639
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Can Patients with HER2-Low Breast Cancer Benefit from Anti-HER2 Therapies? A Review

, , ORCID Icon, &
Pages 281-294 | Received 04 Feb 2023, Accepted 13 Apr 2023, Published online: 21 Apr 2023

Abstract

Breast cancer (BC) poses a severe threat to the health of women worldwide. Currently, different therapeutic regimens are used for BC according to the pathological classification of HER2-positive or HER2-negative. Clinical reports of HER2-low expression indicate that the condition is HER2-negative, which was ineligible for HER2-targeted therapy. In contrast to HER2-zero tumors, however, HER2-low BC is a heterogeneous disease with unique genetic characteristics, prognoses, and different therapeutic responses. Clinical efficacy has been demonstrated by numerous potent and innovative anti-HER2 medications, particularly antibody–drug conjugates (ADCs). Certain ADCs, including T-DXd, have demonstrated good efficacy in some trials either used alone or in conjunction with other medications. To enhance outcomes in individuals with HER2-low BC, immunotherapy and other treatments are frequently combined with HER2-targeted therapy. There are also alternative strategies that target both HER2 and HER3 or other antigenic sites. We hope more individuals with HER2-low BC will benefit from more precise treatment regimens in the future. This article provides a review of existing research and clinical trials.

Introduction

Breast cancer (BC) has become the most commonly diagnosed tumor in women worldwide.Citation1 Human epidermal growth factor receptor 2 (HER2)-positive BC accounted for about 15% of all cases and was considered a poor prognostic predictor in the past. However, the survival outcomes of these patients have improved considerably and were similar to HER2-negative with the evolution of anti-HER2 agents.Citation2–6 In the past, HER2-low BC could not gain clinical benefit from conventional anti-HER2 agents like trastuzumab.Citation7 However, with the introduction of novel anti-HER2 compounds, BC with low levels of HER2 expression and no detectable Erb-B2 receptor tyrosine kinase 2 (ERBB2) gene amplification might also derive benefit. The efficacy was found when anti-HER2 agents were combined with other therapies such as immunotherapy.

According to the updated 2018 American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) guidelines, the HER2 status is assessed to evaluate HER2 protein expression levels by IHC (immunohistochemistry) and HER2 gene status by ISH (in situ hybridization).Citation8 HER2-low is defined as IHC1+ or IHC2+ without gene amplification, accounting for about 55%.Citation9,Citation10 The application of the new guidelines leads to an increase in the negativity rate on HER2 testing for the reclassification of Group 2 and Group 4 cases.Citation11 The refinement of future diagnostics may be needed to assess HER2 status more accurately.

Based on the expression of hormone receptors (HR) and the status of HER2, BC is classified into four intrinsic molecular subtypes: luminal A (HR-positive and HER2-negative), luminal B (HR-positive and HER2-positive), HER2 enriched (HR-negative and HER2-positive), and triple-negative BC (TNBC, HR-negative and HER2-negative).Citation12 The four subtypes of tumors have different prognoses, and TNBC is generally considered to be the worst.Citation13 The possibility of improving the treatment of HER2-low BC has great clinical significance. Also, different treatment approaches for the four subtypes, respectively. In clinical practice, HER2-low BC was reported as HER2-negative, which was classified as TNBC or luminal-like. However, HER2-low BC is a heterogeneous disease, which showed distinctive molecular features.Citation9,Citation14 Compared with HER2-zero tumors, HER2-low BC has a higher rate of HR-positive (64–88%),Citation9,Citation15 a higher disease-free survival (DFS) and overall survival (OS),Citation16 and a limited immune response.Citation17 This implies that they may have differences in response to therapy, and different therapeutic strategies are needed. Even the HR-positive BC, there are about 50% of these tumors present either inherent or acquired resistance to hormonal therapy.Citation18 Thus, the HER2-low, luminal B BC with endocrine-resistant may also benefit from anti-HER2 therapies.

The NSABP B-47 trial showed that the addition of trastuzumab to adjuvant chemotherapy did not improve the outcomes of patients with HER2-low BC.Citation7 Pertuzumab failed to improve the outcomes either.Citation19 It should be noted that this study used pertuzumab monotherapy, which was ineffective in HER2-positive tumors. HER2-low BC was thought not eligible for anti-HER2 monoclonal antibodies.Citation20 A range of 100,0000–500,000 HER2 receptor molecules is present on the membrane of score 1+ and 2+ BC cells.Citation21 So, it still makes sense that anti-HER2 agents could have clinical applications on HER2-low BC mechanistically. Novel and more potent anti-HER2 agents may give the tumor tissue a sufficient chemotherapeutic agent. Antibody–drug conjugates (ADCs) are effective and show clinical efficacy when combined with other treatments such as immunotherapy. We need to reconceptualize the significance of HER2-low and explore anti-HER2 treatments that apply to these patients.

HER2-Targeted Therapies

Monoclonal Antibodies

Although the efficacy of trastuzumab and pertuzumab in HER2-low BC was not found,Citation7,Citation19 there are still novel monoclonal antibodies being tested for treating HER2-low BC. Margetuximab is a second-generation monoclonal antibody which binds to the same epitope on HER2 as trastuzumab but has higher activity against HER2-positive cancer cells than trastuzumab. Based on the Phase III SOPHIA trial (NCT02492711) results, margetuximab was approved by the FDA in December 2020 to treat HER2+ MBC when combined with chemotherapy.Citation22 In mice, margetuximab showed activity against HER2-low expressing tumor cells.Citation23

Although margetuximab demonstrated an acceptable safety profile in Phase I, II, and III studies,Citation24 in a Phase II study, it has not shown sufficient clinical efficacy. In 22 showing 2+ by IHC without amplification patients whose efficacy results were available, no responses were achieved, and only 6 disease stabilizations were observed. Notably, when combined with pembrolizumab (an anti-PD-1 monoclonal antibody), it showed clinical efficacy in HER2-low gastro-oesophageal adenocarcinoma.Citation25 This suggests that the therapeutic effects of margetuximab may be displayed when combined with other treatments.

Antibody–Drug Conjugates

ADCs combine the targeted specificity of monoclonal antibodies with the antitumor’s ability of cytotoxic drugs to improve the therapeutic index. An ADC is composed of three elements, a monoclonal antibody to hit the specific molecular target of tumor cells and internalize the entire ADC complex via endocytosis, a cytotoxic agent (also called “payload”) to kill tumor cells, and a chemical linker to attach the cytotoxic agent to the antibody.Citation26 A factor influencing the therapeutic index is the number of molecules of payload linked to each antibody, which is called the drug-to-antibody ratio (DAR). The treatment may be less effective if the DAR is lower, while it may be more difficult to tolerate adverse effects if the DAR is higher.Citation27 If the released payload is permeable, it can enter and kill the neighboring cells, not only the antigen‐positive cells. The phenomenon is called the “bystander effect”, which is also one of the reasons for its efficacy to treat HER2-low BC.Citation28 summarizes the ADCs which are useful for the treatment of HER2-low BC.

Table 1 The ADCs and Their Trials for the Treatment of HER2-Low BC

T-DM1 was the first ADC approved by the US Food and Drug Administration (FDA) for the treatment of HER2-positive metastatic breast cancer (MBC) in 2013. The payload is DM1, which connect with trastuzumab via a non-cleavable linker, and the DAR is 3.5.Citation29 Retrospective analyses found there was no benefit that can be derived from T-DM1 in the group of HER2-low BC by reviewing HER2 status. In the phase II trial of TDM4258g, HER2-low BC had a poorer clinical efficacy than HER2-positive with an insufficient progression-free survival (PFS) (2.6 vs 8.2 months) and a lower objective response rate (ORR) (4.8% vs 33.8%).Citation30 Similar results were also shown in the TDM4374g trial.Citation31 The lack of bystander effects limits its utility in tumors with HER2-low.

