Next Article in Journal
HCM-Associated MuRF1 Variants Compromise Ubiquitylation and Are Predicted to Alter Protein Structure
Previous Article in Journal
Single-Cell RNA Sequencing Outperforms Single-Nucleus RNA Sequencing in Analyzing Pancreatic Cell Diversity and Gene Expression in Goats
Previous Article in Special Issue
Tumor Microenvironment Dynamics of Triple-Negative Breast Cancer Under Radiation Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors

Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(8), 3920; https://doi.org/10.3390/ijms26083920
Submission received: 12 March 2025 / Revised: 13 April 2025 / Accepted: 16 April 2025 / Published: 21 April 2025
(This article belongs to the Special Issue Breast Cancers: From Molecular Basis to Therapy)

Abstract

:
The systemic treatment of breast cancer has evolved remarkably over the past decades. With the introduction of immune checkpoint inhibitors (ICIs), clinical outcomes for solid tumor malignancies have significantly improved. However, in breast cancer, the indication for ICIs is currently limited to triple-negative breast cancer (TNBC) only. In high-risk luminal B hormone receptor-positive (HR+) breast cancer (BC) and HER2-positive (HER2+) BC, modest efficacy of ICI and chemotherapy combinations were identified in the neoadjuvant setting. To address the unmet need, several novel immunotherapy strategies are being tested in ongoing clinical trials as summarized in the current review: bispecific antibodies, chimeric antigen receptor T-cell therapy (CAR-T), T-cell receptors (TCRs), tumor-infiltrating lymphocytes (TILs), tumor vaccines, and oncolytic virus therapy.

Graphical Abstract

1. Introduction

The treatment landscape of breast cancer (BC) has been considerably transformed with the introduction of multiple targeted therapies that resulted in a significant improvement in patient survival [1,2]. In HR+ BC, CDK 4/6 inhibitor, mTOR inhibitor, and PI3K/AKT inhibitor have expanded therapeutic options for HR+ BC and delayed the initiation of chemotherapy [3,4,5,6,7,8]. Human epidermal growth factor receptor 2 (HER2)-targeted therapy is the mainstream for HER2-positive (HER2+) BC. In addition to conventional dual HER2-targeted monoclonal antibodies, HER2 tyrosine kinase inhibitors (TKIs), antibody–drug conjugates (ADCs) such as trastuzumab emtansine (T-DM1) or trastuzumab–deruxtecan (T-DXd) have significantly improved clinical outcomes of HER2+ BC [9,10,11]. Moreover, poly-adenosine diphosphate–ribose polymerase inhibitors (PARPis) have shown promising clinical benefits in both high-risk early-stage BC and advanced BC, in patients with germline BRCA 1/2 mutation [12,13,14]. In addition, ADC targeting trophoblast cell surface antigen 2 (TROP2) or HER2-low and HER2-ultra-low BC also showed improved outcomes in HR+ and TNBC [15,16]. Lastly, emerging data from immune checkpoint inhibitors (ICIs) in TNBC have changed the landscape of treatment for TNBC [17].
Among all breast cancer subtypes, TNBC is considered the most immunogenic, with higher tumor-infiltrating lymphocyte (TIL) and programmed death-ligand 1 (PD-L1) levels [18,19]. In the KEYNOTE 355 trial for metastatic TNBC, the addition of pembrolizumab to chemotherapy demonstrated modest progression-free survival (PFS) and overall survival (OS) benefits in PD-L1-positive mTNBC [17]. In early-stage TNBC, the incorporation of ICIs with neoadjuvant chemotherapy has significantly improved pathological complete response (pCR), a surrogate marker for survival. Pembrolizumab in combination with chemotherapy received FDA approval for TNBC based on the KEYNOTE522 trial, with a pCR of 64.8% vs. 51.2% with chemotherapy alone [20]. The estimated OS at 5 years was 86.6% in the pembrolizumab–chemotherapy group, as compared with 81.7% in the placebo–chemotherapy group (p = 0.002) [21]. A non-anthracycline-containing regimen (NEOPACT) using carboplatin docetaxel and pembrolizumab for six cycles showed a comparative pCR rate of 58% with an encouraging toxicity profile [22]. The ongoing SWOG phase III SCARLET trial is evaluating the NEOPACT regimen in comparison with the KEYNOTE522 regimen (NCT05929768) [20,22]. The utilization of ICIs is currently limited to PD-L1+ TNBC in the metastatic setting, which accounts for 25–40% of the mTNBC population. This underscores substantial unmet needs in the novel immunotherapy strategies for breast cancer.
This review aims to provide a comprehensive overview of immunotherapy modalities beyond ICIs in breast cancer.

2. Immunotherapies in Breast Cancer

2.1. Bispecific T-Cell Engagers and Bispecific Antibodies

Dual-specific protein targeting is a promising approach in developmental immunotherapy. Bispecific T-cell engagers (BiTEs) consist of two parts—one binds to tumor-specific antigen (TSA), while the other binds to the CD3 receptor expressed on T cells, and upon binding, T cells release cytotoxic molecules such as perforin and granzymes. Aside from their direct killing effects, BiTEs can enhance the proliferation of T cells even at low concentrations and modulate the tumor microenvironment—by recruiting antigen-presenting cells (APCs)—to promote favorable outcomes [23]. Several efforts are underway to investigate the role of BiTEs in breast cancer treatment [24]. Surface proteins upregulated in BC such as HER2, epithelial cell adhesion molecule (EpCAM), or TROP2 are common targets [25,26]. Ertumaxomab, a BiTE targeting HER2 and CD3, has exhibited antitumor effects in HER2-low tumors refractory to trastuzumab [27]. In the phase I trial, dose-limiting toxicity (DLT) was not reached and one of fourteen patients achieved a partial response [28,29]. A BiTE targeting P-cadherin and CD3 (PF-06671008) showed preclinical activity in mouse models [30]. However, the phase I trial in humans did not show efficacy, with significant treatment-related adverse events (TRAEs); the most common was cytokine release syndrome (CRS), leading to permanently discontinued treatment in 25% of the patients (NCT 02659631) [25]. Preclinical data on bispecific antibody targeting Trop2 and CEACAM5 demonstrated antitumor efficacy in xenograft models [31,32,33].
Other than BiTEs, there are several bispecific antibodies (BsAbs) which bind at least two epitopes of TSA instead of T-cell markers. Lymphocyte activation gene 3 (LAG3), a protein that acts as an inhibitory receptor, usually in exhausted T cells, is considered a promising target for immunotherapy [34]. Tebotelimab, a BsAbs targeting PD-1 and LAG-3, was evaluated in a phase I trial and showed a 6% response rate in the TNBC cohort (n = 31) (NCT03219268) [35]. In HER2+ BC, the combination of tebotelimab and margetuximab achieved an overall response rate (ORR) of 19% (14/72) [35]. There were 5 confirmed responses among the 28 PD-L1 patients who had disease progression on prior anti-HER2 therapy (18% ORR). B7-H3 is frequently overexpressed in breast cancer and modulates the proliferation and activation of T cells [36]. Over 30 clinical trials targeting B7-H3 through a variety of mechanisms are conducted: bispecific antibodies, ADCs, CAR-T, CAR-NK, and radioimmunotherapy [37,38]. B7-H3 and CD3 BsAbs showed inhibitory effects on the xenograft model [39]. The combination of B7-H3/CD3 BsAbs with ICIs is ongoing (NCT03406949).
Ivonescimab is a BsAb that targets both PD-1 and vascular endothelial growth factor (VEGF). Ouyang et al. reported its excellent antitumor activity and a manageable safety profile (NCT05227664) [40,41]. In a phase I/II study (n = 36), patients with mTNBC received ivonescimab with paclitaxel or nab-paclitaxel as the first line, and a PFS of 9.4 months was achieved. Interestingly, ORR reached 80% regardless of PD-L1 status. In the patients with PD-L1 combined positive score (CPS) < 10% group, ORR was 79%. All patients (n = 36) experienced a treatment-emergent adverse event (TRAE) of any grade; 18 (50.0%) patients experienced a TRAE of grade 3 or higher, and 9 (25.0%) experienced serious TRAEs. The most common TRAEs of grade 3 or higher were neutropenia at 19.4%, and increased liver enzyme at 5.6%. Another BsAb, PM8002/BNT327 targeting PD-L1 and VEGF-A, also demonstrated promising clinical benefit when combined with nab-paclitaxel in a phase Ib/II study (NCT05918133, n = 42) [42]. PFS was 13.3 months, and the ORR was 73.8%. More strikingly, the ORR was 77%, and the median PFS was 18 months in patients with CPS < 1%. Over half of patients experienced grade 3 or 4 TRAEs (54.8%). A phase III randomized trial is planned for verification of these findings. Both agents showed remarkable efficacies in PD-L1-negative mTNBC, which could potentially become practice-changing. Clinical trials with BiTEs in breast cancer are described in Table 1.

