. Garcia EP, Minkovsky A, Jia Y, et al. Validation of OncoPanel: A Targeted Next-Generation Sequencing Assay for the Detection of Somatic Variants in Cancer. Arch Pathol Lab Med 2017;141:751-8. [Crossref] [PubMed]
  52. Robinson CL, Harrison BT, Ligon AH, et al. Detection of ERBB2 amplification in uterine serous carcinoma by next-generation sequencing: an approach highly concordant with standard assays. Mod Pathol 2021;34:603-12. [Crossref] [PubMed]
  53. Hicks DG, Buscaglia B, Goda H, et al. A novel detection methodology for HER2 protein quantitation in formalin-fixed, paraffin embedded clinical samples using fluorescent nanoparticles: an analytical and clinical validation study. BMC Cancer 2018;18:1266. [Crossref] [PubMed]
  54. Hou Y, Nitta H, Li Z. HER2 Gene Protein Assay Is Useful to Determine HER2 Status and Evaluate HER2 Heterogeneity in HER2 Equivocal Breast Cancer. Am J Clin Pathol 2017;147:89-95. [Crossref] [PubMed]
  55. Prat A, Guarneri V, Pascual T, et al. Development and validation of the new HER2DX assay for predicting pathological response and survival outcome in early-stage HER2-positive breast cancer. EBioMedicine 2022;75:103801. [Crossref] [PubMed]
  56. Vu T, Claret FX. Trastuzumab: Updated Mechanisms of Action and Resistance in Breast Cancer. Front Oncol 2012;2:62.
  57. Wachter RF, Briggs GP, Pedersen CE Jr. Precipitation of phase I antigen of Coxiella burnetii by sodium sulfite. Acta Virol 1975;19:500. [PubMed]
  58. Santin AD, Bellone S, Gokden M, et al. Overexpression of HER-2/neu in uterine serous papillary cancer. Clin Cancer Res 2002;8:1271-9. [PubMed]
  59. Jewell E, Secord AA, Brotherton T, et al. Use of trastuzumab in the treatment of metastatic endometrial cancer. Int J Gynecol Cancer 2006;16:1370-3. [Crossref] [PubMed]
  60. Abu-Rustum N, Yashar C, Arend R, et al. Uterine Neoplasms, Version 1.2023, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2023;21:181-209. [Crossref] [PubMed]
  61. Batman S, Bohn J, Weisenberger MW, et al. Trastuzumab with carboplatin/paclitaxel for treatment of advanced stage and recurrent uterine papillary serous carcinoma: A cost-effectiveness analysis. Gynecol Oncol 2021;160:214-8. [Crossref] [PubMed]
  62. Tymon-Rosario J, Siegel ER, Bellone S, et al. Trastuzumab tolerability in the treatment of advanced (stage III-IV) or recurrent uterine serous carcinomas that overexpress HER2/neu. Gynecol Oncol 2021;163:93-9. [Crossref] [PubMed]
  63. Santin AD, Bellone S, Roman JJ, et al. Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu. Int J Gynaecol Obstet 2008;102:128-31. [Crossref] [PubMed]
  64. Vandenput I, Vanden Bempt I, Leunen K, et al. Limited clinical benefit from trastuzumab in recurrent endometrial cancer: two case reports. Gynecol Obstet Invest 2009;67:46-8. [Crossref] [PubMed]
  65. Pelligra S, Buza N, Hui P, et al. Selection of HER2/NEU negative tumor cells as a mechanism of resistance to trastuzumab in uterine serous carcinoma. Gynecol Oncol Rep 2020;32:100554. [Crossref] [PubMed]
  66. Rossi EC. Identifying the worst of the worst: HER2 positive early stage uterine papillary serous carcinoma. Gynecol Oncol 2020;159:1-2. [Crossref] [PubMed]
  67. Capelan M, Pugliano L, De Azambuja E, et al. Pertuzumab: new hope for patients with HER2-positive breast cancer. Ann Oncol 2013;24:273-82. [Crossref] [PubMed]
  68. El-Sahwi K, Bellone S, Cocco E, et al. In vitro activity of pertuzumab in combination with trastuzumab in uterine serous papillary adenocarcinoma. Br J Cancer 2010;102:134-43. [Crossref] [PubMed]
  69. English DP, Bellone S, Schwab CL, et al. T-DM1, a novel antibody-drug conjugate, is highly effective against primary HER2 overexpressing uterine serous carcinoma in vitro and in vivo. Cancer Med 2014;3:1256-65. [Crossref] [PubMed]
  70. Black J, Menderes G, Bellone S, et al. SYD985, a Novel Duocarmycin-Based HER2-Targeting Antibody-Drug Conjugate, Shows Antitumor Activity in Uterine Serous Carcinoma with HER2/Neu Expression. Mol Cancer Ther 2016;15:1900-9. [Crossref] [PubMed]
  71. Schwab CL, Bellone S, English DP, et al. Afatinib demonstrates remarkable activity against HER2-amplified uterine serous endometrial cancer in vitro and in vivo. Br J Cancer 2014;111:1750-6. [Crossref] [PubMed]