Trastuzumab deruxtecan (T-DXd) is the second ADC approved by the FDA. It is composed of humanized HER2-targeted antibody and DXd (derivative of exatecan), conjugated through enzyme-cleavable linker making it stable in plasma.Citation32,Citation33 The payload is cell membrane permeable, so it has bystander effect to kill tumor cells in proximity regardless of their HER2 expression status.Citation34 At the same time, DXd is a topoisomerase I inhibitor, which is different from the mechanism of chemotherapy drugs commonly used in the treatment of BC, resulting in a reduced risk of cross-drug resistance. Besides, T-DXd also has a higher DAR than T-DM1 (8 vs 3.5), allowing it to deliver more payload molecules to targeted tumor cells.

Based on the results of DESTINY-Breast01 (NCT03248492), T-DXd has been approved for posterior line therapy in patients with HER2-positive BC by the FDA with a confirmed ORR of 61.4% (95% CI 54.0 to 68.5) and a median duration of response (mDOR) of 20.8 months (95% CI 15.0 to NE).Citation35 A Phase I trial (NCT02564900) has reported the efficacy of T-DXd on HER2-low MBC. Preliminary antitumor activity has been demonstrated with the confirmed ORR of 37.0% (20/54; 95% CI, 24.3% to 51.3%) and the DOR was 10.4 months (95% CI, 8.8 months to not evaluable).Citation36 Another study showed that ORR and DCR of HER2-low BC patients were marginally lower than HER2-positive BC ones who prior treated with T-DM1 alone or in combination with pertuzumab.Citation37 DESTINY-Breast04 (NCT03734029) is the first phase III trial to test the efficacy of T-DXd in HER2-low MBC patients. Compared with the physician’s choice arm, superior PFS and OS were observed in the T-DXd group (mPFS: 10.1 vs 5.4 months; OS: 23.9 vs 17.5 months). Fewer grade ≥3 treatment-emergent adverse events occurred (52.6% vs 67.4%).Citation38 Benefit also showed the improvement in health-related quality of life.Citation39 T-DXd was well tolerated and had significant activity effective in patients with HER2-low BC. T-DXd also demonstrated evidence of central nervous system activity in HER2-positive and HER2-low orthotopic patient-derived xenografts models of MBC with brain metastases, which are resistant to T-DM1.Citation40 Unlike T-DM1, T-DXd is effective in patients with low-HER2 BC. The DEBBRAH trial assessed the efficacy and safety of T-DXd in HER2-positive and HER2-low ABC patients with a history of brain metastases and/or leptomeningeal carcinomatosis. The trail has been completed, but the data are not reported.Citation41 Another three trials evaluating the efficacy of T-DXd in treating HER2-low BC are ongoing (NCT04494425, NCT04556773, NCT04553770),Citation42–44 and one is evaluating the clinical activity of neoadjuvant T-DXd and T-DXd/endocrine therapy in patients with HR+/HER2-low early BC.Citation45 The publication of the results of these studies may further expand the therapeutic scope of T-DXd, which is effective in both HER2-low and HER2-positive BC.

Trastuzumab duocarmazine (SYD985) is a novel ADC composed of trastuzumab, duocarmycin, and a cleavable linker.Citation46 Despite it has a lower DAR of 2.8, SYD985 has been shown significantly more potent than T-DM1 in HER2-low BC. Dokter et al predicted that its membrane-permeable nature and the cleavable linker in SYD985 make it has great bystander effects.Citation47 It has shown efficacy on HER2-low MBC in a phase I trial (NCT02277717), and the ORR in HR-positive and triple-negative BC were 27% (N=9) and 40% (N=7) respectively, compared with 33% (N=16) in HER2-positive BC.Citation48 That indicates SYD985 might be another treatment option in future for the treatment of HER2-low BC.

MRG002 refers to a novel ADC conjugating monomethyl auristatin E derivative (MMAE) to humanized monoclonal antibody of MAB802 via a protease cleavable valine-citrulline linker with an average DAR of  3.8.Citation49 At the 2022 AACO Annual Meeting, the results of a phase II (NCT04742153) clinical trial were released. This trial tested MRG002 in 66 patients with HER2-low MBC. The trial showed promising efficacy and a favorable safety profile with an ORR of 34.7% and a DCR of 75.5%, and the most common treatment related adverse events were grade 1 or 2. ORR was similar in the HER2 IHC1+ and IHC2+ subgroups (34.1% VS 37.5%). TNBC patients also benefited from MAB802 with ORR (37.5%) and DCR (62.5%).Citation50 MRG002 is a highly promising ADC for the treatment of HER2-low BC.

A66 is also an ADC target HER2, which adopts a stable protease-cleavable valine citrulline linker and monomethyl auristatin F derivative. In Phase I–II, the first-in-human study for A166 (NCT03602079), the efficacy of HER2-low BC was evaluated in Phase II, cohort 3 experimental group. The relevant results will be published shortly.Citation51 PF-06804103 is an ADC with anti-HER2 immunoglobulin G1, which is conjugated with a cleavable linker to the cytotoxic agent auristatin microtubule inhibitor Aur0101.Citation52 A site-specific ADC PF-06804103 was tested in patients with HER2-positive and HER2-negative BC in the first-in-human trial (NCT03284723).Citation53 In this study, HER2-negative performed status of 0 or 1. It also explored the combination of PF-06804103, palbociclib and letrozole. The study has been completed but the results are not yet available. RC48-ADC was generated by conjugating MMAE to a humanized anti-HER2 antibody via a protease cleavable valine-citrulline linker. A pooled analysis of two studies (C001 CANCER, NCT02881138; C003 CANCER, NCT03052634) reported the ORR of HER2-low BC was 39.6% and mPFS was 5.7 months. Patients with IHC2+/FISH- BC had better outcomes than IHC1+ ones.Citation54 BC with different HER2 status responds differently to RC48-ADC. Meanwhile, a phase III trial (NCT04400695) is ongoing to compare RC48-ADC with the physician’s choice in HER2-low BC.

HER2-Targeted Bispecific Antibodies (bsAbs)

bsAbs have two different antigen-binding sites to address different antigens or epitopes, promoting immune cell recruitment and activation.Citation55 ZW25 (zanidatamab) is a novel HER2-targeted bsAb, targeting HER2 domains ECD2 and ECD4, for which antitumor activity, synergy and additivity with multiple chemotherapeutic agents have been exhibited in HER2-low to -high expressing models.Citation56 A phase I clinical trial (NCT02892123) is investigating ZW25 in HER2-expressing tumors, of which cohort 4 and 7 in part 1, cohort 1 in part 2 and part 3 involves HER2 IHC2+/FISH- BC. The study is still ongoing with no results about HER2-low BC published. Combining a novel auristatin payload with ZW25, an ADC ZW49 is designed. ZW49 has demonstrated antitumor activity in HER2-low BC cell lines and patient-derived xenograft (PDX) models.Citation57 However, there are no relevant clinical trials evaluating efficacy in the HER2-low BC population.