2.2. Cancer Vaccines

Cancer vaccines can be classified into several types by antigen-recognizing platforms: peptide-, protein-, carbohydrate antigen-, tumor cell-, DNA-, and dendritic cell (DC)-based vaccines [43]. The most common type of cancer vaccine is peptide-based, which usually consists of 20–30 amino acids derived from tumor-associated antigens (TAAs). It can be taken up by DCs and presented on major histocompatibility complex (MHC) class I molecules to form the peptide–MHC-I complex [44], allowing it to be recognized by cytotoxic T cells and ultimately leading to the elimination of cancer cells. HER2 is considered a potential target for peptide-based vaccines. Despite early success in inducing systemic immune responses and having excellent safety profiles, NeuVaxTM in adjuvant settings failed to show significant benefit in phase III trials [45]. In the phase III trial (NCT01479244) of NeuVaxTM, a total of 758 patients with operable low HER2 BC were enrolled, and there was no significant difference in disease-free survival (DFS) between the control and experimental arms (p = 0.069) [45]. GP2 is a nucleopeptide derived from HER2. A recent phase II trial for patients with residual disease or high-risk pCR after receiving neoadjuvant or postoperative anti-HER2 agents reported that the GP2 vaccine had no significant benefit in DFS in the total population, yet a subset analysis showed a trend toward improved DFS in the HER2+ group that received the GP2 vaccine (p = 0.052) [46]. A phase III trial of GLSI 100 (GP2 vaccine plus granulocyte–macrophage colony-stimulating factor (GM-CSF)) will be launched (NCT05232916) [47].
Human telomerase reverse transcriptase (hTERT) is a subunit of the telomerase complex that facilitates cell proliferation. It is known to be related to unfavorable prognosis in several cancers [48,49]. A vaccine targeting hTERT (VX-001) was tested in human leukocyte antigen (HLA)-A2+ patients with MBC (n = 19). No objective response was observed and 50% of patients had stable disease (SD) [50]. Tissue analysis did reveal increased TILs and tumor necrosis, indicating augmented intratumoral immune responses [50]. Additional trials are ongoing for further investigation of this vaccine in MBC (NNCT00573495 and NCT01660529).
A whole protein-based vaccine which presents TAAs was investigated in HER2+ BC. Compared to peptide-based vaccines, it contains both HLA class I and II epitopes, allowing it to overcome HLA restriction. In a phase I study, a vaccine targeting intracellular domains of HER2 (amino acids 676 to 1255 of the full-length HER-2/neu) demonstrated the immunogenic profile in 29 HER2+ breast and ovarian cancer patients. A total of 89% of patients developed T cell-mediated immunity against HER2 and 82% had an increased level of HER2-specific immunoglobulin G antibodies [51]. A phase I study (NCT00058526) evaluated the efficacy of recombinant HER2 protein (dHER2) combined with the AS15 immunostimulant [52]. In this study, patients with stage II-III HER2+ BC were enrolled. After 5 years of follow-up, 62% of patients were disease-free, and dHER2-specific antibody responses were induced, with the rate of responders increasing with the dHER2 dose and the number and frequency of immunizations. The study was limited by its single-arm nature without vaccine-free control.
Glycans are the most common surface antigens presented by cancer cells, and glycosylation plays a pivotal role in cancer development and progression. Therefore, various efforts targeting glycans as an anticancer vaccine were made over the years, resulting in many clinical trials that have proven unsuccessful, largely due to the immune tolerance associated with them [53]. Novel strategies such as the selection of appropriate domains or antigen modification were used to enhance carbohydrate-based cancer vaccines [54]. Sialyl-Tn (STn) is a carbohydrate antigen that is related to poor prognosis in solid tumors. The vaccine STn-KLH (Theratope®) was the first carbohydrate-based vaccine exhibiting efficacy in a phase II trial [55]. Based on this success, a randomized, double-blinded, phase III trial was conducted and 1028 patients with metastatic BC received cyclophosphamide with this vaccine [56]. Overall, the treatment was well tolerated, and considerable antigen-specific immunoglobulin generation was observed. However, the vaccine failed to improve PFS and OS [57].
The DNA-based vaccine uses DNA fragments that encode TAAs. It is delivered by plasmids or viral vectors and engulfed by APCs, which are subsequently recognized by T cells [58]. The selection of targeting antigens and the formulation of the optimal delivery approaches remain challenges for DNA-based vaccines. One approach targets the transmembrane glycoprotein mucin1 (MUC-1), which is overexpressed on cancer cells and promotes cancer cell immigration and invasion by activating the transforming growth factor beta (TGF-β)-related signaling pathway [59]. PANVAC is a vaccine with carcinoembryonic antigen (CEA) and MUC-1 DNA fragments encapsulated in viral vectors [60]. A phase II trial study tested PANVAC with/without docetaxel in patients with MBC (n = 48). Median PFS (mPFS) was numerically better in the PANVAC arm, but not statistically significant (7.9 vs. 3.9 months, p = 0.09) [61]. Mammaglobin-A (MAM-A) is a TAA overexpressed in 40–80% breast cancers and is known to affect tumor growth by changing chemotherapy sensitivity [62]. A phase I trial was conducted to evaluate the safety and efficacy of the DNA-based vaccine targeting MAM-A [63,64]. HLA-A2 or 3 were initially selected based on preclinical findings (n = 8), and patients without HLA-A2 were also enrolled later on (n = 6), and no statistically significant improvement in PFS was found [65,66,67].
HER-2 DNA vaccine with GM-CSF and interleukin-2 (IL-2) was tested in a Swedish trial, and increased immune activity was observed [68]. A phase I trial testing a DNA vaccine co-targeting HER2 and CEA was conducted in patients with II-IV BC, and no definite cell-mediated immune response was observed [69]. A DNA vaccine targeting HER2 ICD was tested in a phase I dose-escalation trial in patients with stage III and IV HER2+ BC (n = 66). Most patients experienced grade 1 or 2 adverse events, and the patients receiving higher doses of vaccine were correlated with a higher magnitude of HER2 IDC type I immune responses (NCT00436254) [70]. At the time of the report, the median OS and PFS had not been reached in any arm [70]. Another phase I trial utilizing a xenogeneic HER2 DNA-based vaccine for patients with advanced-stage BC is currently ongoing (NCT00393783).
A DC-based cancer vaccine is also under development. DC-based vaccines are produced in ex vivo environments, allowing them to adopt an individual’s immune characteristics by being exposed to various TAAs [71]. This individualized processing is a technical hurdle and makes it challenging to assess the response [72]. In a phase 1 trial including ductal carcinoma in situ (DCIS) and early-stage HER2+ BC, variable immune responses in peripheral blood and tumor specimens were observed (66.7% to 89.5%). Patients with DCIS had a higher pCR than HER2+ BC patients (28.6% vs. 8.3%) [73]. In addition to HER2, p53-derived peptide DC vaccine, with co-simulation by IL-4 and GM-CSF, was tested in patients with metastatic BC (n = 26). Eight patients had stable disease, with a positive correlation between response and p53-specific cellular immune response [74].
Using tumor cells is one of the classic concepts in cancer vaccine development. The intact tumor cells can induce humoral and cellular immunity by themselves, and the antigens derived from the cells can also induce substantial immune reactions [75]. However, this can often lead to off-target immune responses, including autoimmune diseases, because of their allogeneic source [76]. A combination vaccine with autologous DCs and tumor cells was evaluated in a phase I trial [77], in which patients (n = 32) with metastatic breast or renal cancer were enrolled and modest efficacy was observed, with 1 patient achieving near complete response (CR) and 1 patient having partial response (PR) in the breast cancer cohort (n = 16). BriaCell is a human immortalized cell line, engineered to trigger both adaptive and innate responses [78]. In heavily pretreated patients with mBC, a phase 1/2 study (NCT03066947) showed that low-dose cyclophosphamide followed by the SV-BR-1-GM regimen plus retifanlimab, then low-dose local pegylated interferon alpha (IFN)-α, demonstrated improved PFS and OS with acceptable tolerability, and the disease control rate (DCR) reached 50% [79]. A sequential phase 3 trial displayed higher median overall survival (OS) (13.4), an objective response rate (ORR) of 9.5%, and a clinical benefit rate (CBR) of 55% (NCT06072612) [80]. Another phase I/II trial of a V-BR-1-GM regimen in combination with retifanlimab is ongoing in patients with metastatic or locally recurrent breast cancer who have failed standard therapy (NCT03328026) [81]. Completed and ongoing trials are described in Table 2.
Messenger RNA (mRNA) vaccination’s success in combating coronavirus disease (COVID-19) has propelled recent interest in mRNA vaccination in breast cancer [82]. mRNA vaccination has the potential to convert immune-cold breast cancer into immune-hot breast cancer and synergizes with immune checkpoint inhibitors. Effective mRNA vaccine design for in vivo function requires consideration of many factors, including effective vaccine targets, mRNA structures, transport vectors, and injection routes. In a recent phase 1 trial, intratumoral immunotherapy with pembrolizumab and mRNA-2752, a lipid nanoparticle-encapsulated mRNA encoding three immunomodulatory proteins, OX40L, interleukin (IL)-23, and IL-36γ, was tested in patients with ductal carcinoma in situ (DCIS) [83]. The combination was associated with flulike symptoms and grade 1 or 2 local reactions with no serious systemic toxic effects and resulted in an 80% overall response rate. All patients with ERBB2-positive or hormone receptor-negative DCIS experienced at least a partial response, with one-third experiencing complete resolution of the tumor, and high-baseline immune cells were associated with response.

2.3. Adoptive T-Cell Therapy

Adoptive T-cell therapy is one of the immunotherapy strategies that utilizes infusion of the manufactured T cells to eliminate cancer cells [84]. Manufactured T cells can be derived from autologous or allogenic sources and then modified to increase immunogenicity and antigen recognition.

2.3.1. CAR-T Therapy

Chimeric antigen receptor T cells (CAR-T) utilize T cells with antigen recognition domains specific to TAAs and immune-boosting signaling domains. CAR-T has been widely used in routine clinical care for patients with hematologic malignancies, with significant benefits in extending PFS and OS [84]. Its adaptation in solid tumors has been challenging, which is largely attributed to tumor heterogeneity and the hostile tumor microenvironment. There are several target molecules used for CAR-T therapy: HER2, Mesothelin, c-Met, ROR-1, and NKG2D.
HER2 overexpression in breast cancer renders it a highly actionable cancer target. Early HER2 CAR-T development encountered a variable challenge. An early trial reported a patient developed fatal acute respiratory failure after lymphodepleting chemotherapy and infusion of a high dose of HER2 CART designed with the trastuzumab single-chain variable fragment (scFv) and high-dose interleukin (IL)-2 [85,86]. Additional trials administered a lower dose of HER2-specific CAR-T designed from non-trastuzumab scFvs after no or low-intensity lymphodepletion and without cytokine support; although toxicity was not observed in the regimen, antitumor activity was limited [87,88,89]. Other Her2 CAR-T trials such as NCT02713984 and NCT04650451 were terminated due to safety concerns. Currently, a phase 1 trial of CCT 303–406 is anticipated to recruit patients with metastatic HER2-positive solid tumors (NCT04511871). At the same time, HER2 CAR-monocytes/macrophages (HER2 CAR-M) became an appealing option due to the enrichment of macrophages/monocytes in the TME, with potentially better tumor penetration. HER2 CAR-M CT-0508 showed modest antitumor activity and a tolerable safety profile in patients in a phase 1 study (NCT04660929). Among patients who received CT-0508 (n = 14), SD was found in 28.6% of patients [90,91,92].
Mesothelin is overexpressed in most TNBC (~67%); therefore, several clinical trials aimed to evaluate mesothelin-targeted CAR-T products for TNBC. Wang et al. generated PD-1 and T-cell receptor (TCR)-deficient CAR-T products and conducted a phase 1 trial (NCT03545815). In this trial, one mTNBC patient was enrolled, showing 20% disease regression after receiving two doses [93]. Several new early-phase trials are recruiting patients now (NCT05623488, etc.).
C-Met is a member of the MET family, and disrupting the C-Met signaling pathway leads to the promotion of malignant cells. C-Met expressions in breast cancer varied from 14% to 54% and showed higher expression in TNBC than in other subtypes [94]. A few CAR-T trials were conducted for solid tumor expression of C-Met protein. A phase 0 trial in intratumoral injection of mRNA-transfected c-Met-CAR T cells (NCT01837602) aimed to assess safety and feasibility in mTNBC. Among six patients, CAR T messenger ribonucleic acid (mRNA) was detectable in the peripheral blood of two patients and the injected tumor tissues of four patients. The injections were well tolerated, without drug-related adverse effects greater than grade 1 [95]. Shah et al. performed a phase 1 trial of autologous C-Met CAR-T in metastatic melanoma and TNBC patients (NCT03060356) [96]. Among patients who expressed the protein more than 30% by IHC, four patients with TNBC and three patients with melanoma were treated with the CAR-T product. All patients experienced Gr 1-2 AEs, and half of the TNBC cohort achieved SD. Additionally, mRNA response was detected in all patients [97,98,99].
Receptor Tyrosine Kinase-like Orphan Receptor 1 (ROR-1) is involved in embryonal development [100], and it is highly expressed in breast cancer and related to its growth [101]. An anti-ROR-1 CAR-T trial (NCT02706392) showed that two of four TNBC patients reached SD after 15 weeks and 19 weeks of treatment and that none of the four patients experienced severe neurotoxicity or CRS [102]. A phase 1 trial of another ROR-1-targeted CAR-T treatment (LYL797) is currently recruiting solid tumor patients (NCT05274451) [103].
NKG2D (natural-killer group 2, member D) plays a role in the immune activation of NK cells and T-cell subsets by recognizing tumor cells [104]. The first-generation NKG2D-targeted CAR-T therapy showed an antitumor effect in in vivo experiments with the MDA-MB-231 TNBC cell line [105]. Therapeutic Immunotherapy with NKR-2 (THINK) is a multinational open-label phase I study that will evaluate the safety and efficacy of autologous NKR-2 CAR-T cells (NCT03018405). In addition, NCT04107142 will evaluate the access feasibility and safety of haplo/allogeneic NKG2DL-targeting CAR-T cells in solid tumors. In addition to these trials, there are a number of CAR-T trials in BC (Table 3).