  72. Schwab CL, English DP, Roque DM, et al. Neratinib shows efficacy in the treatment of HER2/neu amplified uterine serous carcinoma in vitro and in vivo. Gynecol Oncol 2014;135:142-8. [Crossref] [PubMed]
  73. Yadav G, Roque DM, Bellone S, et al. Synergistic activity of neratinib in combination with olaparib in uterine serous carcinoma overexpressing HER2/neu. Gynecol Oncol 2022;166:351-7. [Crossref] [PubMed]
  74. Zhu L, Lopez S, Bellone S, et al. Dacomitinib (PF-00299804), a second-generation irreversible pan-erbB receptor tyrosine kinase inhibitor, demonstrates remarkable activity against HER2-amplified uterine serous endometrial cancer in vitro. Tumour Biol 2015;36:5505-13. [Crossref] [PubMed]
  75. Groeneweg JW, Hernandez SF, Byron VF, et al. Dual HER2 targeting impedes growth of HER2 gene-amplified uterine serous carcinoma xenografts. Clin Cancer Res 2014;20:6517-28. [Crossref] [PubMed]
  76. English DP, Bellone S, Cocco E, et al. Oncogenic PIK3CA gene mutations and HER2/neu gene amplifications determine the sensitivity of uterine serous carcinoma cell lines to GDC-0980, a selective inhibitor of Class I PI3 kinase and mTOR kinase (TORC1/2). Am J Obstet Gynecol 2013;209:465.e1-9. [Crossref] [PubMed]
  77. Lopez S, Schwab CL, Cocco E, et al. Taselisib, a selective inhibitor of PIK3CA, is highly effective on PIK3CA-mutated and HER2/neu amplified uterine serous carcinoma in vitro and in vivo. Gynecol Oncol 2014;135:312-7. [Crossref] [PubMed]
  78. Growdon WB, Groeneweg J, Byron V, et al. HER2 over-expressing high grade endometrial cancer expresses high levels of p95HER2 variant. Gynecol Oncol 2015;137:160-6. [Crossref] [PubMed]
  79. Black JD, Lopez S, Cocco E, et al. PIK3CA oncogenic mutations represent a major mechanism of resistance to trastuzumab in HER2/neu overexpressing uterine serous carcinomas Br J Cancer 2015;113:1020-6. [published correction appears in Br J Cancer 2015 Dec 1;113(11):1641]. [Crossref] [PubMed]
  80. Lopez S, Cocco E, Black J, et al. Dual HER2/PIK3CA Targeting Overcomes Single-Agent Acquired Resistance in HER2-Amplified Uterine Serous Carcinoma Cell Lines In vitro and In vivo. Mol Cancer Ther 2015;14:2519-26. [Crossref] [PubMed]
  81. Modi S, Saura C, Yamashita T, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N Engl J Med 2020;382:610-21. [Crossref] [PubMed]
  82. Meric-Bernstam F, Anoka C, Dobrowolska A, et al. A phase 2, multicenter, open-label study evaluating trastuzumab deruxtecan (T-DXd) for the treatment of select human epi-dermal growth factor receptor 2 (HER2)-expressing solid tumors (DESTI-NY-PanTumor02). J Clin Oncol 2022;40:abstr TPS623.
  83. Meric-Bernstam F, Makker V, Oaknin A, et al. Efficacy and safety of trastuzumab deruxtecan (T-DXd) in patients (pts) with HER2-expressing solid tumors: DESTI-NY-PanTumor02 (DP-02) interim results. J Clin Oncol 2023;41:abstr LBA3000.
  84. Hainsworth JD, Meric-Bernstam F, Swanton C, et al. Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results From MyPathway, an Open-Label, Phase IIa Multiple Basket Study. J Clin Oncol 2018;36:536-42. [Crossref] [PubMed]
  85. National Cancer Institute (NCI). A Phase I Study of DS-8201a in Combination With Olaparib in HER2-Expressing Malignancies [Internet]. clinicaltrials.gov; 2023 Jul [cited 2023 Jul 13]. Report No.: NCT04585958. Available online: https://clinicaltrials.gov/study/NCT04585958
  86. National Cancer Institute (NCI). A Phase II/III Study of Paclitaxel/Carboplatin Alone or Combined With Either Trastuzumab and Hyaluronidase-oysk (HERCEPTIN HYLECTA) or Pertuzumab, Trastuzumab, and Hyaluronidase-zzxf (PHESGO) in HER2 Positive, Stage I-IV Endometrial Serous Carcinoma or Carcinosarcoma [Internet]. clini-caltrials.gov; 2023 Jul [cited 2023 Jul 12]. Report No.: NCT05256225. Available online: https://clinicaltrials.gov/study/NCT05256225
  87. Nishikawa T, Hasegawa K, Matsumoto K, et al. Trastuzumab Deruxtecan for Human Epidermal Growth Factor Receptor 2–Expressing Advanced or Recurrent Uterine Carcinosarcoma (NCCH1615): The STATICE Trial. J Clin Oncol 2023;41:2789-99. [Crossref] [PubMed]
Cite this article as: Jayraj AS, Abdul-Aziz S, Mburu A, Upadhyay A, Singh N, Ghatage P. Narrative review on the evolving role of HER2/neu targeting in uterine serous cancers. Ann Transl Med 2024;12(4):69. doi: 10.21037/atm-23-1465

Download Citation