HER2/HER3-Targeted Therapies

ErBb family includes four distinct receptors, HER1 (EGFR or ErbB1), HER2, HER3, and HER4. HER2 overexpression is associated with the activation of HER3, and some models show that HER2-driven tumors do not develop or grow if HER3 expression is absent.Citation58–60 Treatments show efficacy in HER2-low BC when targeting both HER2 and HER3. The HER2/HER3 heterodimer induces phosphorylation of the ER and endocrine resistance.Citation61 Preclinical data showed that the triple combination of anti-HER2 (pertuzumab), anti-HER3 (lumretuzumab), and anti-estrogen therapy (fulvestrant) lead to long-lasting responses in ER+/HER2-low/HER3+ human BC HBCx-19 xenograft model.Citation62 A study (NCT01918254) evaluated the clinical activity and safety of lumretuzumab, pertuzumab and paclitaxel in HER3-positive, HER2-low MBC. Chronic diarrhea is the major side effect; thus, the initial antitumor activity could not be confirmed.Citation63 Mitigating the toxic effects of combination therapies is a key issue in achieving effectiveness.

The bsAbs targeting HER2/HER3 are undergoing clinical development. MCLA-128 (zenocutuzumab) is a humanized bsAb targeting HER2 and HER3. The addition of MCLA-128 to estrogen therapy demonstrated to a better antitumor effect than estrogen therapy alone in HER2-low BC xenograft models.Citation64 It also showed clinical activity in patients with ER-positive, HER2-low BC after estrogen therapy and CDK4/6i failure according to a phase II trial (NCT03321981). The disease control rate (DCR) was 45% with 2 patients having an unconfirmed partial response and 19 patients keeping the disease stable. It also showed a favorable safety profile with no grade 5 adverse events or diarrhea requiring treatment discontinuation observed.Citation65 Bispecific humanized IgG1 antibody is safer than the combination therapies when targeting HER2/HER3.

HER2/Other Antigenic Sites-Targeted Therapies

Some therapeutic approaches block both HER2 and other antigenic sites. Evorpacept (ALX148) is a high-affinity CD47-blocking fusion protein that enhances the activity of other antitumor therapies. The safety and antitumor activity of HER2-targeted bsAbs ZX25 in combination with ALX148 is being assessed in a phase Ib/II study (NCT05027139).Citation66 SAR443216 is a trispecific antibody with binding sites for HER2, CD3 and CD28. It is a HER2-targeted T cell engager that can activate CD4 and CD8 T-cells and has the activity of T cell-dependent cellular cytotoxicity (TDCC) against HER2-expressing tumor.Citation67 Two hundred patients with HER2-expressing (various levels of HER2 expression) solid tumors including BC participate in a trial (NCT05013554). The preliminary clinical activity of SAR443216 after intravenous and subcutaneous administration will be assessed.

Immunotherapy

Immune Checkpoint Inhibitor

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of immunogenic cancers by enabling the priming and infiltration of T-cells into the tumor microenvironment, promoting cytotoxic signaling pathways and affecting tumor cytolysis by specifically recognizing, binding to cancer cells and killing them.Citation68 BC is a moderately immunogenic cancer,Citation69 in which TNBC and HER2-positive BC have the highest immunogenic potential.Citation70 However, intrinsic resistance is a big problem to the use of ICIs, which occurs in 60–85% of the patients with TNBC.Citation71 Some cytotoxic drugs can induce immunogenic cell death in tumor cells, promote tumor antigen release and activate the immune system.Citation72 HER2-targeted drugs can kill tumor cells by the immune-mediated mechanism of antibody-dependent cellular cytotoxicity (ADCC). Therefore, ICIs are often combined with monoclonal antibodies and ADCs. The combination of trastuzumab with ICIs could overcome trastuzumab resistance in a study.Citation73 Six of 40 programmed cell death 1 ligand 1 (PD-L1) -positive-patients achieved an objective response, but no objective responders among the PD-L1-negative patients.Citation74 The payload DXd of T-DXd is a topoisomerase inhibitor, which has gained attention as an immunomodulator.Citation75 Mechanically, T-DXd is an ideal partner for ICIs. In preclinical studies, T-DXd increased the efficacy of ICIs by increasing immune cell infiltration and upregulating PD-L1 expression when combined with anti-PD-L1 antibody.Citation76

Programmed cell death protein 1 (PD-1) and its ligand PD-L1 have been evaluated as putative markers of response to immunotherapy with PD-1/PD-L1 blockade in BC.Citation77 Anti-PD-1 or anti-PD-L1 therapies have been investigated in combination with HER2-targeted therapies. T-DXd combined with a PD-1 inhibition was more effective than monotherapy with either agent in vitro studies. Nivolumab is the first PD-1 blocking antibody approved for clinical use, and there are many ongoing trials to assess the safety and efficacy of nivolumab combined with other therapies for BC. The combination of T-DXd and nivolumab was tested in a phase Ib trial (NCT03523572). Cohort 2 involved 16 patients with HER2-low MBC who had exhausted any clinically meaningful treatments. After 12.7 months of follow-up, the confirmed ORR (cORR) was 50% and the median PFS was 7.0 months comparing the HER2-positive cohort with a cORR of 65.6% and median PFS of 11.6 months.Citation78 Combining nivolumab may result in an improved ORR compared to T-DXd alone. Pembrolizumab is another PD-1 inhibition showing efficacy in this subgroup when combined with T-DXd. In the dose extension of part 2 of a trial (NCT04042701), patients with HER2-low BC were treated with pembrolizumab and T-DXd.Citation79 The estimated completion date of the trial is in May 2023 and ORR is the primary outcome measure.

PD-L1 binds to PD-1 and CD80 receptors, resulting in the inhibition of T-cell function, which can be blocked by durvalumab.Citation80 Randomized phase Ib/II study BEGONIA reported its initial results, in which cORR was 100% (4/4) and median DOR was not reached.Citation81 Additionally, DESTINY-Breast08 is also comparing the combination of T-DXd, durvalumab and paclitaxel with others (T-DXd combines with endocrine therapy, chemotherapy, and immunotherapy). No deaths or cases of interstitial lung disease/pneumonitis were reported so far.Citation82 T-DM1 in combination with atezolizumab (a PD-L1 inhibitor) was effective only in PD-L1-positive BC in the trial KATE2 (NCT02924883), which suggested that the benefit of the combination of PD-L1 inhibitors may be limited to PD-L1-positive patients.Citation83 It may also be helpful to assess the patient’s PD-L1 status to better evaluate the efficacy of T-DXd in combination with immunotherapy. The combination trials with ICIs to treat HER2-low BC are listed in .

Table 2 Trials of ICIs in Combination with Other Drugs

Apart from these, immune-stimulating antibody conjugates (ISACs) are also under development in HER2-positive BC and TNBC.Citation84 An ISAC comprises a tumor-targeting monoclonal antibody conjugated to an immune agonist, exerting durable antitumor immunity.Citation85 They are well-tolerated in vivo. For instance, BDC-1001 a novel ISAC and it did not induce interstitial lung disease, cytokine release syndrome, or thrombocytopenia in non-human primate studies.Citation86

HER2-Derived Vaccines

Unlike trastuzumab and pertuzumab, which are passive immunotherapy, HER2-derived vaccines are active immunotherapy. Cancer vaccines are designed to elicit or enhance antitumor immune responses by activating immune cells to induce a therapeutic effect.Citation87 HER2 is a tumor-associated antigen identified as an appropriate vaccine source in BC.Citation88 HER2-derived vaccines have been studied extensively in HER2-positive BC, and several trials are also conducted in HER-low BC recently.