2.3.2. TCR Gene Therapy

T-cell receptor gene therapy is another type of adoptive cell therapy. An engineered T-cell receptor is expected to overcome the limitations of CAR-T by increasing the presentation of membrane and intracellular proteins and thus increasing MHC processing [106]. Therefore, TCR gene therapy is more sensitive than CAR-T even in lower-density epitopes. NY-ESO-1 (New York esophageal squamous cell carcinoma 1) is a cancer–testis antigen and is presented in multiple types of solid tumors [107]. Despite its potential for immunotherapy, several TCR-based therapies were conducted in early-phase clinical trials without success. Its effectiveness in humans has been limited due to several potential causes: heterogeneous antigen expression leading to incomplete tumor eradication; limited persistence of TCR-T cells; tumor immune escape mechanism through downregulation of HLA-A molecules; or loss of tumor NY-ESO-1 expression [108,109].
CEA is another antigen that is expressed in less than half of breast cancer cases [110], while elevated serum level is also associated with mBC [111]. Autologous T lymphocytes genetically engineered to express a murine T-cell receptor (TCR) CEA were tested in a phase I trial and the results for the first three patients were reported. All patients experienced profound decreases in serum CEA levels (74–99%), and one patient had an objective regression of cancer metastatic to the lung and liver. However, severe transient inflammatory colitis that represented a dose-limiting toxicity was found in all three patients, which led to the termination of the trial [112]. A phase 1 trial of CEA-targeted therapy (NCT00673829) aimed to address safety and efficacy in mBC patients, but the trial was suspended in 2016 with no result reported.

2.3.3. Tumor-Infiltrating Lymphocyte Therapy

Tumor-infiltrating lymphocyte (TIL) therapy in an intratumoral environment is considered a key factor in antitumor immunity [113]. Due to prior activation, TILs isolated from patients’ tumor tissues demonstrate enhanced recognition of target antigens compared to naive T cells. The first drug for TIL therapy, lifileucel (Amtinagvi), was approved for melanoma, and its indication has expanded to other solid tumors. A pilot phase II trial (NCT 01174121) demonstrated a promising result for BC; six patients received autologous TIL with pembrolizumab, and ORR was 50%, including one case of complete response and two partial responses [114]. The trials for TNBC are currently recruiting more patients (NCT04842812 and NCT04111510).

2.4. Oncolytic Virus

Oncolytic virus is an emerging immunotherapy that induces antitumor immunity indirectly and oncolysis directly [115,116]. Talimogene laherparepvec (TVEC) is based on the herpes simplex virus (HSV) and is genetically engineered to penetrate cancer cells. A phase 1 trial included 14 patients with MBC among a total of 30 patients with solid tumors, where 13 patients received a single dose, and 17 patients received multiple doses without DLT. A total of 19 of 26 patients with post-treatment biopsies had residual diseases, with 14 having tumor necrosis [117,118,119]. Furthermore, a phase II trial with neoadjuvant TVEC therapy in early TNBC was conducted (NCT02779855) [120]. In the study, the RCB0 rate was 45.9% and RCB0-1 was 65% without DLT. Not all results were promising, as seen in a phase II trial (NCT02658812) which explored intratumoral administration of TVEC for inoperable breast cancer; the result seemed disappointing. There was no completion of the planned treatment. Seven of nine terminated the study due to rapid deterioration, and SD was the best response in the remaining two patients [121]. Similarly, when TVEC was combined with atezolizumab in a phase 1 trial (NCT03256344), only 1 patient had PR among 11 patients with TNBC [122]. However, another phase 2 trial of TVEC injection followed by stereotactic body irradiation and pembrolizumab in mTNBC (n = 28) presented a different result [123]. Patients tolerated the treatment well, two patients reached CR, and one patient reached PR with a durable response. Additionally, a phase 1 trial for HER2-negative, metastatic, or unresectable disease (NCT03554044) demonstrated a safety profile and local disease control near the injection site [124].
LTX 315 is an oncolytic peptide which showed a potential demolishing effect when combined with doxorubicin or ICI. According to the preclinical data, LTX 315 and doxorubicin resulted in tumor regression and local necrosis in TNBC mice models [125]. A phase 1 study of LTX 315 with pembrolizumab or ipilimumab included 17 patients with TNBC. The concurrent pembrolizumab group showed a PR rate of 17% among patients with evaluable disease (NCT01986426) [126].
Pelareorep is derived from human reovirus. A phase 2 BRACELET-1 study (NCT04215146) evaluated the efficacy of pelareorep in patients with metastatic HR+/HER2− disease [127]. Patients who received pelareorep plus paclitaxel showed a median PFS of 12.1 m, compared to 6.4 m in the paclitaxel-alone arm, with a hazard ratio of 0.39. In the AWARE-1 trial (NCT04102618) (n = 38), pelareoprep plus atezolizumab was compared to pelareoprop monotherapy [128], and TIL was increased in the atezolizumab arm. Another phase 1 study of pelareoprep with paclitaxel in metastatic HR+/HER2 patients (n = 15) showed an ORR of 20% [129].
Oncolytic virus based on an adenovirus platform has a long history of development. Onyx-015 was the first drug to enter clinical trials and showed limited efficacy in mBC [130]. To overcome the limitations, deletion of E1ACR2 was introduced to increase clinical activity. A phase 1 trial (ICOVIR-7) gathered a total of 21 patients with metastatic solid tumors, 3 of whom had mBC. All patients showed manageable toxicity, and one patient with BC obtained a stabilized tumor marker [131].
Other viruses like pox, polio, or vaccinia have been utilized to develop oncolytic peptides. Early-phase clinical trials are active or recruiting populations with solid tumors including TNBC (NCT03564782, NCT02630368, NCT04301011, and NCT02432963). The novel chimeric orthopoxvirus, CF33-hNIS-anti-PD-L1 (CHECKvacc), encodes two transgenes, human sodium–iodide symporter (hNIS) and anti-PD-L1, with robust anticancer activity in TNBC xenografts. TNBC cells infected with this virus express functional hNIS and anti-PD-L1 proteins. CHECKvacc intratumoral injection is currently undergoing investigation in patients with mTNBC as a single agent (NCT05081492) [132].

2.5. Cytokine Genes

Cytokines induce inflammatory responses among cells and ultimately lead to tumor death [133]. Recombinant interferon-α and interleukin (IL)-2 were approved for the treatment of some malignancies such as melanoma. NKTR-214, also known as bempegaldesleukin (BEMPEG), is an engineered IL-2-specific agonist that stimulates the expansion and proliferation of CD8 T cells and NK cells. NKTR-214 has been tested in a phase 1 trial (NCT02869295) in advanced or metastatic solid tumors (n = 28 total; n = 2 BC) and proved its clinical efficacy, with tolerable toxicity and evidence of increased CD4 and CD8 T cells in the tumor microenvironment (TME) [134].
IL-12 is a proinflammatory cytokine that increases the production of interferon-γ (IFN-γ), and has generated interest in its combination with immunotherapy. In an in vivo study with a BC cell line, tumor shrinkage was observed when recombinant adenovirus encoding IL-12 was administered [135]. A phase 1 trial was initiated to evaluate therapeutic potential as a monotherapy in TNBC, and increased TIL was observed in locoregional TME [136]. Moreover, cytokine therapy has expanded its indication when combined with other immunotherapies. The phase 2 study KEYNOTE 890 (NCT03567720) was aimed at assessing the efficacy of intratumoral tavokinogene telseplasmid, a plasmid encoding IL-12, followed by electroporation and pembrolizumab in patients with mTNBC. Eleven patients completed response evaluation after treatment, three (27.3%) achieved PR, and evidence of enhanced immune response related to IL-12 was detected in blood [137].

2.6. Immune-Modulating Agents That Target the Innate Immune System

SD-101 is an intratumoral toll-like receptor 9 (TLR9) agonist. Its mechanism is to increase the production of IL-2, which leads to cytotoxic activity in TME [138]. The combination of SD101 and the known KEYNOTE regimen was studied in I-spy2 (NCT01042379). Despite showing an enhanced achievement rate of pCR in patients with high-risk, HR-positive/HER2-negative stage II/III breast cancer (n = 75), the regimen did not meet the pre-specified threshold for graduation [139].
Rintatolimod is a chemokine modulator used to treat myalgic myeloencephalitis/chronic fatigue syndrome. It acts on TLR-3 and enhances the innate immune system. A phase 2 trial was completed in an mTNBC cohort in combination with IFN-2a and celecoxib (NCT03599453), and the result from a phase 1 trial for early TNBC announced that toxicity profiles were manageable and the rate of pCR plus ypT1mic was 66% [140].

3. Conclusions

Despite the emergence of immune checkpoint inhibitors, there are several limitations that ICIs cannot fulfill in the treatment of breast cancer. They are closely related to the original pathologic mechanisms of BC and its tumor-favorable microenvironment. Modulating those circumstances is the key to advancing immunotherapy, as several classes of drugs were introduced to manage the limitations. As well as addressing TME, the effective recognition of target antigens is another key to drug development. In addition to HER2, several proteins have proved their potential. Moreover, improving binding power and infiltration of recognized T cells is an important challenge. A newer class of antibodies, vaccines, adoptive T-cell therapy, etc., provides ways to overcome those obstacles. Immune cells like B cells, NK cells, or macrophages are considered targets for immunotherapy, and the regulation of immune-suppressive cells is also part of new immunotherapy. More studies are still warranted, but we foresee these advancements will provide a new paradigm in the immunotherapy of breast cancer.

Author Contributions

Conceptualization, Y.C. and Y.Y.; methodology, Y.C., J.S.L. and Y.Y.; validation of data, J.S.L., J.T. and Y.Y.; formal analysis, Y.C. and J.T.; data curation, Y.C., J.T., D.L., J.S.L. and Y.Y.; writing—original draft, Y.C., J.T. and J.S.L.; review and editing, J.T., D.L., J.S.L. and Y.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This work was supported in part by Cedars-Sinai Cancer Center.

Conflicts of Interest

Y.Y. has contracted research sponsored by Agenus, Merck, and Summit, and is a consultant for Gilead, Novartis, Genentech, and Astra Zeneca. The authors declare no conflicts of interest.