E75 (Nelipepimut-S, NPS, KIFGLSAFL) vaccine is one of the most widely researched BC vaccines against HER2. In an early clinical study, patients with HER2-low specifically IHC 1(+) BC benefited more from E75 vaccine than those with higher levels of HER2 expression.Citation89 However, the phase III trial PRESENT (NCT01479244) terminated due to failure to show a significant difference in DFS in 758 patients with HER2-low, node-positive BC.Citation90 A phase IIb trial (NCT01570036) enrolled 275 disease-free patients after standard therapy completion. Treated with the combination of E75, granulocyte-macrophage colony-stimulating factor and trastuzumab, the estimated DFS of HER2-low patients did not significantly differ from the control group (HR, 0.62; 95% CI, 0.31–1.25; P= 0.18).Citation91 However, the combination improved 36-month DFS among patients with TNBC (HR, 0.25; 95% CI, 0.08–0.78, p = 0.01) and HLA-A24 (HR, 0.41; 95% CI, 0.16–1.04; P= 0.05) positivity according to a subgroup analysis.Citation92 There may be a synergistic effect between E75 and HER2-targeted therapy, and further investigation is warranted to be confirmed.

AE37 is another HER2-related peptide vaccine to treat BC which can induce CD8 and CD4 cells.

A trial (NCT00524277) enrolled 298 patients with disease-free node-positive and high-risk node-negative BC expressing any degree of HER2. After receiving AE37 and GM-CSF, 5-year DFS was 77.2% versus 65.7% of the control group in planned subset analyses of HER2-low patients (P=0.21). Specifically, patients with TNBC are more likely to have clinical benefits in DFS (77.7% vs 49.0%, P = 0.12).Citation93

The above two vaccines with a tolerable and favorable safety profile. There are some other vaccines tested in patients with HER2-low BC. Cornerstone-001 (NCT05163223) is a phase II trial to assess the efficacy and safety of pNGVL3-hICD (AST-301) plasmid-based vaccine in patients with HER2-low and HR-negative BC with residual disease after neoadjuvant treatment. The trial started on 28 February 2022 and is expected to be completed in December 2025. ETBX-021 is another HER2-targeted vaccine that is being evaluated in phase I clinical trial (NCT02751528) with locally advanced or metastatic HER2-low BC.

CDK4/6 Inhibitors

CDK4/6 is downstream of HER2 and driving resistance to HER2-targeted therapies.Citation94 CDK4/6 inhibitors not only induce tumor cell cycle arrest but also promote antitumor immunity by enhancing tumor antigen presentation and impacting immunosuppressive cells such as CD4 T-cells.Citation95,Citation96 The phase II trial of NA-PHER2 evaluated the effects of Ki67 and apoptosis after neoadjuvant pertuzumab, trastuzumab, fulvestrant and palbociclib (a CDK4/6 inhibitor). In cohort C, 23 women with ER-positive and HER2-low BC participated. More than 90% tumors showed a drop of Ki67 below 10% at week 2 and complete cell cycle arrest was achieved in 65%. The ORR before surgery was 78.3%, but no patient achieved a pathologic complete response.Citation97 HER2-low status does not affect survival outcomes of patients with MBC,Citation98 and CDK4/6 inhibitors may play an important role in the treatment of HER2-low BC when combined with HER2-targeted therapies.

Others

EZH1 and EZH2 are two markers of aggressive BC and are associated with invasion and cancer progression.Citation99,Citation100 Valemetostat (DS-3201) is a novel and potent EZH1/2 dual inhibitor. A study (NCT05633979) is designed to test the efficacy and safety of the combination of valemetostat and T-DXd in HER2 low/ultra-low/null (IHC2+/ISH-, IHC1+; IHC0 with detectable faint/barely perceptible incomplete staining in ≤10% tumor cells; IHC0 without any observed tumor cell staining) MBC patients. The study is estimated to start on 23 May 2023. Some tyrosine kinase inhibitors are pan-HER kinase inhibitors already widely used in HER2-positive MBC.Citation101 Lapatinib (a tyrosine kinase inhibitor) increased HER2 levels and potentiated ADCC in preclinical models.Citation102 Adding endocrine therapy may improve the efficacy of HER2- and HER3-targeted agents according to a study.Citation62 Likewise, HER2-targeted agents play a role in the resistance to endocrine therapy,Citation103 and the HER2-negative patients gained greater benefit from aromatase inhibitors following the TRANS-AIOG meta-analysis.Citation104

Discussion

IHC/ISH is the gold standard to define HER2 expression. However, some factors remarkably affect the analytical reliability.Citation105 Although HER2-low expression has not been formally defined, HER2-low BC is different from HER2-negative cases for which the former can benefit from many anti-HER2 therapies. HER2-low BC with IHC1+ and 2+ may respond differently to HER2-targeted therapies.Citation54 So more precise identification is needed by harmonizing testing strategy and technique. HER2 gene–protein assay (HER2 GPA) can be used for assessing HER2 status of HER2-positive, equivocal, and -negative BC by combining HER2 ISH and HER2 IHC assays.Citation106 Other techniques also facilitate the determination of HER2 status such as automated image analysis and digital PCR.Citation107,Citation108

Approximately half of BC was HER2-lowCitation109; in some cases, HER2-negative eventually converted to HER2-low phenotype.Citation110 A study showed that after neoadjuvant therapy, HER2-low converted from 14.8% of the primary tumors to 8.9% of residual disease.Citation110 Patients with ER-positive HER2-low residual disease have a high risk of relapse compared to ER-positive HER2-negative cases, who may need HER2-targeted therapies.Citation110 A similar evolution of HER2-low expression can also be observed from primary to recurrent.Citation111 HR-positive HER2-negative BC faces the problem of drug resistance to endocrine therapy and TNBC has limited treatment options. A large population of patients have BC, which is a highly unmet medical need.

ADCs play an important role in the treatment of this subtype and T-DXd may be firstly approved because of the bystander effect and its cell membrane permeable payload. HER2-targeted therapies often show clinical efficacy when combined with other treatments, such as immunotherapy.Citation79 There are also strategies that target both HER2 and HER3 or other antigenic sites.Citation64 These treatments are usually well tolerated. In many cases, serious adverse events occur after using different drugs, such as the combination of lumretuzumab, pertuzumab and paclitaxel.Citation63 The design of the conjugates better limits the adverse effects. More and more studies will be conducted to explore the efficacy of HER2-targeted therapies for HER2-low BC in the future, and more patients will benefit from these treatments.

Disclosure

There are no conflicts of interest.