References

  1. Cardoso, F.; O’Shaughnessy, J.; Liu, Z.; McArthur, H.; Schmid, P.; Cortes, J.; Harbeck, N.; Telli, M.L.; Cescon, D.W.; Fasching, P.A.; et al. Pembrolizumab and chemotherapy in high-risk, early-stage, ER(+)/HER2(-) breast cancer: A randomized phase 3 trial. Nat. Med. 2025, 31, 442–448. [Google Scholar] [CrossRef] [PubMed]
  2. Loi, S.; Curigliano, G.; Salgado, R.F.; Romero Diaz, R.I.; Delaloge, S.; Rojas, C.; Kok, M.; Saura Manich, C.; Harbeck, N.; Mittendorf, E.A.; et al. LBA20 A randomized, double-blind trial of nivolumab (NIVO) vs placebo (PBO) with neoadjuvant chemotherapy (NACT) followed by adjuvant endocrine therapy (ET) ± NIVO in patients (pts) with high-risk, ER+ HER2− primary breast cancer (BC). Ann. Oncol. 2023, 34, S1259–S1260. [Google Scholar] [CrossRef]
  3. Turner, N.C.; Slamon, D.J.; Ro, J.; Bondarenko, I.; Im, S.A.; Masuda, N.; Colleoni, M.; DeMichele, A.; Loi, S.; Verma, S.; et al. Overall Survival with Palbociclib and Fulvestrant in Advanced Breast Cancer. N. Engl. J. Med. 2018, 379, 1926–1936. [Google Scholar] [CrossRef]
  4. Hortobagyi, G.N.; Stemmer, S.M.; Burris, H.A.; Yap, Y.S.; Sonke, G.S.; Hart, L.; Campone, M.; Petrakova, K.; Winer, E.P.; Janni, W.; et al. Overall Survival with Ribociclib plus Letrozole in Advanced Breast Cancer. N. Engl. J. Med. 2022, 386, 942–950. [Google Scholar] [CrossRef] [PubMed]
  5. Johnston, S.R.D.; Harbeck, N.; Hegg, R.; Toi, M.; Martin, M.; Shao, Z.M.; Zhang, Q.Y.; Martinez Rodriguez, J.L.; Campone, M.; Hamilton, E.; et al. Abemaciclib Combined With Endocrine Therapy for the Adjuvant Treatment of HR+, HER2-, Node-Positive, High-Risk, Early Breast Cancer (monarchE). J. Clin. Oncol. 2020, 38, 3987–3998. [Google Scholar] [CrossRef]
  6. Rugo, H.S.; Lerebours, F.; Ciruelos, E.; Drullinsky, P.; Ruiz-Borrego, M.; Neven, P.; Park, Y.H.; Prat, A.; Bachelot, T.; Juric, D.; et al. Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): One cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol. 2021, 22, 489–498, Retraction and republication in Lancet Oncol. 2024, 25, e629–e638. https://doi.org/10.1016/S1470-2045(24)00673-9. [Google Scholar] [CrossRef]
  7. Turner, N.C.; Oliveira, M.; Howell, S.J.; Dalenc, F.; Cortes, J.; Gomez Moreno, H.L.; Hu, X.; Jhaveri, K.; Krivorotko, P.; Loibl, S.; et al. Capivasertib in Hormone Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2023, 388, 2058–2070. [Google Scholar] [CrossRef] [PubMed]
  8. Turner, N.C.; Im, S.A.; Saura, C.; Juric, D.; Loibl, S.; Kalinsky, K.; Schmid, P.; Loi, S.; Sunpaweravong, P.; Musolino, A.; et al. Inavolisib-Based Therapy in PIK3CA-Mutated Advanced Breast Cancer. N. Engl. J. Med. 2024, 391, 1584–1596. [Google Scholar] [CrossRef]
  9. Cortes, J.; Kim, S.B.; Chung, W.P.; Im, S.A.; Park, Y.H.; Hegg, R.; Kim, M.H.; Tseng, L.M.; Petry, V.; Chung, C.F.; et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N. Engl. J. Med. 2022, 386, 1143–1154. [Google Scholar] [CrossRef]
  10. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.Y.; Dieras, V.; Guardino, E.; et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef]
  11. von Minckwitz, G.; Huang, C.S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef] [PubMed]
  12. Robson, M.; Im, S.A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N. Engl. J. Med. 2017, 377, 523–533, Corrected in N. Engl. J. Med. 2017, 377, 1700. https://doi.org/10.1056/nejmx170012. [Google Scholar] [CrossRef] [PubMed]
  13. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; de Azambuja, E.; Fielding, A.; Balmana, J.; et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef]
  14. Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Goncalves, A.; Lee, K.H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef]
  15. Rugo, H.S.; Bardia, A.; Marme, F.; Cortes, J.; Schmid, P.; Loirat, D.; Tredan, O.; Ciruelos, E.; Dalenc, F.; Gomez Pardo, P.; et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): A randomised, open-label, multicentre, phase 3 trial. Lancet 2023, 402, 1423–1433. [Google Scholar] [CrossRef]
  16. Curigliano, G.; Hu, X.; Dent, R.A.; Yonemori, K.; Barrios, C.H.; O’Shaughnessy, J.; Wildiers, H.; Zhang, Q.; Im, S.-A.; Saura, C.; et al. Trastuzumab deruxtecan (T-DXd) vs physician’s choice of chemotherapy (TPC) in patients (pts) with hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-low or HER2-ultralow metastatic breast cancer (mBC) with prior endocrine therapy (ET): Primary results from DESTINY-Breast06 (DB-06). J. Clin. Oncol. 2024, 42, LBA1000. [Google Scholar] [CrossRef]
  17. Cortes, J.; Rugo, H.S.; Cescon, D.W.; Im, S.A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Perez-Garcia, J.; Iwata, H.; et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2022, 387, 217–226. [Google Scholar] [CrossRef]
  18. Valenza, C.; Taurelli Salimbeni, B.; Santoro, C.; Trapani, D.; Antonarelli, G.; Curigliano, G. Tumor Infiltrating Lymphocytes across Breast Cancer Subtypes: Current Issues for Biomarker Assessment. Cancers 2023, 15, 767. [Google Scholar] [CrossRef]
  19. Leon-Ferre, R.A.; Jonas, S.F.; Salgado, R.; Loi, S.; de Jong, V.; Carter, J.M.; Nielsen, T.O.; Leung, S.; Riaz, N.; Chia, S.; et al. Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer. JAMA 2024, 331, 1135–1144. [Google Scholar] [CrossRef]
  20. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kummel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef]
  21. Schmid, P.; Cortes, J.; Dent, R.; McArthur, H.; Pusztai, L.; Kummel, S.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Overall Survival with Pembrolizumab in Early-Stage Triple-Negative Breast Cancer. N. Engl. J. Med. 2024, 391, 1981–1991. [Google Scholar] [CrossRef] [PubMed]
  22. Sharma, P.; Stecklein, S.R.; Yoder, R.; Staley, J.M.; Schwensen, K.; O’Dea, A.; Nye, L.; Satelli, D.; Crane, G.; Madan, R.; et al. Clinical and Biomarker Findings of Neoadjuvant Pembrolizumab and Carboplatin Plus Docetaxel in Triple-Negative Breast Cancer: NeoPACT Phase 2 Clinical Trial. JAMA Oncol. 2024, 10, 227–235. [Google Scholar] [CrossRef] [PubMed]
  23. Tian, Z.; Liu, M.; Zhang, Y.; Wang, X. Bispecific T cell engagers: An emerging therapy for management of hematologic malignancies. J. Hematol. Oncol. 2021, 14, 75. [Google Scholar] [CrossRef]
  24. Dees, S.; Ganesan, R.; Singh, S.; Grewal, I.S. Bispecific Antibodies for Triple Negative Breast Cancer. Trends Cancer 2021, 7, 162–173. [Google Scholar] [CrossRef] [PubMed]
  25. Harding, J.J.; Garrido-Laguna, I.; Chen, X.; Basu, C.; Dowlati, A.; Forgie, A.; Hooper, A.T.; Kamperschroer, C.; Max, S.I.; Moreau, A.; et al. A Phase 1 Dose-Escalation Study of PF-06671008, a Bispecific T-Cell-Engaging Therapy Targeting P-Cadherin in Patients With Advanced Solid Tumors. Front. Immunol. 2022, 13, 845417. [Google Scholar] [CrossRef]
  26. Hong, R.; Zhou, Y.; Tian, X.; Wang, L.; Wu, X. Selective inhibition of IDO1, D-1-methyl-tryptophan (D-1MT), effectively increased EpCAM/CD3-bispecific BiTE antibody MT110 efficacy against IDO1(hi)breast cancer via enhancing immune cells activity. Int. Immunopharmacol. 2018, 54, 118–124. [Google Scholar] [CrossRef] [PubMed]
  27. Kiewe, P.; Thiel, E. Ertumaxomab: A trifunctional antibody for breast cancer treatment. Expert. Opin. Investig. Drugs 2008, 17, 1553–1558. [Google Scholar] [CrossRef]
  28. Haense, N.; Pauligk, C.; Marme, F.; Werner, D.; Sorgius, D.; Hozaeel, W.; Rafiyan, M.-R.; Ottmann, O.G.; Klunker, D.; Jäger, M.; et al. Interim analysis of a phase I/II open label, dose-escalating study to investigate safety, tolerability, and preliminary efficacy of the trifunctional anti-HER2/neu x anti-CD3 antibody ertumaxomab in patients with HER2/neu expressing solid tumors progressing after standard therapy. J. Clin. Oncol. 2014, 32 (Suppl. 15), 3055. [Google Scholar] [CrossRef]
  29. Haense, N.; Atmaca, A.; Pauligk, C.; Steinmetz, K.; Marme, F.; Haag, G.M.; Rieger, M.; Ottmann, O.G.; Ruf, P.; Lindhofer, H.; et al. A phase I trial of the trifunctional anti Her2 x anti CD3 antibody ertumaxomab in patients with advanced solid tumors. BMC Cancer 2016, 16, 420. [Google Scholar] [CrossRef]
  30. Fisher, T.S.; Hooper, A.T.; Lucas, J.; Clark, T.H.; Rohner, A.K.; Peano, B.; Elliott, M.W.; Tsaparikos, K.; Wang, H.; Golas, J.; et al. A CD3-bispecific molecule targeting P-cadherin demonstrates T cell-mediated regression of established solid tumors in mice. Cancer Immunol. Immunother. 2018, 67, 247–259. [Google Scholar] [CrossRef]
  31. Chang, C.H.; Wang, Y.; Li, R.; Rossi, D.L.; Liu, D.; Rossi, E.A.; Cardillo, T.M.; Goldenberg, D.M. Combination Therapy with Bispecific Antibodies and PD-1 Blockade Enhances the Antitumor Potency of T Cells. Cancer Res. 2017, 77, 5384–5394. [Google Scholar] [CrossRef] [PubMed]