Additional information

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References

  • Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.21660
  • Dawood S, Broglio K, Buzdar AU, Hortobagyi GN, Giordano SH. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J Clin Oncol. 2010;28(1):92–98. doi:10.1200/jco.2008.19.9844
  • Ishii K, Morii N, Yamashiro H. Pertuzumab in the treatment of HER2-positive breast cancer: an evidence-based review of its safety, efficacy, and place in therapy. Core Evid. 2019;14:51–70. doi:10.2147/ce.S217848
  • Swain SM, Miles D, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): end-of-study results from a double-blind, randomised, placebo-controlled, Phase 3 study. Lancet Oncol. 2020;21(4):519–530. doi:10.1016/s1470-2045(19)30863-0
  • Diéras V, Miles D, Verma S, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18(6):732–742. doi:10.1016/s1470-2045(17)30312-1
  • Keam SJ. Trastuzumab deruxtecan: first approval. Drugs. 2020;80(5):501–508. doi:10.1007/s40265-020-01281-4
  • Fehrenbacher L, Cecchini RS, Geyer CE, et al. NSABP B-47/NRG oncology phase III randomized trial comparing adjuvant chemotherapy with or without trastuzumab in high-risk invasive breast cancer negative for HER2 by FISH and with IHC 1+ or 2. J Clin Oncol. 2020;38(5):444–453. doi:10.1200/jco.19.01455
  • Ahn S, Woo JW, Lee K, Park SY. HER2 status in breast cancer: changes in guidelines and complicating factors for interpretation. Jptm. 2019;54(1):34–44. doi:10.4132/jptm.2019.11.03
  • Schettini F, Chic N, Brasó-Maristany F, et al. Clinical, pathological, and PAM50 gene expression features of HER2-low breast cancer. Npj Breast Cancer. 2021;7(1):1. doi:10.1038/s41523-020-00208-2
  • Marchiò C, Annaratone L, Marques A, Casorzo L, Berrino E, Sapino A. Evolving concepts in HER2 evaluation in breast cancer: heterogeneity, HER2-low carcinomas and beyond. Semin Cancer Biol. 2021;72:123–135. doi:10.1016/j.semcancer.2020.02.016
  • Gordian-Arroyo AM, Zynger DL, Tozbikian GH. Impact of the 2018 ASCO/CAP HER2 guideline focused update. Am J Clin Pathol. 2019;152(1):17–26. doi:10.1093/ajcp/aqz012
  • Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30(8):1194–1220. doi:10.1093/annonc/mdz173
  • Li X, Yang J, Peng L, et al. Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res Treat. 2017;161(2):279–287. doi:10.1007/s10549-016-4059-6
  • Berrino E, Annaratone L, Bellomo SE, et al. Integrative genomic and transcriptomic analyses illuminate the ontology of HER2-low breast carcinomas. Genome Med. 2022;14(1):98. doi:10.1186/s13073-022-01104-z
  • Horisawa N, Adachi Y, Takatsuka D, et al. The frequency of low HER2 expression in breast cancer and a comparison of prognosis between patients with HER2-low and HER2-negative breast cancer by HR status. Breast Cancer. 2022;29(2):234–241. doi:10.1007/s12282-021-01303-3
  • Denkert C, Seither F, Schneeweiss A, et al. Clinical and molecular characteristics of HER2-low-positive breast cancer: pooled analysis of individual patient data from four prospective, neoadjuvant clinical trials. Lancet Oncol. 2021;22(8):1151–1161. doi:10.1016/s1470-2045(21)00301-6
  • van den Ende NS, Smid M, Timmermans A, et al. HER2-low breast cancer shows a lower immune response compared to HER2-negative cases. Sci Rep. 2022;12(1):12974. doi:10.1038/s41598-022-16898-6
  • Clarke R, Liu MC, Bouker KB, et al. Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene. 2003;22(47):7316–7339. doi:10.1038/sj.onc.1206937
  • Gianni L, Lladó A, Bianchi G, et al. Open-label, phase II, multicenter, randomized study of the efficacy and safety of two dose levels of Pertuzumab, a human epidermal growth factor receptor 2 dimerization inhibitor, in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol. 2010;28(7):1131–1137. doi:10.1200/jco.2009.24.1661
  • Ramić S, Asić K, Balja MP, Paić F, Benković V, KneŽEviĆ F. Correlation of phosphorylated HER2 with clinicopathological characteristics and efficacy of trastuzumab treatment for breast cancer. Anticancer Res. 2013;33(6):2509.
  • Onsum MD, Geretti E, Paragas V, et al. Single-cell quantitative HER2 measurement identifies heterogeneity and distinct subgroups within traditionally defined HER2-positive patients. Am J Pathol. 2013;183(5):1446–1460. doi:10.1016/j.ajpath.2013.07.015
  • Rugo HS, Im S-A, Wright GLS, et al. SOPHIA primary analysis: a phase 3 (P3) study of margetuximab (M) + chemotherapy (C) versus trastuzumab (T) + C in patients (pts) with HER2+ metastatic (met) breast cancer (MBC) after prior anti-HER2 therapies (Tx). J Clin Oncol. 2019;37(15_suppl):1000. doi:10.1200/JCO.2019.37.15_suppl.1000
  • Nordstrom JL, Gorlatov S, Zhang W, et al. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res. 2011;13(6):R123. doi:10.1186/bcr3069
  • Im S-A, Cardoso F, Cortes J, et al. Abstract PS10-12: integrated safety summary of single agent and combination margetuximab in Phase 1, 2, and 3 studies of HER2-positive advanced cancers and metastatic breast cancer (MBC). Cancer Res. 2021;81(4_Supplement):PS10-12-PS10-12. doi:10.1158/1538-7445.Sabcs20-ps10-12
  • Catenacci DVT, Kang Y-K, Park H, et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22–05): a single-arm, phase 1b–2 trial. Lancet Oncol. 2020;21(8):1066–1076. doi:10.1016/S1470-2045(20)30326-0
  • Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct Target Ther. 2022;7(1):93. doi:10.1038/s41392-022-00947-7
  • Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. 2017;16(5):315–337. doi:10.1038/nrd.2016.268
  • Kovtun YV, Audette CA, Ye Y, et al. Antibody-drug conjugates designed to eradicate tumors with homogeneous and heterogeneous expression of the target antigen. Cancer Res. 2006;66(6):3214–3221. doi:10.1158/0008-5472.Can-05-3973
  • Lambert JM, Chari RV. Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J Med Chem. 2014;57(16):6949–6964. doi:10.1021/jm500766w
  • Burris HA, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol. 2011;29(4):398–405. doi:10.1200/jco.2010.29.5865
  • Krop IE, LoRusso P, Miller KD, et al. A Phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2–positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol. 2012;30(26):3234–3241. doi:10.1200/JCO.2011.40.5902
  • Xu Z, Guo D, Jiang Z, et al. Novel HER2-targeting antibody-drug conjugates of trastuzumab beyond T-DM1 in breast cancer: trastuzumab deruxtecan (DS-8201a) and (Vic-)trastuzumab duocarmazine (SYD985). Eur J Med Chem. 2019;183:111682. doi:10.1016/j.ejmech.2019.111682