  32. Sharkey, R.M.; van Rij, C.M.; Karacay, H.; Rossi, E.A.; Frielink, C.; Regino, C.; Cardillo, T.M.; McBride, W.J.; Chang, C.H.; Boerman, O.C.; et al. A new Tri-Fab bispecific antibody for pretargeting Trop-2-expressing epithelial cancers. J. Nucl. Med. 2012, 53, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  33. Seckinger, A.; Majocchi, S.; Moine, V.; Nouveau, L.; Ngoc, H.; Daubeuf, B.; Ravn, U.; Pleche, N.; Calloud, S.; Broyer, L.; et al. Development and characterization of NILK-2301, a novel CEACAM5xCD3 kappalambda bispecific antibody for immunotherapy of CEACAM5-expressing cancers. J. Hematol. Oncol. 2023, 16, 117. [Google Scholar] [CrossRef]
  34. Aggarwal, V.; Workman, C.J.; Vignali, D.A.A. LAG-3 as the third checkpoint inhibitor. Nat. Immunol. 2023, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  35. Luke, J.J.; Patel, M.R.; Blumenschein, G.R.; Hamilton, E.; Chmielowski, B.; Ulahannan, S.V.; Connolly, R.M.; Santa-Maria, C.A.; Wang, J.; Bahadur, S.W.; et al. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: A phase 1 trial. Nat. Med. 2023, 29, 2814–2824. [Google Scholar] [CrossRef]
  36. Mei, J.; Cai, Y.; Zhu, H.; Jiang, Y.; Fu, Z.; Xu, J.; Chen, L.; Yang, K.; Zhao, J.; Song, C.; et al. High B7-H3 expression with low PD-L1 expression identifies armored-cold tumors in triple-negative breast cancer. NPJ Breast Cancer 2024, 10, 11. [Google Scholar] [CrossRef]
  37. Getu, A.A.; Tigabu, A.; Zhou, M.; Lu, J.; Fodstad, O.; Tan, M. New frontiers in immune checkpoint B7-H3 (CD276) research and drug development. Mol. Cancer 2023, 22, 43. [Google Scholar] [CrossRef] [PubMed]
  38. Kontos, F.; Michelakos, T.; Kurokawa, T.; Sadagopan, A.; Schwab, J.H.; Ferrone, C.R.; Ferrone, S. B7-H3: An Attractive Target for Antibody-based Immunotherapy. Clin. Cancer Res. 2021, 27, 1227–1235. [Google Scholar] [CrossRef]
  39. Zhang, Z.; Jiang, C.; Liu, Z.; Yang, M.; Tang, X.; Wang, Y.; Zheng, M.; Huang, J.; Zhong, K.; Zhao, S.; et al. B7-H3-Targeted CAR-T Cells Exhibit Potent Antitumor Effects on Hematologic and Solid Tumors. Mol. Ther. Oncolytics 2020, 17, 180–189. [Google Scholar] [CrossRef]
  40. Ouyang, Q.; Wang, X.; Tian, C.; Shao, X.; Huang, J.; Chen, Z.H.; Wang, Y.; Sun, T.; Yi, T.; Yu, X.; et al. 347MO The safety and efficacy of ivonescimab in combination with chemotherapy as first-line (1L) treatment for triple-negative breast cancer (TNBC). Ann. Oncol. 2024, 35, S360–S361. [Google Scholar] [CrossRef]
  41. Ouyang, Q.; Wang, X.; Tian, C.; Shao, X.; Huang, J.; Chen, Z.; Wang, Y.; Sun, T.; Yi, T.; Yu, X.; et al. Evaluation of the safety and efficacy of ivonescimab in combination with chemotherapy as first-line treatment for triple-negative breast cancer. Brain 2024, 3, 61. [Google Scholar]
  42. Wu, J.; Zhang, J.; Tong, Z.; Zhang, Q.; Wang, Y.; Cheng, Q.; Chen, X.; Li, Z.; Yin, Y.; Du, Y.; et al. 348MO A phase Ib/II study to assess the safety and efficacy of PM8002/BNT327 in combination with nab-paclitaxel for first-line treatment of locally advanced or metastatic triple-negative breast cancer. Ann. Oncol. 2024, 35, S361. [Google Scholar] [CrossRef]
  43. Al-Hawary, S.I.S.; Saleh, E.A.M.; Mamajanov, N.A.; Gilmanova, N.S.; Alsaab, H.O.; Alghamdi, A.; Ansari, S.A.; Alawady, A.H.R.; Alsaalamy, A.H.; Ibrahim, A.J. Breast cancer vaccines; A comprehensive and updated review. Pathol. Res. Pract. 2023, 249, 154735. [Google Scholar] [CrossRef]
  44. Purcell, A.W.; McCluskey, J.; Rossjohn, J. More than one reason to rethink the use of peptides in vaccine design. Nat. Rev. Drug Discov. 2007, 6, 404–414. [Google Scholar] [CrossRef] [PubMed]
  45. Mittendorf, E.A.; Lu, B.; Melisko, M.; Price Hiller, J.; Bondarenko, I.; Brunt, A.M.; Sergii, G.; Petrakova, K.; Peoples, G.E. 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, 4248–4254. [Google Scholar] [CrossRef]
  46. Brown, T.A., 2nd; Mittendorf, E.A.; Hale, D.F.; Myers, J.W., 3rd; Peace, K.M.; Jackson, D.O.; Greene, J.M.; Vreeland, T.J.; Clifton, G.T.; Ardavanis, A.; et al. Prospective, randomized, single-blinded, multi-center phase II trial of two HER2 peptide vaccines, GP2 and AE37, in breast cancer patients to prevent recurrence. Breast Cancer Res. Treat. 2020, 181, 391–401. [Google Scholar] [CrossRef]
  47. Patel, S.; Thompson, J.; Patel, M.; Daugherty, F.J.; Rimawi, M.F. Phase III study to evaluate the efficacy and safety of GLSI-100 (GP2 + GM-CSF) in breast cancer patients with residual disease or high-risk PCR after both neoadjuvant and postoperative adjuvant anti-HER2 therapy: Flamingo-01. J. Clin. Oncol. 2023, 41 (Suppl. 16), TPS617. [Google Scholar] [CrossRef]
  48. Wang, K.; Wang, R.L.; Liu, J.J.; Zhou, J.; Li, X.; Hu, W.W.; Jiang, W.J.; Hao, N.B. The prognostic significance of hTERT overexpression in cancers: A systematic review and meta-analysis. Medicine 2018, 97, e11794. [Google Scholar] [CrossRef]
  49. Zhu, C.Q.; Cutz, J.C.; Liu, N.; Lau, D.; Shepherd, F.A.; Squire, J.A.; Tsao, M.S. Amplification of telomerase (hTERT) gene is a poor prognostic marker in non-small-cell lung cancer. Br. J. Cancer 2006, 94, 1452–1459. [Google Scholar] [CrossRef]
  50. Domchek, S.M.; Recio, A.; Mick, R.; Clark, C.E.; Carpenter, E.L.; Fox, K.R.; DeMichele, A.; Schuchter, L.M.; Leibowitz, M.S.; Wexler, M.H.; et al. Telomerase-specific T-cell immunity in breast cancer: Effect of vaccination on tumor immunosurveillance. Cancer Res. 2007, 67, 10546–10555. [Google Scholar] [CrossRef]
  51. Disis, M.L.; Schiffman, K.; Guthrie, K.; Salazar, L.G.; Knutson, K.L.; Goodell, V.; dela Rosa, C.; Cheever, M.A. Effect of dose on immune response in patients vaccinated with an her-2/neu intracellular domain protein--based vaccine. J. Clin. Oncol. 2004, 22, 1916–1925. [Google Scholar] [CrossRef]
  52. Limentani, S.A.; Campone, M.; Dorval, T.; Curigliano, G.; de Boer, R.; Vogel, C.; White, S.; Bachelot, T.; Canon, J.L.; Disis, M.; et al. A non-randomized dose-escalation Phase I trial of a protein-based immunotherapeutic for the treatment of breast cancer patients with HER2-overexpressing tumors. Breast Cancer Res. Treat. 2016, 156, 319–330. [Google Scholar] [CrossRef] [PubMed]
  53. Livingston, P.O.; Zhang, S.; Lloyd, K.O. Carbohydrate vaccines that induce antibodies against cancer. 1. Rationale. Cancer Immunol. Immunother. 1997, 45, 1–9. [Google Scholar] [CrossRef] [PubMed]
  54. Sorieul, C.; Papi, F.; Carboni, F.; Pecetta, S.; Phogat, S.; Adamo, R. Recent advances and future perspectives on carbohydrate-based cancer vaccines and therapeutics. Pharmacol. Ther. 2022, 235, 108158. [Google Scholar] [CrossRef] [PubMed]
  55. Miles, D.; Papazisis, K. Rationale for the clinical development of STn-KLH (Theratope) and anti-MUC-1 vaccines in breast cancer. Clin. Breast Cancer 2003, 3 (Suppl. 4), S134–S138. [Google Scholar] [CrossRef]
  56. Miles, D.; Roche, H.; Martin, M.; Perren, T.J.; Cameron, D.A.; Glaspy, J.; Dodwell, D.; Parker, J.; Mayordomo, J.; Tres, A.; et al. Phase III multicenter clinical trial of the sialyl-TN (STn)-keyhole limpet hemocyanin (KLH) vaccine for metastatic breast cancer. Oncologist 2011, 16, 1092–1100. [Google Scholar] [CrossRef]
  57. Ibrahim, N.K.; Murray, J.L.; Zhou, D.; Mittendorf, E.A.; Sample, D.; Tautchin, M.; Miles, D. Survival Advantage in Patients with Metastatic Breast Cancer Receiving Endocrine Therapy plus Sialyl Tn-KLH Vaccine: Post Hoc Analysis of a Large Randomized Trial. J. Cancer 2013, 4, 577–584. [Google Scholar] [CrossRef]
  58. Pandya, A.; Shah, Y.; Kothari, N.; Postwala, H.; Shah, A.; Parekh, P.; Chorawala, M.R. The future of cancer immunotherapy: DNA vaccines leading the way. Med. Oncol. 2023, 40, 200. [Google Scholar] [CrossRef]
  59. Chen, W.; Zhang, Z.; Zhang, S.; Zhu, P.; Ko, J.K.; Yung, K.K. MUC1: Structure, Function, and Clinic Application in Epithelial Cancers. Int. J. Mol. Sci. 2021, 22, 6567. [Google Scholar] [CrossRef]
  60. Madan, R.A.; Arlen, P.M.; Gulley, J.L. PANVAC-VF: Poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert. Opin. Biol. Ther. 2007, 7, 543–554. [Google Scholar] [CrossRef]
  61. Heery, C.R.; Ibrahim, N.K.; Arlen, P.M.; Mohebtash, M.; Murray, J.L.; Koenig, K.; Madan, R.A.; McMahon, S.; Marte, J.L.; Steinberg, S.M.; et al. Docetaxel Alone or in Combination With a Therapeutic Cancer Vaccine (PANVAC) in Patients With Metastatic Breast Cancer: A Randomized Clinical Trial. JAMA Oncol. 2015, 1, 1087–1095. [Google Scholar] [CrossRef]
  62. Picot, N.; Guerrette, R.; Beauregard, A.P.; Jean, S.; Michaud, P.; Harquail, J.; Benzina, S.; Robichaud, G.A. Mammaglobin 1 promotes breast cancer malignancy and confers sensitivity to anticancer drugs. Mol. Carcinog. 2016, 55, 1150–1162. [Google Scholar] [CrossRef]
  63. Tiriveedhi, V.; Tucker, N.; Herndon, J.; Li, L.; Sturmoski, M.; Ellis, M.; Ma, C.; Naughton, M.; Lockhart, A.C.; Gao, F.; et al. Safety and preliminary evidence of biologic efficacy of a mammaglobin-a DNA vaccine in patients with stable metastatic breast cancer. Clin. Cancer Res. 2014, 20, 5964–5975. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, S.W.; Goedegebuure, P.; Gillanders, W.E. Mammaglobin-A is a target for breast cancer vaccination. Oncoimmunology 2016, 5, e1069940. [Google Scholar] [CrossRef] [PubMed]
  65. Bharat, A.; Benshoff, N.; Fleming, T.P.; Dietz, J.R.; Gillanders, W.E.; Mohanakumar, T. Characterization of the role of CD8+T cells in breast cancer immunity following mammaglobin-A DNA vaccination using HLA-class-I tetramers. Breast Cancer Res. Treat. 2008, 110, 453–463. [Google Scholar] [CrossRef] [PubMed]
  66. Jaramillo, A.; Majumder, K.; Manna, P.P.; Fleming, T.P.; Doherty, G.; Dipersio, J.F.; Mohanakumar, T. Identification of HLA-A3-restricted CD8+ T cell epitopes derived from mammaglobin-A, a tumor-associated antigen of human breast cancer. Int. J. Cancer 2002, 102, 499–506. [Google Scholar] [CrossRef]
  67. Jaramillo, A.; Narayanan, K.; Campbell, L.G.; Benshoff, N.D.; Lybarger, L.; Hansen, T.H.; Fleming, T.P.; Dietz, J.R.; Mohanakumar, T. Recognition of HLA-A2-restricted mammaglobin-A-derived epitopes by CD8+ cytotoxic T lymphocytes from breast cancer patients. Breast Cancer Res. Treat. 2004, 88, 29–41. [Google Scholar] [CrossRef]
  68. Norell, H.; Poschke, I.; Charo, J.; Wei, W.Z.; Erskine, C.; Piechocki, M.P.; Knutson, K.L.; Bergh, J.; Lidbrink, E.; Kiessling, R. Vaccination with a plasmid DNA encoding HER-2/neu together with low doses of GM-CSF and IL-2 in patients with metastatic breast carcinoma: A pilot clinical trial. J. Transl. Med. 2010, 8, 53. [Google Scholar] [CrossRef]
  69. Diaz, C.M.; Chiappori, A.; Aurisicchio, L.; Bagchi, A.; Clark, J.; Dubey, S.; Fridman, A.; Fabregas, J.C.; Marshall, J.; Scarselli, E.; et al. Phase 1 studies of the safety and immunogenicity of electroporated HER2/CEA DNA vaccine followed by adenoviral boost immunization in patients with solid tumors. J. Transl. Med. 2013, 11, 62. [Google Scholar] [CrossRef]
  70. Disis, M.L.N.; Guthrie, K.A.; Liu, Y.; Coveler, A.L.; Higgins, D.M.; Childs, J.S.; Dang, Y.; Salazar, L.G. Safety and Outcomes of a Plasmid DNA Vaccine Encoding the ERBB2 Intracellular Domain in Patients With Advanced-Stage ERBB2-Positive Breast Cancer: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol. 2023, 9, 71–78. [Google Scholar] [CrossRef]
  71. Perez, C.R.; De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat. Commun. 2019, 10, 5408. [Google Scholar] [CrossRef] [PubMed]
  72. Shumway, N.M.; Ibrahim, N.; Ponniah, S.; Peoples, G.E.; Murray, J.L. Therapeutic breast cancer vaccines: A new strategy for early-stage disease. BioDrugs 2009, 23, 277–287. [Google Scholar] [CrossRef]
  73. Lowenfeld, L.; Mick, R.; Datta, J.; Xu, S.; Fitzpatrick, E.; Fisher, C.S.; Fox, K.R.; DeMichele, A.; Zhang, P.J.; Weinstein, S.P.; et al. Dendritic Cell Vaccination Enhances Immune Responses and Induces Regression of HER2(pos) DCIS Independent of Route: Results of Randomized Selection Design Trial. Clin. Cancer Res. 2017, 23, 2961–2971. [Google Scholar] [CrossRef] [PubMed]
  74. Svane, I.M.; Pedersen, A.E.; Johansen, J.S.; Johnsen, H.E.; Nielsen, D.; Kamby, C.; Ottesen, S.; Balslev, E.; Gaarsdal, E.; Nikolajsen, K.; et al. Vaccination with p53 peptide-pulsed dendritic cells is associated with disease stabilization in patients with p53 expressing advanced breast cancer; monitoring of serum YKL-40 and IL-6 as response biomarkers. Cancer Immunol. Immunother. 2007, 56, 1485–1499. [Google Scholar] [CrossRef]
  75. Perez-Banos, A.; Gleisner, M.A.; Flores, I.; Pereda, C.; Navarrete, M.; Araya, J.P.; Navarro, G.; Quezada-Monras, C.; Tittarelli, A.; Salazar-Onfray, F. Whole tumour cell-based vaccines: Tuning the instruments to orchestrate an optimal antitumour immune response. Br. J. Cancer 2023, 129, 572–585. [Google Scholar] [CrossRef]
  76. Tay, B.Q.; Wright, Q.; Ladwa, R.; Perry, C.; Leggatt, G.; Simpson, F.; Wells, J.W.; Panizza, B.J.; Frazer, I.H.; Cruz, J.L.G. Evolution of Cancer Vaccines-Challenges, Achievements, and Future Directions. Vaccines 2021, 9, 535. [Google Scholar] [CrossRef] [PubMed]
  77. Avigan, D.; Vasir, B.; Gong, J.; Borges, V.; Wu, Z.; Uhl, L.; Atkins, M.; Mier, J.; McDermott, D.; Smith, T.; et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses. Clin. Cancer Res. 2004, 10, 4699–4708. [Google Scholar] [CrossRef]
  78. Hurvitz, S.A.; Chumsri, S.; Brufsky, A.; Nangia, C.S.; Del Priore, G.; Cristofanilli, M. Study of the Bria-IMT regimen and CPI vs physicians’ choice in advanced metastatic breast cancer (BRIA-ABC). J. Clin. Oncol. 2024, 42 (Suppl. S16), TPS1137. [Google Scholar] [CrossRef]
  79. Wiseman, C.L.; Holmes, J.P.; Calfa, C.; Dakhil, S.R.; Bhattacharya, S.; Peoples, G.E.; Lacher, M.D.; Lopez-Lago, M.; Kharazi, A.; Del Priore, G.; et al. Results of a phase I/IIa trial of SV-BR-1-GM inoculation with low-dose cyclophosphamide and interferon alpha (Bria-IMT) in metastatic breast cancer. Hum. Vaccin. Immunother. 2024, 20, 2379864. [Google Scholar] [CrossRef]
  80. Chumsri, S. Overall survival results of BRIA-IMT allogenic whole cell-based cancer vaccine. In Proceedings of the 2024 San Antonio Breast Cancer Symposium, San Antonio, TX, USA, 10–14 December 2024. [Google Scholar]
  81. Calfa, C.J.; Nangia, C.S.; Barve, M.A.; Rowland, K.M.; Boccia, R.V.; Knecht, J.G.; Chang, M.; Salgado, M.; Bayer, B.; Aghajanian, T.; et al. Outcomes of advanced/metastatic breast cancer (aMBC) treated with BRIA-IMT, an allogeneic whole cell immunotherapy. J. Clin. Oncol. 2024, 42 (Suppl. S16), 1022. [Google Scholar] [CrossRef]
  82. Jiang, X.T.; Liu, Q. mRNA vaccination in breast cancer: Current progress and future direction. J. Cancer Res. Clin. Oncol. 2023, 149, 9435–9450. [Google Scholar] [CrossRef] [PubMed]
  83. Ramalingam, K.; Woody, R.; Glencer, A.; Schwartz, C.J.; Mori, H.; Wong, J.; Hirst, G.; Rosenbluth, J.; Onishi, N.; Gibbs, J.; et al. Intratumoral Injection of mRNA-2752 and Pembrolizumab for High-Risk Ductal Carcinoma In Situ: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol. 2025, 11, 288–292. [Google Scholar] [CrossRef]
  84. June, C.H. Adoptive T cell therapy for cancer in the clinic. J. Clin. Invest. 2007, 117, 1466–1476. [Google Scholar] [CrossRef]
  85. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef]
  86. Shabaneh, T.B.; Stevens, A.R.; Stull, S.M.; Shimp, K.R.; Seaton, B.W.; Gad, E.A.; Jaeger-Ruckstuhl, C.A.; Simon, S.; Koehne, A.L.; Price, J.P.; et al. Systemically administered low-affinity HER2 CAR T cells mediate antitumor efficacy without toxicity. J. Immunother. Cancer 2024, 12, e008566. [Google Scholar] [CrossRef]
  87. Akhoundi, M.; Mohammadi, M.; Sahraei, S.S.; Sheykhhasan, M.; Fayazi, N. CAR T cell therapy as a promising approach in cancer immunotherapy: Challenges and opportunities. Cell. Oncol. 2021, 44, 495–523. [Google Scholar] [CrossRef]
  88. Feng, K.; Liu, Y.; Guo, Y.; Qiu, J.; Wu, Z.; Dai, H.; Yang, Q.; Wang, Y.; Han, W. Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell 2018, 9, 838–847. [Google Scholar] [CrossRef] [PubMed]
  89. Hegde, M.; Joseph, S.K.; Pashankar, F.; DeRenzo, C.; Sanber, K.; Navai, S.; Byrd, T.T.; Hicks, J.; Xu, M.L.; Gerken, C.; et al. Tumor response and endogenous immune reactivity after administration of HER2 CAR T cells in a child with metastatic rhabdomyosarcoma. Nat. Commun. 2020, 11, 3549. [Google Scholar] [CrossRef] [PubMed]
  90. Abdou, Y.; Dees, E.C.; Mortimer, J.E.; Pohlmann, P.R.; Johnson, M.L.; Maziarz, R.T.; Specht, J.M.; Yuan, Y.; Ueno, N.T.; Angelos, M.; et al. A phase 1, first-in-human (FIH) study of autologous macrophages engineered to express an anti-HER2 chimeric antigen receptor (CAR) in participants (pts) with HER2-overexpressing solid tumors. J. Clin. Oncol. 2023, 41 (Suppl. S16), TPS2666. [Google Scholar] [CrossRef]
  91. Klichinsky, M. Harnessing the power of engineered macrophages. In Proceedings of the 8th Annual CAR-TCR Summit, Boston, MA, USA, 30 August–1 September 2023. [Google Scholar]
  92. Reiss, K.A.; Angelos, M.G.; Dees, E.C.; Yuan, Y.; Ueno, N.T.; Pohlmann, P.R.; Johnson, M.L.; Chao, J.; Shestova, O.; Serody, J.S.; et al. CAR-macrophage therapy for HER2-overexpressing advanced solid tumors: A phase 1 trial. Nat. Med. 2025, 31, 1171–1182. [Google Scholar] [CrossRef]
  93. Wang, Z.; Li, N.; Feng, K.; Chen, M.; Zhang, Y.; Liu, Y.; Yang, Q.; Nie, J.; Tang, N.; Zhang, X.; et al. Phase I study of CAR-T cells with PD-1 and TCR disruption in mesothelin-positive solid tumors. Cell. Mol. Immunol. 2021, 18, 2188–2198. [Google Scholar] [CrossRef] [PubMed]
  94. Yan, S.; Jiao, X.; Zou, H.; Li, K. Prognostic significance of c-Met in breast cancer: A meta-analysis of 6010 cases. Diagn. Pathol. 2015, 10, 62. [Google Scholar] [CrossRef]
  95. Tchou, J.; Zhao, Y.; Levine, B.L.; Zhang, P.J.; Davis, M.M.; Melenhorst, J.J.; Kulikovskaya, I.; Brennan, A.L.; Liu, X.; Lacey, S.F.; et al. Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T Cells in Metastatic Breast Cancer. Cancer Immunol. Res. 2017, 5, 1152–1161. [Google Scholar] [CrossRef]