  • Indini A, Rijavec E, Grossi F. Trastuzumab deruxtecan: changing the destiny of HER2 expressing solid tumors. Int J Mol Sci. 2021;22(9):4774.
  • Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107(7):1039–1046. doi:10.1111/cas.12966
  • Modi S, Saura C, Yamashita T, et al. Abstract PD3-06: updated results from DESTINY-breast01, a Phase 2 trial of trastuzumab deruxtecan (T-DXd) in HER2 positive metastatic breast cancer. Cancer Res. 2021;81(4_Supplement):PD3-06-PD3-06. doi:10.1158/1538-7445.Sabcs20-pd3-06
  • Modi S, Park H, Murthy RK, et al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-Low-expressing advanced breast cancer: results from a Phase Ib study. J Clin Oncol. 2020;38(17):1887–1896. doi:10.1200/jco.19.02318
  • Modi S, Tsurutani J, Takahashi S, et al. Abstract PD3-07: safety and efficacy results from a phase 1 study of DS-8201a in patients with HER2 expressing breast cancers. Cancer Res. 2018;78(4_Supplement):PD3-07-PD3-07. doi:10.1158/1538-7445.Sabcs17-pd3-07
  • Modi S, Jacot W, Yamashita T, et al. Trastuzumab deruxtecan (T-DXd) versus treatment of physician’s choice (TPC) in patients (pts) with HER2-low unresectable and/or metastatic breast cancer (mBC): results of DESTINY-Breast04, a randomized, phase 3 study. J Clin Oncol. 2022;40(17_suppl):LBA3–LBA3. doi:10.1200/JCO.2022.40.17_suppl.LBA3
  • Ueno NT, Jacot W, Yamashita T, et al. 217O Patient-reported outcomes (PROs) from DESTINY-Breast04, a randomized phase III study of trastuzumab deruxtecan (T-DXd) vs treatment of physician’s choice (TPC) in patients (pts) with HER2-low metastatic breast cancer (MBC). Ann Oncol. 2022;33:S632–S633. doi:10.1016/j.annonc.2022.07.256
  • Kabraji S, Ni J, Sammons S, et al. Preclinical and clinical efficacy of trastuzumab deruxtecan in breast cancer brain metastases. Clin Cancer Res. 2022. doi:10.1158/1078-0432.Ccr-22-1138
  • Batista MV, Cortez P, Ruiz M, et al. Abstract PD4-06: trastuzumab deruxtecan in patients with HER2[+] or HER2-low-expressing advanced breast cancer and central nervous system involvement: preliminary results from the DEBBRAH phase 2 study. Cancer Res. 2022;82(4_Supplement):PD4-06-PD4-06. doi:10.1158/1538-7445.Sabcs21-pd4-06
  • Bardia A, Barrios C, Dent R, et al. Abstract OT-03-09: trastuzumab deruxtecan (T-DXd; DS-8201) vs investigator’s choice of chemotherapy in patients with hormone receptor-positive (HR+), HER2 low metastatic breast cancer whose disease has progressed on endocrine therapy in the metastatic setting: a randomized, global phase 3 trial (DESTINY-Breast06). Cancer Res. 2021;81(4_Supplement):OT-03-09-OT-03-09. doi:10.1158/1538-7445.Sabcs20-ot-03-09
  • Jhaveri K, Hamilton E, Loi S, et al. Abstract OT-03-05: trastuzumab deruxtecan (T-DXd; DS-8201) in combination with other anticancer agents in patients with HER2-low metastatic breast cancer: a phase 1b, open-label, multicenter, dose-finding and dose-expansion study (DESTINY-Breast08). Cancer Res. 2021;81(4_Supplement):OT-03-05-OT-03-05. doi:10.1158/1538-7445.Sabcs20-ot-03-05
  • Hurvitz SA, Peddi PF, Tetef ML, et al. TRIO-US B-12 TALENT: phase II neoadjuvant trial evaluating trastuzumab deruxtecan with or without anastrozole for HER2-low, HR+ early stage breast cancer. J Clin Oncol. 2021;39(15_suppl):TPS603. doi:10.1200/JCO.2021.39.15_suppl.TPS603
  • Hurvitz SA, Wang LS, Chan D, et al. TRIO-US B-12 TALENT: phase II neoadjuvant trial evaluating trastuzumab deruxtecan with or without anastrozole for HER2-low, HR+ early-stage breast cancer. J Clin Oncol. 2022;40(16_suppl):TPS623. doi:10.1200/JCO.2022.40.16_suppl.TPS623
  • Elgersma RC, Coumans RG, Huijbregts T, et al. Design, synthesis, and evaluation of linker-duocarmycin payloads: toward selection of HER2-targeting antibody-drug conjugate SYD985. Mol Pharm. 2015;12(6):1813–1835. doi:10.1021/mp500781a
  • Dokter W, Ubink R, van der Lee M, et al. Preclinical profile of the HER2-targeting ADC SYD983/SYD985: introduction of a new duocarmycin-based linker-drug platform. Mol Cancer Ther. 2014;13(11):2618–2629. doi:10.1158/1535-7163.Mct-14-0040-t
  • Saura C, Thistlethwaite F, Banerji U, et al. A phase I expansion cohorts study of SYD985 in heavily pretreated patients with HER2-positive or HER2-low metastatic breast cancer. J Clin Oncol. 2018;36(15_suppl):1014. doi:10.1200/JCO.2018.36.15_suppl.1014
  • Li H, Zhang X, Xu Z, et al. Preclinical evaluation of MRG002, a novel HER2-targeting antibody-drug conjugate with potent antitumor activity against HER2-positive solid tumors. Antib Ther. 2021;4(3):175–184. doi:10.1093/abt/tbab017
  • Jiang Z, Sun T, Wang X, et al. A multiple center, open-label, single-arm, phase II clinical trial of MRG002, an HER2-targeted antibody-drug conjugate, in patients with HER2-low expressing advanced or metastatic breast cancer. J Clin Oncol. 2022;40(16_suppl):1102. doi:10.1200/JCO.2022.40.16_suppl.1102
  • Liu Y, Lian W, Zhao X, et al. A first in-human study of A166 in patients with locally advanced/metastatic solid tumors which are HER2-positive or HER2-amplified who did not respond or stopped responding to approved therapies. J Clin Oncol. 2020;38(15_suppl):1049. doi:10.1200/JCO.2020.38.15_suppl.1049
  • Graziani EI, Sung M, Ma D, et al. PF-06804103, A site-specific anti-HER2 antibody–drug conjugate for the treatment of HER2-expressing breast, gastric, and lung cancers. Mol Cancer Ther. 2020;19(10):2068–2078. doi:10.1158/1535-7163.Mct-20-0237
  • Meric-Bernstam F, Calvo E, Moreno V, et al. A phase I dose escalation study evaluating the safety and tolerability of a novel anti-HER2 antibody-drug conjugate (PF-06804103) in patients with HER2-positive solid tumors. J Clin Oncol. 2020;38(15_suppl):1039. doi:10.1200/JCO.2020.38.15_suppl.1039
  • Wang J, Liu Y, Zhang Q, et al. RC48-ADC, a HER2-targeting antibody-drug conjugate, in patients with HER2-positive and HER2-low expressing advanced or metastatic breast cancer: a pooled analysis of two studies. J Clin Oncol. 2021;39(15_suppl):1022. doi:10.1200/JCO.2021.39.15_suppl.1022
  • Kontermann RE, Brinkmann U. Bispecific antibodies. Drug Discov Today. 2015;20(7):838–847. doi:10.1016/j.drudis.2015.02.008
  • Weisser N, Wickman G, Davies R, Rowse G. Abstract 31: preclinical development of a novel biparatopic HER2 antibody with activity in low to high HER2 expressing cancers. Cancer Res. 2017;77(13_Supplement):31. doi:10.1158/1538-7445.Am2017-31
  • Hamblett K, Barnscher S, Davies R, et al. Abstract P6-17-13: ZW49, a HER2 targeted biparatopic antibody drug conjugate for the treatment of HER2 expressing cancers. Cancer Res. 2019;79(4_Supplement):P6-17-13-P6-17-13. doi:10.1158/1538-7445.Sabcs18-p6-17-13
  • Vaught DB, Stanford JC, Young C, et al. HER3 is required for HER2-induced preneoplastic changes to the breast epithelium and tumor formation. Cancer Res. 2012;72(10):2672–2682. doi:10.1158/0008-5472.Can-11-3594
  • Lee-Hoeflich ST, Crocker L, Yao E, et al. A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy. Cancer Res. 2008;68(14):5878–5887. doi:10.1158/0008-5472.Can-08-0380
  • Lazrek Y, Dubreuil O, Garambois V, et al. Anti-HER3 domain 1 and 3 antibodies reduce tumor growth by hindering HER2/HER3 dimerization and AKT-induced MDM2, XIAP, and FoxO1 phosphorylation. Neoplasia. 2013;15(3):335–IN40. doi:10.1593/neo.121960
  • Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29(2):217–233. doi:10.1210/er.2006-0045
  • Collins D, Jacob W, Cejalvo JM, et al. Direct estrogen receptor (ER) / HER family crosstalk mediating sensitivity to lumretuzumab and pertuzumab in ER+ breast cancer. PLoS One. 2017;12(5):e0177331. doi:10.1371/journal.pone.0177331
  • Schneeweiss A, Park-Simon T-W, Albanell J, et al. Phase Ib study evaluating safety and clinical activity of the anti-HER3 antibody lumretuzumab combined with the anti-HER2 antibody pertuzumab and paclitaxel in HER3-positive, HER2-low metastatic breast cancer. Invest New Drugs. 2018;36(5):848–859. doi:10.1007/s10637-018-0562-4
  • Geuijen CAW, De Nardis C, Maussang D, et al. Unbiased combinatorial screening identifies a bispecific IgG1 that potently inhibits HER3 signaling via HER2-guided ligand blockade. Cancer Cell. 2018;33(5):922–936.e10. doi:10.1016/j.ccell.2018.04.003
  • Pistilli B, Wildiers H, Hamilton EP, et al. Clinical activity of MCLA-128 (zenocutuzumab) in combination with endocrine therapy (ET) in ER+/HER2-low, non-amplified metastatic breast cancer (MBC) patients (pts) with ET-resistant disease who had progressed on a CDK4/6 inhibitor (CDK4/6i). J Clin Oncol. 2020;38(15_suppl):1037. doi:10.1200/JCO.2020.38.15_suppl.1037
  • Hurvitz SA, Chaves J, Brufsky A, et al. Abstract OT1-14-01: zanidatamab in combination with ALX148 in advanced human epidermal growth factor receptor 2 (HER2)-expressing cancers, including breast cancer: a phase 1b/2, multicenter, open-label, dose-finding and cohort-expansion study (ZWI-ZW25-204). Cancer Res. 2022;82(4_Supplement):OT1-14-01-OT1-14-01. doi:10.1158/1538-7445.Sabcs21-ot1-14-01
  • Sha W, Vadde S, Song Z, et al. Abstract 1825: SAR443216, a novel trispecific T cell engager with potent T cell-dependent cytotoxicity for HER2-low tumors. Cancer Res. 2021;81(13_Supplement):1825. doi:10.1158/1538-7445.Am2021-1825
  • Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10. doi:10.1016/j.immuni.2013.07.012
  • Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi:10.1038/nature12477
  • Brigham & Women’s Hospital & Harvard Medical School Chin Lynda 9 11 Park Peter J. 12 Kucherlapati Raju 13, Genome data analysis: Baylor College of Medicine Creighton Chad J. 22 23 Donehower Lawrence A. 22 23 24 25, Institute for Systems Biology Reynolds Sheila 31 Kreisberg Richard B. 31 Bernard Brady 31 Bressler Ryan 31 Erkkila Timo 32 Lin Jake 31 Thorsson Vesteinn 31 Zhang Wei 33 Shmulevich Ilya 31, Oregon Health & Science University Anur Pavana 37 Spellman Paul T. 37. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi:10.1038/nature11412
  • Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer — expanded options, evolving needs. Nat Rev Clin Oncol. 2022;19(2):91–113. doi:10.1038/s41571-021-00565-2
  • Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2014;21(1):15–25. doi:10.1038/cdd.2013.67
  • Loi S, Giobbe-Hurder A, Gombos A, et al. Abstract GS2-06: Phase Ib/II study evaluating safety and efficacy of pembrolizumab and trastuzumab in patients with trastuzumab-resistant HER2-positive metastatic breast cancer: results from the PANACEA (IBCSG 45-13/BIG 4-13/KEYNOTE-014) study. Cancer Res. 2018;78(4_Supplement):GS2-06-GS2-06. doi:10.1158/1538-7445.Sabcs17-gs2-06
  • Loi S, Giobbie-Hurder A, Gombos A, et al. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b–2 trial. Lancet Oncol. 2019;20(3):371–382. doi:10.1016/S1470-2045(18)30812-X
  • Kitai Y, Kawasaki T, Sueyoshi T, et al. DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-dependent pathway and reinforce antitumor immunity. J Immunol. 2017;198(4):1649–1659. doi:10.4049/jimmunol.1601694
  • Iwata TN, Ishii C, Ishida S, Ogitani Y, Wada T, Agatsuma T. A HER2-targeting antibody–drug conjugate, trastuzumab deruxtecan (DS-8201a), enhances antitumor immunity in a mouse model. Mol Cancer Ther. 2018;17(7):1494–1503. doi:10.1158/1535-7163.Mct-17-0749
  • Zerdes I, Matikas A, Bergh J, Rassidakis GZ, Foukakis T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene. 2018;37(34):4639–4661. doi:10.1038/s41388-018-0303-3
  • Hamilton EP, Shapiro CL, Boni V, et al. 162O primary analysis from DS8201-A-U105: a 2-part, open label, phase Ib trial assessing trastuzumab deruxtecan (T-DXd) with nivolumab (nivo) in patients (pts) with HER2-expressing advanced breast cancer. Ann Oncol. 2022;33:S196. doi:10.1016/j.annonc.2022.03.181
  • Borghaei H, Besse B, Bardia A, et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in combination with pembrolizumab in patients with advanced/metastatic breast or non-small cell lung cancer (NSCLC): a phase Ib, multicenter, study. J Clin Oncol. 2020;38(15_suppl):TPS1100. doi:10.1200/JCO.2020.38.15_suppl.TPS1100
  • Stewart R, Morrow M, Hammond SA, et al. Identification and characterization of MEDI4736, an antagonistic anti-PD-L1 monoclonal antibody. Cancer Immunol Res. 2015;3(9):1052–1062. doi:10.1158/2326-6066.Cir-14-0191
  • Schmid P, Im S-A, Armstrong A, et al. BEGONIA: Phase 1b/2 study of durvalumab (D) combinations in locally advanced/metastatic triple-negative breast cancer (TNBC)—Initial results from arm 1, d+paclitaxel (P), and arm 6, d+trastuzumab deruxtecan (T-DXd). J Clin Oncol. 2021;39(15_suppl):1023. doi:10.1200/JCO.2021.39.15_suppl.1023
  • Andre F, Hamilton EP, Loi S, et al. Dose-finding and -expansion studies of trastuzumab deruxtecan in combination with other anti-cancer agents in patients (pts) with advanced/metastatic HER2+ (DESTINY-Breast07 [DB-07]) and HER2-low (DESTINY-Breast08 [DB-08]) breast cancer (BC). J Clin Oncol. 2022;40(16_suppl):3025. doi:10.1200/JCO.2022.40.16_suppl.3025
  • Emens LA, Esteva FJ, Beresford M, et al. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised, double-blind trial. Lancet Oncol. 2020;21(10):1283–1295. doi:10.1016/S1470-2045(20)30465-4