  96. Shah, P.D.; Huang, A.C.; Xu, X.; Orlowski, R.; Amaravadi, R.K.; Schuchter, L.M.; Zhang, P.; Tchou, J.; Matlawski, T.; Cervini, A.; et al. Phase I Trial of Autologous RNA-electroporated cMET-directed CAR T Cells Administered Intravenously in Patients with Melanoma and Breast Carcinoma. Cancer Res. Commun. 2023, 3, 821–829. [Google Scholar] [CrossRef]
  97. Bamdad, C.C.; Yuan, Y.; Specht, J.M.; Stewart, A.K.; Smagghe, B.J.; Lin, S.C.-M.; Carter, M.G.; Synold, T.W.; Frankel, P.H.; Parekh, V.; et al. Phase I/II first-in-human CAR T–targeting MUC1 transmembrane cleavage product (MUC1*) in patients with metastatic breast cancer. J. Clin. Oncol. 2002, 40 (Suppl. S16), TPS1130. [Google Scholar] [CrossRef]
  98. Posey, A.D., Jr.; Schwab, R.D.; Boesteanu, A.C.; Steentoft, C.; Mandel, U.; Engels, B.; Stone, J.D.; Madsen, T.D.; Schreiber, K.; Haines, K.M.; et al. Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. Immunity 2016, 44, 1444–1454. [Google Scholar] [CrossRef] [PubMed]
  99. Zhou, R.; Yazdanifar, M.; Roy, L.D.; Whilding, L.M.; Gavrill, A.; Maher, J.; Mukherjee, P. Corrigendum: CAR T Cells Targeting the Tumor MUC1 Glycoprotein Reduce Triple-Negative Breast Cancer Growth. Front. Immunol. 2020, 11, 628776. [Google Scholar] [CrossRef]
  100. Balakrishnan, A.; Goodpaster, T.; Randolph-Habecker, J.; Hoffstrom, B.G.; Jalikis, F.G.; Koch, L.K.; Berger, C.; Kosasih, P.L.; Rajan, A.; Sommermeyer, D.; et al. Analysis of ROR1 Protein Expression in Human Cancer and Normal Tissues. Clin. Cancer Res. 2017, 23, 3061–3071. [Google Scholar] [CrossRef]
  101. Zhang, S.; Chen, L.; Cui, B.; Chuang, H.Y.; Yu, J.; Wang-Rodriguez, J.; Tang, L.; Chen, G.; Basak, G.W.; Kipps, T.J. ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth. PLoS ONE 2012, 7, e31127. [Google Scholar] [CrossRef]
  102. Specht, J.M.; Lee, S.M.; Turtle, C.; Berger, C.; Balakrishnan, A.; Srivastava, S.; Viollet, V.; Veatch, J.; Gooley, T.; Mullane, E.; et al. Abstract P2-09-13: A phase I study of adoptive immunotherapy for ROR1+ advanced triple negative breast cancer (TNBC) with defined subsets of autologous T cells expressing a ROR1-specific chimeric antigen receptor (ROR1-CAR). Cancer Res. 2019, 79 (Suppl. S4), P2-09-13. [Google Scholar] [CrossRef]
  103. Park, S.; Simianer, C.; Spadinger, S.; Wang, X.; Sundar, P.; Potluri, S.; Lynn, R.; Boldajipour, B.; Wang, G.; Sharma, N.; et al. Abstract 2754: LYL797, a ROR1 CAR T-cell therapy with genetic and epigenetic reprogramming for solid tumors. Cancer Res. 2022, 82 (Suppl. 12), 2754. [Google Scholar] [CrossRef]
  104. Curio, S.; Jonsson, G.; Marinovic, S. A summary of current NKG2D-based CAR clinical trials. Immunother. Adv. 2021, 1, ltab018. [Google Scholar] [CrossRef]
  105. Han, Y.; Xie, W.; Song, D.G.; Powell, D.J., Jr. Control of triple-negative breast cancer using ex vivo self-enriched, costimulated NKG2D CAR T cells. J. Hematol. Oncol. 2018, 11, 92. [Google Scholar] [CrossRef] [PubMed]
  106. Baulu, E.; Gardet, C.; Chuvin, N.; Depil, S. TCR-engineered T cell therapy in solid tumors: State of the art and perspectives. Sci. Adv. 2023, 9, eadf3700. [Google Scholar] [CrossRef] [PubMed]
  107. Thomas, R.; Al-Khadairi, G.; Roelands, J.; Hendrickx, W.; Dermime, S.; Bedognetti, D.; Decock, J. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front. Immunol. 2018, 9, 947. [Google Scholar] [CrossRef]
  108. Gyurdieva, A.; Zajic, S.; Chang, Y.F.; Houseman, E.A.; Zhong, S.; Kim, J.; Nathenson, M.; Faitg, T.; Woessner, M.; Turner, D.C.; et al. Biomarker correlates with response to NY-ESO-1 TCR T cells in patients with synovial sarcoma. Nat. Commun. 2022, 13, 5296. [Google Scholar] [CrossRef]
  109. Frankiw, L.; Singh, A.; Peters, C.; Comin-Anduix, B.; Berent-Maoz, B.; Macabali, M.; Shammaie, K.; Quiros, C.; Kaplan-Lefko, P.; Baselga Carretero, I.; et al. Immunotherapy resistance driven by loss of NY-ESO-1 expression in response to transgenic adoptive cellular therapy with PD-1 blockade. J. Immunother. Cancer 2023, 11, e006930. [Google Scholar] [CrossRef]
  110. Jansen, K.; Kornfeld, L.; Lennartz, M.; Blessin, N.C.; Höflmeyer, D.; Dwertmann Rico, S.; Kind, S.; Viehweger, F.; Fraune, C.; Gorbokon, N.; et al. Carcinoembryonic antigen (CEA) expression in human tumors: A tissue microarray study on 15,413 tumors. J. Clin. Oncol. 2023, 41 (Suppl. 16), e15181. [Google Scholar] [CrossRef]
  111. Anoop, T.M.; Joseph, P.R.; Soman, S.; Chacko, S.; Mathew, M. Significance of serum carcinoembryonic antigen in metastatic breast cancer patients: A prospective study. World J. Clin. Oncol. 2022, 13, 529–539. [Google Scholar] [CrossRef]
  112. Parkhurst, M.R.; Yang, J.C.; Langan, R.C.; Dudley, M.E.; Nathan, D.A.; Feldman, S.A.; Davis, J.L.; Morgan, R.A.; Merino, M.J.; Sherry, R.M.; et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther. 2011, 19, 620–626. [Google Scholar] [CrossRef]
  113. Zhao, Y.; Deng, J.; Rao, S.; Guo, S.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; et al. Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers 2022, 14, 4160. [Google Scholar] [CrossRef] [PubMed]
  114. Zacharakis, N.; Huq, L.M.; Seitter, S.J.; Kim, S.P.; Gartner, J.J.; Sindiri, S.; Hill, V.K.; Li, Y.F.; Paria, B.C.; Ray, S.; et al. Breast Cancers Are Immunogenic: Immunologic Analyses and a Phase II Pilot Clinical Trial Using Mutation-Reactive Autologous Lymphocytes. J. Clin. Oncol. 2022, 40, 1741–1754. [Google Scholar] [CrossRef]
  115. Lin, D.; Shen, Y.; Liang, T. Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduct. Target. Ther. 2023, 8, 156. [Google Scholar] [CrossRef]
  116. Shalhout, S.Z.; Miller, D.M.; Emerick, K.S.; Kaufman, H.L. Therapy with oncolytic viruses: Progress and challenges. Nat. Rev. Clin. Oncol. 2023, 20, 160–177. [Google Scholar] [CrossRef] [PubMed]
  117. Andtbacka, R.H.I.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
  118. Hu, J.C.; Coffin, R.S.; Davis, C.J.; Graham, N.J.; Groves, N.; Guest, P.J.; Harrington, K.J.; James, N.D.; Love, C.A.; McNeish, I.; et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin. Cancer Res. 2006, 12, 6737–6747. [Google Scholar] [CrossRef] [PubMed]
  119. Senzer, N.N.; Kaufman, H.L.; Amatruda, T.; Nemunaitis, M.; Reid, T.; Daniels, G.; Gonzalez, R.; Glaspy, J.; Whitman, E.; Harrington, K.; et al. Phase II Clinical Trial of a Granulocyte-Macrophage Colony-Stimulating Factor–Encoding, Second-Generation Oncolytic Herpesvirus in Patients With Unresectable Metastatic Melanoma. J. Clin. Oncol. 2009, 27, 5763–5771. [Google Scholar] [CrossRef] [PubMed]
  120. Soliman, H.; Hogue, D.; Han, H.; Mooney, B.; Costa, R.; Lee, M.C.; Niell, B.; Williams, A.; Chau, A.; Falcon, S.; et al. Oncolytic T-VEC virotherapy plus neoadjuvant chemotherapy in nonmetastatic triple-negative breast cancer: A phase 2 trial. Nat. Med. 2023, 29, 450–457. [Google Scholar] [CrossRef]
  121. Kai, M.; Marx, A.N.; Liu, D.D.; Shen, Y.; Gao, H.; Reuben, J.M.; Whitman, G.; Krishnamurthy, S.; Ross, M.I.; Litton, J.K.; et al. A phase II study of talimogene laherparepvec for patients with inoperable locoregional recurrence of breast cancer. Sci. Rep. 2021, 11, 22242. [Google Scholar] [CrossRef]
  122. Hecht, J.R.; Raman, S.S.; Chan, A.; Kalinsky, K.; Baurain, J.F.; Jimenez, M.M.; Garcia, M.M.; Berger, M.D.; Lauer, U.M.; Khattak, A.; et al. Phase Ib study of talimogene laherparepvec in combination with atezolizumab in patients with triple negative breast cancer and colorectal cancer with liver metastases. ESMO Open 2023, 8, 100884. [Google Scholar] [CrossRef]
  123. Sun, K.; Xu, Y.; Zhang, L.; Niravath, P.; Darcourt, J.; Patel, T.; Teh, B.S.; Farach, A.M.; Guerrero, C.; Mathur, S.; et al. A Phase 2 Trial of Enhancing Immune Checkpoint Blockade by Stereotactic Radiation and In Situ Virus Gene Therapy in Metastatic Triple-Negative Breast Cancer. Clin. Cancer Res. 2022, 28, 4392–4401. [Google Scholar] [CrossRef] [PubMed]
  124. Huppert, L.A.; Gliwa, A.S.; Tait, M.; Quintal, L.; Starzinski, S.; Cheung, A.; Moasser, M.M.; Majure, M.; Melisko, M.E.; Rugo, H.S.; et al. Phase Ib study of talimogene laherparepvec (T-VEC) in combination with chemotherapy (CT) or endocrine therapy (ET) in patients with metastatic, unresectable, or locoregionally recurrent HER2-negative breast cancer (BC). J. Clin. Oncol. 2023, 41 (Suppl. S16), 1091. [Google Scholar] [CrossRef]
  125. Camilio, K.A.; Wang, M.Y.; Mauseth, B.; Waagene, S.; Kvalheim, G.; Rekdal, O.; Sveinbjornsson, B.; Maelandsmo, G.M. Combining the oncolytic peptide LTX-315 with doxorubicin demonstrates therapeutic potential in a triple-negative breast cancer model. Breast Cancer Res. 2019, 21, 9. [Google Scholar] [CrossRef]
  126. Spicer, J.F.; Marabelle, A.; Baurain, J.-F.; Awada, A.; Kristeleit, R.S.; Jossang, D.E.; Jebsen, N.; Loirat, D.; Armstrong, A.C.; Curigliano, G.; et al. A phase I/II study of the oncolytic peptide LTX-315 combined with checkpoint inhibition generates de novo T-cell responses and clinical benefit in patients with advanced solid tumors. J. Clin. Oncol. 2018, 36 (Suppl. S15), 3094. [Google Scholar] [CrossRef]