  • Diamond JR, Henry JT, Falchook GS, et al. Abstract CT249: first-in-human study of TAK-500, a novel STING agonist immune stimulating antibody conjugate (ISAC), alone and in combination with pembrolizumab in patients with select advanced solid tumors. Cancer Res. 2022;82(12_Supplement):CT249. doi:10.1158/1538-7445.Am2022-ct249
  • Ackerman SE, Pearson CI, Gregorio JD, et al. Immune-stimulating antibody conjugates elicit robust myeloid activation and durable antitumor immunity. Nat Cancer. 2021;2(1):18–33. doi:10.1038/s43018-020-00136-x
  • Dumbrava EI, Sharma MR, Carvajal RD, et al. Abstract OT-03-02: Phase 1/2 study of a novel HER2 targeting TLR7/8 immune-stimulating antibody conjugate (ISAC), BDC-1001, as a single agent and in combination with an immune checkpoint inhibitor in patients with advanced HER2-expressing solid tumors. Cancer Res. 2021;81(4_Supplement):OT-03-02-OT-03-02. doi:10.1158/1538-7445.Sabcs20-ot-03-02
  • Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14(2):135–146. doi:10.1038/nrc3670
  • McCarthy PM, Clifton GT, Vreeland TJ, Adams AM, O’Shea AE, Peoples GE. AE37: a HER2-targeted vaccine for the prevention of breast cancer recurrence. Expert Opin Investig Drugs. 2021;30(1):5–11. doi:10.1080/13543784.2021.1849140
  • Benavides LC, Gates JD, Carmichael MG, et al. The impact of HER2/neu expression level on response to the E75 vaccine: from U.S. Military Cancer Institute Clinical Trials Group Study I-01 and I-02. Clin Cancer Res. 2009;15(8):2895–2904. doi:10.1158/1078-0432.Ccr-08-1126
  • Mittendorf EA, Lu B, Melisko M, et al. Efficacy and safety analysis of nelipepimut-S vaccine to prevent breast cancer recurrence: a randomized, multicenter, Phase III clinical trial. Clin Cancer Res. 2019;25(14):4248–4254. doi:10.1158/1078-0432.Ccr-18-2867
  • Clifton GT, Hale D, Vreeland TJ, et al. Results of a randomized Phase IIb trial of nelipepimut-S + trastuzumab versus trastuzumab to prevent recurrences in patients with high-risk HER2 low-expressing breast cancer. Clin Cancer Res. 2020;26(11):2515–2523. doi:10.1158/1078-0432.Ccr-19-2741
  • Chick RC, Clifton GT, Hale DF, et al. Subgroup analysis of nelipepimut-S plus GM-CSF combined with trastuzumab versus trastuzumab alone to prevent recurrences in patients with high-risk, HER2 low-expressing breast cancer. Clin Immunol. 2021;225:108679. doi:10.1016/j.clim.2021.108679
  • Mittendorf EA, Ardavanis A, Symanowski J, et al. Primary analysis of a prospective, randomized, single-blinded phase II trial evaluating the HER2 peptide AE37 vaccine in breast cancer patients to prevent recurrence. Ann Oncol. 2016;27(7):1241–1248. doi:10.1093/annonc/mdw150
  • Witkiewicz AK, Cox D, Knudsen ES. CDK4/6 inhibition provides a potent adjunct to Her2-targeted therapies in preclinical breast cancer models. Genes Cancer. 2014;5(7–8):261–272. doi:10.18632/genesandcancer.24
  • Scirocchi F, Scagnoli S, Botticelli A, et al. Immune effects of CDK4/6 inhibitors in patients with HR+/HER2− metastatic breast cancer: relief from immunosuppression is associated with clinical response. eBioMedicine. 2022;79:104010. doi:10.1016/j.ebiom.2022.104010
  • Goel S, DeCristo MJ, Watt AC, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–475. doi:10.1038/nature23465
  • Gianni L, Colleoni M, Bisagni G, et al. Effects of neoadjuvant trastuzumab, pertuzumab and palbociclib on Ki67 in HER2 and ER-positive breast cancer. NPJ Breast Cancer. 2022;8(1):1. doi:10.1038/s41523-021-00377-8
  • Carlino F, Diana A, Ventriglia A, et al. HER2-low status does not affect survival outcomes of patients with Metastatic Breast Cancer (MBC) undergoing first-line treatment with endocrine therapy plus palbociclib: results of a multicenter, retrospective cohort study. Cancers. 2022;14(20):4981.
  • An R, Li Y-Q, Lin Y-L, Xu F, Li M-M, Liu Z. EZH1/2 as targets for cancer therapy. Cancer Gene Ther. 2022. doi:10.1038/s41417-022-00555-1
  • Lee Shuet S, Li Z, Wu Z, et al. Context-specific regulation of NF-κB target gene expression by EZH2 in breast cancers. Mol Cell. 2011;43(5):798–810. doi:10.1016/j.molcel.2011.08.011
  • Ulrich L, Okines AFC. Treating advanced unresectable or metastatic HER2-positive breast cancer: a spotlight on tucatinib. Breast Cancer. 2021;13:361–381. doi:10.2147/bctt.S268451
  • Collins D, O’Donovan N, Mahgoub T, et al. Trastuzumab (T) and pertuzumab (P)-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) in tyrosine kinase inhibitor (TKI)-treated breast cancer (BC) cell lines.. J Clin Oncol. 2014;32(15_suppl):643. doi:10.1200/jco.2014.32.15_suppl.643
  • Roßwag S, Cotarelo CL, Pantel K, et al. Functional characterization of Circulating Tumor Cells (CTCs) from metastatic ER+/HER2− breast cancer reveals dependence on HER2 and FOXM1 for endocrine therapy resistance and tumor cell survival: implications for treatment of ER+/HER2− breast cancer. Cancers. 2021;13(8):1810. doi:10.3390/cancers13081810
  • Bartlett JMS, Ahmed I, Regan MM, et al. HER2 status predicts for upfront AI benefit: a TRANS-AIOG meta-analysis of 12,129 patients from ATAC, BIG 1–98 and TEAM with centrally determined HER2. Eur J Cancer. 2017;79:129–138. doi:10.1016/j.ejca.2017.03.033
  • Bussolati G, Annaratone L, Maletta F. The pre-analytical phase in surgical pathology. Recent Results Cancer Res. 2015;199:1–13. doi:10.1007/978-3-319-13957-9_1
  • Nitta H, Kelly BD, Allred C, et al. The assessment of HER2 status in breast cancer: the past, the present, and the future. Pathol Int. 2016;66(6):313–324. doi:10.1111/pin.12407
  • Holten-Rossing H, Møller Talman M-L, Kristensson M, Vainer B. Optimizing HER2 assessment in breast cancer: application of automated image analysis. Breast Cancer Res Treat. 2015;152(2):367–375. doi:10.1007/s10549-015-3475-3
  • Otsuji K, Sasaki T, Tanaka A, et al. Use of droplet digital PCR for quantitative and automatic analysis of the HER2 status in breast cancer patients. Breast Cancer Res Treat. 2017;162(1):11–18. doi:10.1007/s10549-016-4092-5
  • Tarantino P, Hamilton E, Tolaney SM, et al. HER2-low breast cancer: pathological and clinical landscape. J Clin Oncol. 2020;38(17):1951–1962. doi:10.1200/jco.19.02488
  • Miglietta F, Griguolo G, Bottosso M, et al. HER2-low-positive breast cancer: evolution from primary tumor to residual disease after neoadjuvant treatment. NPJ Breast Cancer. 2022;8(1):66. doi:10.1038/s41523-022-00434-w
  • Miglietta F, Griguolo G, Bottosso M, et al. Evolution of HER2-low expression from primary to recurrent breast cancer. NPJ Breast Cancer. 2021;7(1):137. doi:10.1038/s41523-021-00343-4