  127. Clark, A.S.; Zhao, F.; Klein, P.; Montero, A.J.; Falkson, C.I.; Krill-Jackson, E.; Rowland, K.M.; Sardesai, S.D.; Incorvati, J.A.; Dillon, P.M.; et al. BRACELET-1 (PrE0113): Inducing an inflammatory phenotype in metastatic HR+/HER2- breast cancer with the oncolytic reovirus pelareorep in combination with paclitaxel and avelumab. J. Clin. Oncol. 2023, 41 (Suppl. S16), 1012. [Google Scholar] [CrossRef]
  128. Manso, L.; Salvador, F.; Villagrasa, P.; Chic, N.; Bermejo, B.; Cejalvo, J.M.; Izarzugaza, Y.; Cantos, B.; Blanch, S.; Margeli, M.; et al. Abstract CT191: A window-of-opportunity study with atezolizumab and the oncolytic virus pelareorep in early breast cancer (AWARE-1). Cancer Res. 2021, 81 (Suppl. S13), CT191. [Google Scholar] [CrossRef]
  129. Li, W.; Yin, Y.; Cui, J.; Wang, W.; Liang, Y.; Liang, H.; Xu, B. Abstract P3-07-04: A multicenter, single-arm, open-label Phase I study of AN1004 (Pelareorep) oncolytic virus plus paclitaxel in Chinese patients with Hormone receptor-positive and HER2-negative advanced/metastatic breast cancer (REO 026-1). Cancer Res. 2023, 83 (Suppl. S5), P3-07-04. [Google Scholar] [CrossRef]
  130. Nemunaitis, J.; Senzer, N.; Sarmiento, S.; Zhang, Y.A.; Arzaga, R.; Sands, B.; Maples, P.; Tong, A.W. A phase I trial of intravenous infusion of ONYX-015 and enbrel in solid tumor patients. Cancer Gene Ther. 2007, 14, 885–893. [Google Scholar] [CrossRef]
  131. Nokisalmi, P.; Pesonen, S.; Escutenaire, S.; Sarkioja, M.; Raki, M.; Cerullo, V.; Laasonen, L.; Alemany, R.; Rojas, J.; Cascallo, M.; et al. Oncolytic adenovirus ICOVIR-7 in patients with advanced and refractory solid tumors. Clin. Cancer Res. 2010, 16, 3035–3043. [Google Scholar] [CrossRef]
  132. Yuan, Y.; Egelston, C.; Colunga Flores, O.; Chaurasiya, S.; Lin, D.; Chang, H.; Chong, L.M.O.; Seiz, A.; Shah, M.; Meisen, W.H.; et al. CF33-hNIS-anti-PD-L1 oncolytic virus followed by trastuzumab-deruxtecan in a patient with metastatic triple negative breast cancer: A case study. Ther. Adv. Med. Oncol. 2023, 15, 17588359231210675. [Google Scholar] [CrossRef]
  133. Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Perez-Gracia, J.L.; Rodriguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castanon, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [PubMed]
  134. Bentebibel, S.E.; Hurwitz, M.E.; Bernatchez, C.; Haymaker, C.; Hudgens, C.W.; Kluger, H.M.; Tetzlaff, M.T.; Tagliaferri, M.A.; Zalevsky, J.; Hoch, U.; et al. A First-in-Human Study and Biomarker Analysis of NKTR-214, a Novel IL2Rbetagamma-Biased Cytokine, in Patients with Advanced or Metastatic Solid Tumors. Cancer Discov. 2019, 9, 711–721. [Google Scholar] [CrossRef]
  135. Divino, C.M.; Chen, S.H.; Yang, W.; Thung, S.; Brower, S.T.; Woo, S.L. Anti-tumor immunity induced by interleukin-12 gene therapy in a metastatic model of breast cancer is mediated by natural killer cells. Breast Cancer Res. Treat. 2000, 60, 129–134. [Google Scholar] [CrossRef] [PubMed]
  136. Telli, M.L.; Nagata, H.; Wapnir, I.; Acharya, C.R.; Zablotsky, K.; Fox, B.A.; Bifulco, C.B.; Jensen, S.M.; Ballesteros-Merino, C.; Le, M.H.; et al. Intratumoral Plasmid IL12 Expands CD8(+) T Cells and Induces a CXCR3 Gene Signature in Triple-negative Breast Tumors that Sensitizes Patients to Anti-PD-1 Therapy. Clin. Cancer Res. 2021, 27, 2481–2493. [Google Scholar] [CrossRef]
  137. Telli, M.L.; Wapnir, I.; Devitt, B.; Cuff, K.; Soliman, H.; Vinayak, S.; Canton, D.A.; Twitty, C.; Foerter, K.M.; Joshi, R. Abstract P3-09-04: Phase 2, open-label study of intratumoral tavokinogene telseplasmid (tavo) plus electroporation in combination with intravenous pembrolizumab therapy in patients with inoperable locally advanced or metastatic triple-negative breast cancer (mTNBC) (KEYNOTE- 890/OMS-I141). Cancer Res. 2020, 80 (Suppl. S4), P3-09-04. [Google Scholar] [CrossRef]
  138. Ribas, A.; Medina, T.; Kummar, S.; Amin, A.; Kalbasi, A.; Drabick, J.J.; Barve, M.; Daniels, G.A.; Wong, D.J.; Schmidt, E.V.; et al. SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase Ib, Multicenter Study. Cancer Discov. 2018, 8, 1250–1257. [Google Scholar] [CrossRef] [PubMed]
  139. Chien, A.J.; Soliman, H.H.; Ewing, C.A.; Boughey, J.C.; Campbell, M.J.; Rugo, H.S.; Wallace, A.M.; Albain, K.S.; Stringer-Reasor, E.M.; Church, A.L.; et al. Evaluation of intra-tumoral (IT) SD-101 and pembrolizumab (Pb) in combination with paclitaxel (P) followed by AC in high-risk HER2-negative (HER2-) stage II/III breast cancer: Results from the I-SPY 2 trial. J. Clin. Oncol. 2021, 39 (Suppl. S15), 508. [Google Scholar] [CrossRef]
  140. Gandhi, S.; Jones, C.; Opyrchal, M.; Slomba, R.; Attwood, K.; Wang, J.; Gomez, E.C.; O’Connor, T.; Levine, E.; Kalinski, P. Abstract PO1-18-03: Chemokine-modulating regimen (rintatolimod, IFN-α2b, celecoxib): New strategy to drive CD8+T-cells into triple negative breast cancer. Cancer Res. 2024, 84 (Suppl. S9), PO11-18-03. [Google Scholar] [CrossRef]
Table 1. Clinical trials for breast cancer using Bispecific antibody.
Table 1. Clinical trials for breast cancer using Bispecific antibody.
DrugTarget AntigenPopulationPhaseTrial No.Status
ErtumaxomabHER2 and CD3Low Her2 breast cancerINCT00522457
NCT00452140
Terminated
PF-06671008P-cadherin and CD3Advanced solid tumorINCT02659631Terminated
TebotelimabLAG3 and CD3Unresectable and metastatic malignancyINCT032196268Completed
ZW25ECD4 and 2 in Her2Her2-expressing cancerINCT02892123Active, not recruiting
D3L-001Her2/CD47Her2-positive solid tumorINCT05957536Recruiting
AK117 (Ivonescimab)VEFG and PD-1Metastatic TNBCIINCT05227664Completed
PM8002/BNT327VEGF and PDL-1Unresectable, locally advanced or metastatic TNBCIb/IINCT05918133Recruiting
Table 2. Clinical trials for BC using cancer vaccines.
Table 2. Clinical trials for BC using cancer vaccines.
Target AntigenStudy PopulationPhaseNCTStatus
Peptide-based
Nelipepimut-SHer2Operable breast cancer (T1-3N+)IIINCT01479244Completed
GP2Her2HER2/neu-positive subjects with residual disease or high-risk PCR after both neoadjuvant and postoperative adjuvant trastuzumab-based therapyIIINCT05232916Recruiting
AE37Her2-derived peptideNode-positive and high-risk node-negative breast cancer patientsIINCT00524277Completed
VX-001hTERTAdvanced
breast cancer
INCT00573495 Completed
Metastatic breast cancerINCT01660529Completed
Stage III breast cancerINCT00753415Completed
Protein-based
Her2 intracellular domainHer2-positive breast or ovarian cancerI
Her2Stage II–III breast cancerINCT00058526Completed
Carbohydrate-based
STn-KLH (theratope)Sialyl-TnMetastatic breast cancerIIINCT00003638Completed
DNA-based
PANVACCEA and MUC-1Metastatic breast cancerI/IINCT00179309Completed
Her2Metastatic Her2-expressing breast cancerIDnr151:785/2001Completed
Stage II-III breast cancerI/IINCT00250419 and NCT00647114Completed
Advanced-stage Her2-positive breast cancerINCT00436254Active, not recruiting
Dendritic cell-based
Her2Her2-positive DCIS and early breast cancerINCT02061332Completed
Stage II (≥6 + LN), III, or IV breast cancer with >50% HER2 overexpression after adjuvant surgery INCT00005956Completed
p53HLA-A2+ patients with progressive advanced breast cancerIINot applicable (Denmark)Completed
Tumor cell-based (combined with DC)
Metastatic breast and renal cancerINot applicableCompleted
Table 3. CAR-T therapy in breast cancer.
Table 3. CAR-T therapy in breast cancer.
Target AntigenStudy PopulationPhaseNCTStatus
MesothelinMesothelioma, lung and breast cancersINCT02414269Active, not recruiting
Metastatic solid tumorINCT03545815Not applicable
Metastatic TNBCINCT02792114Active, not recruiting
C-METMetastatic TNBCINCT01837602Completed
Metastatic melanoma and TNBCINCT03060356Terminated
MUC-1Advanced MUC1-positive breast cancerINCT04020575Recruiting
ROR-1Advanced ROR1-positive tumorINCT02706392Terminated
NKG2DColorectal, ovarian, bladder, triple-negative breast, and pancreatic cancersINCT03018405Not applicable
Relapsed or refractory solid tumorINCT04107142Not applicable
Her2Her2-positive metastatic solid tumorINCT02713984Withdrawn
Her2-positive solid tumor1NCT04650451Suspended
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Choi, Y.; Tan, J.; Lin, D.; Lee, J.S.; Yuan, Y. Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2025, 26, 3920. https://doi.org/10.3390/ijms26083920

AMA Style

Choi Y, Tan J, Lin D, Lee JS, Yuan Y. Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors. International Journal of Molecular Sciences. 2025; 26(8):3920. https://doi.org/10.3390/ijms26083920

Chicago/Turabian Style

Choi, Yeonjoo, Jiayi Tan, David Lin, Jin Sun Lee, and Yuan Yuan. 2025. "Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors" International Journal of Molecular Sciences 26, no. 8: 3920. https://doi.org/10.3390/ijms26083920

APA Style

Choi, Y., Tan, J., Lin, D., Lee, J. S., & Yuan, Y. (2025). Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors. International Journal of Molecular Sciences, 26(8), 3920. https://doi.org/10.3390/ijms26083920

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop