Next Article in Journal
SIRT1 Promotes Cisplatin Resistance in Bladder Cancer via Beclin1 Deacetylation-Mediated Autophagy
Previous Article in Journal
Assessment of an Anticancer Effect of the Simultaneous Administration of MM-129 and Indoximod in the Colorectal Cancer Model
Previous Article in Special Issue
Evolving CAR-T-Cell Therapy for Cancer Treatment: From Scientific Discovery to Cures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential

by
Diego F. Chamorro
1,†,
Lauren K. Somes
1,† and
Valentina Hoyos
1,2,*
1
Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
2
Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(1), 124; https://doi.org/10.3390/cancers16010124
Submission received: 5 December 2023 / Revised: 19 December 2023 / Accepted: 21 December 2023 / Published: 26 December 2023
(This article belongs to the Collection Mechanism of Immunotherapy in Cancers)

Abstract

:

Simple Summary

Breast cancer stands as the predominant form of cancer identified in women, underscoring the urgent demand for innovative targeted therapies. Engineered adoptive cell therapies represent a groundbreaking approach, empowering the immune cells of patients to precisely target their tumors through cancer-specific receptors. Encouragingly, preclinical studies have illuminated the immense promise of this strategy. Nonetheless, the translation of this technique into clinical practice hinges on the accumulation of additional robust clinical data. Within this review, we offer a comprehensive examination of the current landscape surrounding engineered cell therapies for breast cancer, delving into both their limitations and the compelling prospects for enhancement.

Abstract

Breast cancer remains a significant health challenge, and novel treatment approaches are critically needed. This review presents an in-depth analysis of engineered adoptive T-cell therapies (E-ACTs), an innovative frontier in cancer immunotherapy, focusing on their application in breast cancer. We explore the evolving landscape of chimeric antigen receptor (CAR) and T-cell receptor (TCR) T-cell therapies, highlighting their potential and challenges in targeting breast cancer. The review addresses key obstacles such as target antigen selection, the complex breast cancer tumor microenvironment, and the persistence of engineered T-cells. We discuss the advances in overcoming these barriers, including strategies to enhance T-cell efficacy. Finally, our comprehensive analysis of the current clinical trials in this area provides insights into the future possibilities and directions of E-ACTs in breast cancer treatment.

1. Introduction

Breast cancer is the most common cancer diagnosed in women and the second most common cause of cancer-related death [1]. Clinically, it is divided into four molecular subtypes based on expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) [2]. Various therapeutic modalities that target these receptors are utilized in the treatment of breast cancer, including anti-HER2 monoclonal antibodies and selective ER degraders (SERDs) [3,4]. Despite the great success of these therapies, there is a need for novel targeted treatments that can benefit a wider variety of breast cancer patients.
Engineered adoptive T-cell therapies (E-ACTs), a subset of immunotherapy, equip a patient’s T-cells with engineered receptors that specifically recognize their cancer. These therapies have revolutionized the treatment of hematologic malignancies; however, none have successfully emerged as a clinically relevant option for breast cancer. A thorough understanding of the factors that limit the success of engineered adoptive T-cell therapies in breast cancer is vital to improving their therapeutic outcomes. In this review, we analyze the main types of E-ACTs for breast cancer, their limitations and challenges, and the current landscape of these therapies in clinical trials.

2. Engineered Adoptive T-Cell Therapies for Breast Cancer

E-ACTs are a type of immunotherapy in which the patient’s immune cells are modified to confer a customized immune response to their cancer [5,6,7]. These therapies can be divided into two major categories: chimeric antigen receptor (CAR) T-cell therapy and T-cell receptor (TCR) T-cell therapy. Both approaches are utilized in breast cancer models with varying degrees of success. Table 1 compares TCR T and CAR T-cell receptors regarding their MHC restriction, sensitivity, antigens recognized, and co-stimulatory molecules.
CAR T-cell therapies combine the specificity of a monoclonal antibody (mAb) with the signaling components of a TCR, resulting in a synthetic receptor that is not major histocompatibility complex (MHC)-restricted (Figure 1) [11]. The typical CAR is composed of a single-chain variable fragment (scFv) fused to hinge and transmembrane domains, followed by an intracellular signaling domain [12]. The scFv is derived from the heavy and light chain variable regions of a mAb and is responsible for antigen recognition [13]. The intracellular signaling domain of first-generation CARs is comprised solely of the CD3ζ or FcRɣ signaling domains; clinical efficacy with these first-generation CARs was limited, however, as it was proven that they do not produce a durable anti-tumor response due to a lack of expansion and persistence [14,15]. Second- and third-generation CAR constructs incorporate the signaling domains of known T-cell co-stimulatory molecules. The two most common co-stimulatory domains used are CD28 and 4-1BB (CD137). Others include inducible T-cell co-stimulator (ICOS), CD27, MyD88, CD40, and OX40 (CD134) [16,17]. Fourth-generation CARs, also known as T-cells redirected for universal cytokine killing (TRUCKs), are engineered to incorporate cytokines or their receptors, which serve to support T-cell activity and survival [18,19], resulting in more durable T-cell responses [20].
TCR T-cell therapies, on the other hand, utilize naturally occurring TCRs isolated from T-cell clones that recognize the tumor antigen of interest. The α and β chains of the isolated TCR are expressed in the recipient’s T-cells, which dimerize and associate with endogenous CD3ε/ɣ/δ/ζ subunits to confer the desired specificity (Figure 1) [21]. Engineered TCR (E-TCR) T-cells can recognize peptides from both intracellular and extracellular tumor antigens presented on surface MHC molecules, including neoantigens arising from tumor-specific mutations [6,21]. Since TCRs recognize tumor antigens in an MHC-dependent manner, E-TCRs are matched to the patient’s expressed MHC alleles [22]. As a significant portion of breast cancer cases have mutations in PIK3CA, TP53, and ESR1 [21,23,24], the use of E-TCRs specific for these mutations is an attractive therapeutic avenue.
Compared to the success seen in hematologic malignancies, E-ACTs for breast cancer are limited in their efficacy and feasibility. Six CAR T-cell products have been approved by the US Food and Drug Administration (FDA) for B-cell malignancies and multiple myeloma [25]; currently, there are no FDA-approved CAR T-cell therapies for solid tumors. In the case of TCR T-cells, there are no FDA-approved TCR T-cell products for hematologic or solid tumors to date, and there is little clinical data regarding their efficacy in breast cancer [21]. Many factors limit the clinical success of engineered adoptive cell therapies in breast cancer, including difficulties identifying suitable tumor targets, the immunosuppressive tumor microenvironment, diminishing T-cell persistence, and the costs associated with treatment.

3. Identification of Suitable Tumor Targets

The ideal target antigen for engineered cell therapies is overexpressed by the tumor and absent from normal cells in surrounding tissue and vital organs. A wide variety of breast cancer targets have been identified; however, many of these targets are also expressed on normal cells to a certain degree. Therefore, careful antigen selection is a crucial aspect of E-ACTs to ensure treatment efficacy and patient safety. The three main classes of tumor antigens are tumor-associated antigens (TAAs), cancer-germline antigens (CGAs), and tumor-specific antigens (TSAs) [6,26,27].
TAAs are expressed in malignant cells and some healthy tissues. As a result, targeting these antigens carries the risk of on-target/off-tumor toxicities. This risk, however, can be managed with proper TAA selection. TAAs can be classified as either differentiation or overexpressed antigens. Differentiation antigens are expressed in the tumor and the corresponding healthy tissue and are thus associated with the most significant risk of on-target/off-tumor toxicities [21,28]. Most breast cancer TAAs are overexpressed antigens. Overexpressed antigens are enriched in tumor cells, with minimal expression in healthy cells; however, there is still a risk of on-target/off-tumor toxicity. To overcome this risk, engineered cell therapies can be optimized to necessitate a high antigen density for receptor activation, therefore minimizing the destruction of normal cells with low antigen density [29].
CGAs, also referred to as cancer/testis antigens (CTAs), are a unique group of TAAs that are expressed exclusively in germ cells and absent from normal somatic tissues [30,31]. Their expression in malignant cells results from aberrant gene expression due to DNA hypomethylation [30,32]. CGAs are desirable targets for E-ACT due to their restricted expression pattern. Potential CGA targets for breast cancer have been evaluated for CAR T and TCR T-cell therapies.
Finally, TSAs, as the name suggests, are exclusively expressed in malignant cells. These antigens may arise from mutations (neoantigens) or the expression of viral elements [28,33]. Neoantigens are ideal targets for TCR T-cell therapy as TCRs can detect mutant peptides harboring a single-point mutation without reactivity to the wildtype peptide [34]. While many mutations are patient-specific or “private” neoantigens, driver mutations in certain genes can result in “public” neoantigens that are shared among many breast cancer patients [35].
Table 2 summarizes the most common breast cancer TAA, CGA, and TSA targets under investigation.

Remaining Challenges: Antigen Heterogeneity and On-Target/Off-Tumor Toxicity

In solid tumors, including breast cancer, many target antigens are not uniformly expressed among all cells within the tumor. As a result, patients may initially respond to therapy but later progress due to the outgrowth of antigen-negative tumor cells [118]. HER2 intratumoral heterogeneity, for example, is reported in up to 40% of breast cancers and is a potential mechanism for resistance [119]. Strategies to overcome intratumoral heterogeneity in solid and hematologic malignancies include using epigenetic modulators to increase surface expression of target antigens [120,121,122] and targeting multiple antigens expressed throughout the tumor simultaneously [120,123,124,125]. Few preclinical studies using these approaches to address tumor heterogeneity in breast cancer models exist, and they will be necessary for the success of these therapies in breast cancer. An early study in a murine breast cancer model found that expression of a murine CGA could be increased by treating the cells with an epigenetic drug that inhibits DNA methylation [126]. More studies are needed to test if these treatments may be combined with E-ACTs in in vivo models.
Multi-antigen targeting also reduces the risk of on-target/off-tumor toxicities. While few preclinical studies utilize breast cancer models, a recent study presented an elegant method for restricting CAR T-cells to dual antigen encounters using targets commonly expressed in breast cancer. These logic-gated intracellular network (LINK) CARs only mount an anti-tumor response when both target antigens are present, thus significantly limiting the potential for on-target/off-tumor toxicity [127]. Future studies are needed, however, to test the efficacy of this approach in solid tumor models.

4. Overcoming the Tumor Microenvironment

The breast cancer tumor microenvironment (TME) consists of various suppressive immune cells, stromal cells, and soluble components that together play a vital role in the growth, survival, and spread of malignant cells (Figure 2) [128,129]. The TME is one of the most significant barriers for E-ACTs to overcome, as T-cells must not only perform their anti-tumor functions but also navigate the hostile TME milieu. Efforts to further dissect the complexity of the TME via single-cell RNA sequencing (scRNAseq) analysis have identified various subclusters of immune and stromal cells that perform a variety of pro-tumoral functions [130,131,132,133]. These studies also identify a wealth of potential prognostic markers and therapeutic targets [132,133,134,135,136]. As the full complexity of the TME is beyond the scope of this review, we will summarize the primary TME components that contribute to the inhibition of E-ACTs.

4.1. Immune Microenvironment of Breast Cancer

The breast cancer TME is divided into different compartments based on their cell composition and proximity to the tumor cells [128,129]. The local, or intratumoral, compartment primarily includes several types of immune cells, including regulatory T-cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs). These cell types perform specific functions that promote tumor growth and suppress anti-tumor immunity.

4.1.1. Regulatory T-Cells (Tregs)

Tregs represent one of the primary immune populations that favor immunotolerance in the breast TME. Tregs are characterized by their expression of the transcription factor Foxp3 and are recruited to the TME via chemokines secreted by the breast tumor cells [128,137,138]. Tregs suppress the functions of effector cells through several mechanisms, including the secretion of IL-10, TGFβ, and adenosine, competitive consumption of IL-2, and expression of CTLA-4/PD-L1 [139,140,141]. While the prognostic significance of Tregs in breast cancer reportedly varies among molecular subtypes [139], studies have identified relationships between Treg infiltration and certain prognostic variables. An analysis of tumor samples from various molecular breast cancer subtypes found that elevated numbers of Tregs are associated with aggressive tumor phenotypes, larger tumor size, and estrogen receptor negativity [142,143]. Similarly, a study of breast tumor resident Tregs found that their frequency increases in TNBC. These Tregs also express high levels of chemokine receptor 8 (CCR8), which is associated with higher-grade tumors and poor survival [144].
Due to their prevalence and immunosuppressive capabilities, Tregs are a desirable target for improving the efficacy of E-ACTs. Broadly, methods to inhibit the effects of Tregs include immune checkpoint inhibitors against various co-inhibitory molecules (CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT), depletion via targeting of Treg-specific surface molecules, agonistic antibodies against tumor necrosis factor receptor superfamily molecules (GITR, 4-1BB, OX40, and others), and small molecule drugs targeting characteristic features of Tregs [145,146]. In breast cancer, genetic depletion of Tregs significantly enhanced the efficacy of checkpoint inhibition in a claudin-low TNBC model; however, the use of pharmacologic methods to deplete Tregs also reduced the numbers of infiltrating CD4+ and CD8+ T-cells [147]. The histone deacetylase (HDAC) inhibitor vorinostat was found to decrease the number of Foxp3+ cells in syngeneic 4T1 TNBC tumors and potentiate the effect of checkpoint inhibition [148]. Studies utilizing cyclophosphamide and letrozole found that both drugs can reduce Treg populations in peripheral blood and in the tumor [149,150]. Additionally, a phase I study of daclizumab, an anti-CD25 mAb, in combination with peptide vaccination reported significant depletion of CD25+Foxp3+ Tregs and CD8 responses to the tumor peptides, despite CD25 expression on effector T-cells [151]. Currently, no preclinical studies focus on targeting Tregs in the context of E-ACTs, likely due to difficulties specifically targeting the Tregs while sparing the engineered T-cells.

4.1.2. Myeloid-Derived Suppressor Cells (MDSCs)

MDSCs are a heterogeneous group of immature myeloid cells that arise due to altered myelopoiesis driven by chronic inflammation in cancer and primarily function to suppress T-cell-mediated immune responses [152,153]. They are divided into two main phenotypic subtypes: monocytic MDSCs (M-MDSCs) and granulocytic/polymorphonuclear (G-/PMN-MDSCs). While phenotypic characterizations can vary, M-MDSCs are typically defined as CD11b+CD14+HLA-DR−/lowCD15 cells, and PMN-MDSCs can be defined as CD11b+CD14CD15+ or CD66b+CD15+CD14−/dimCD33dimHLA-DR cells [152,154,155]. Both subtypes of MDSCs maintain an immunosuppressive environment through a variety of mechanisms, including deprivation of metabolic fuels required by T-cells, induction of oxidative stress, recruitment of Tregs, and expression of high levels of PD-L1, to name a few [153,156]. M-MDSCs predominantly mediate their immunosuppressive effects through elevated ARG1, iNOS, and TGFβ expression. On the other hand, PMN-MDSCs dysregulate T-cell function through cell-to-cell contact and ROS production [153].
In metastatic breast cancer, high levels of M-MDSCs are associated with aggressive disease and shorter survival [157,158]. M-MDSCs have been shown to suppress CAR T-cell efficacy in vitro and orthotopic mouse models [52,159]. Preclinical studies describe various methods to target MDSCs in breast cancer models. Drugs that favor the differentiation of MDSCs into mature, less suppressive cell types, such as all-trans retinoic acid, can diminish the immunosuppressive microenvironment [160]. In addition, the HDAC inhibitor entinostat attenuates the immunosuppressive function of G-/PMN-MDSCs in combination with immune checkpoint inhibitors in transgenic breast cancer models [161]. In the context of CAR T-cell therapy, polyinosinic-polycytidylic acid (poly I:C), a ligand of TLR3, not only enhances CAR T-cell function but also decreases MDSC levels in peripheral blood and attenuates their immunosuppressive activity [159]. Furthermore, olaparib suppresses MDSC recruitment through the SDF1α/CXCR4 axis and enhances CAR T-cell efficacy in syngeneic breast cancer models [162].
Methods to arm engineered T-cells with other chimeric receptors that provide co-stimulation while targeting immunosuppressive cell types have also been explored. MDSCs express a receptor called tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptor 2 (TR2). This receptor induces apoptosis upon TRAIL engagement and has previously been targeted using an agonist mAb [163,164]. A novel co-stimulatory receptor composed of the scFv of the TR2 mAb combined with a 4-1BB endodomain successfully protected anti-MUC1 CAR T-cells from MDSC immunosuppression and promoted superior anti-tumor activity in breast cancer models [52].

4.1.3. Tumor-Associated Macrophages (TAMs)

TAMs are another immunosuppressive tumor-associated myeloid cell type. TAMs are classified into two primary phenotypes that depend on cytokine exposure. M1 macrophages, which possess anti-tumor functions, are stimulated by IFNɣ and TNF. M2 macrophages, on the other hand, activated by IL-4, IL-10, and IL-13, promote tumor growth and suppress anti-tumor immune responses [129]. TAMs suppress T-cells by secreting factors such as IL-10, ARG1, iNOS, PGE2, and TGFβ and recruiting Tregs [165,166,167]. TAMs can also express PD-L1, CD80/86, or death receptor ligands that engage with inhibitory receptors on effector cells [166].
In breast cancer, high levels of TAMs are associated with metastasis, lower rates of survival, and overall poor prognosis [168,169]. Preclinical studies suggest that TAMs may be depleted from the breast TME via CARs targeting proteins expressed by both the tumor and TAMs. One potential target, AXL, is a receptor tyrosine kinase expressed in both breast cancer cells and TAMs. Anti-AXL CAR T-cells demonstrate significant anti-tumor activity in TNBC models and have the potential to overcome the immunosuppressive microenvironment by inhibiting TAM cytokine secretion [26,97,98,170]. Another receptor tyrosine kinase, ephrin receptor A10 (EphA10), is detected in TNBC cells, TAMs, and MDSCs. EphA10-specific CAR T-cells have been shown to inhibit in vivo tumor growth in an orthotopic MDA-MB-231 tumor model. Although the effects of these CAR T-cells remain to be tested in immunocompetent models, anti-EphA10 antibodies increased the infiltration and activity of cytotoxic T-cells in a syngeneic 4T1 tumor model, suggesting that blocking EphA10 on TAMs/MDSCs restores T-cell activity [171]. In addition, pharmacologic inhibition of sphingosine 1-phosphate receptor 3 (S1PR3), a bioactive lipid molecule expressed in breast cancer, resulted in TME remodeling via the recruitment of pro-inflammatory macrophages and improved the efficacy of anti-EpCAM CAR T-cell therapy in a murine breast cancer model [61]. Other potential therapeutic avenues to target TAMs in the breast TME include depletion via CSF-1/CSF1R inhibition [172,173,174,175], modulation via class IIa HDAC inhibition [176], and reprogramming via antibodies targeting the pattern recognition scavenger receptor MARCO [177]. Further studies, however, are needed to assess whether these methods can bolster the efficacy of breast cancer E-ACTs.

4.2. Non-Immune Microenvironment of Breast Cancer

In addition to immune cells that suppress effector functions and support tumor growth, the regional, or breast, compartment of the TME is home to other physical and structural components that influence tumor and T-cell behavior. Acellular components such as the extracellular matrix and hostile metabolic conditions, and cellular components such as cancer-associated fibroblasts (CAFs) and endothelial cells, provide structural support for tumor cells, serve as a barrier for anti-tumor immunity, and provide nutrients, proliferative stimuli, and tumor niche protection.

4.2.1. Extracellular Matrix (ECM)

Breast tumors are encapsulated by a complex and dynamic ECM composed of collagens, fibronectin, laminins, glycosaminoglycans and proteoglycans, matricellular proteins, and ECM remodeling enzymes [178]. Collagen within the breast ECM plays a significant role in cancer progression and metastasis [179,180,181,182] and presents a physical barrier that adoptively transferred T-cells must traverse to infiltrate the tumor tissue [183]. Discoidin domain receptor 1 (DDR1) provides further ECM fortification by promoting collagen fiber alignment. This, in turn, suppresses anti-tumor immunity and promotes breast cancer growth by preventing T-cell infiltration [183,184]. Antibodies to neutralize DDR1 effectively disrupt collagen alignment and support anti-tumor immunity [183]. Furthermore, macrophages are an essential source of matrix metalloproteinases (MPPs) that degrade ECM components. In a preclinical study, macrophages engineered to express an anti-HER2 CAR significantly inhibited HER2-4T1 tumor growth in an immunocompetent model [185].

4.2.2. Cancer-Associated Fibroblasts (CAFs)

CAFs are derived from normal fibroblasts activated by tumor-derived inflammation and are the most prominent cell type in the breast TME. CAFs perform various functions, including remodeling the TME, promoting tumor malignancy and angiogenesis, suppressing immune cells, and acting as a physical barrier to infiltrating T-cells [186,187,188]. Breast cancer CAFs express high levels of stromal cell-derived factor-1 (SDF-1), which is responsible for promoting angiogenesis via recruitment of endothelial progenitor cells (EPCs) and stimulating tumor growth [189]. In addition, CAF-derived exomes contain micro RNAs (miRNAs) that promote breast cancer progression and metastasis [190].
Due to their prevalence in the breast TME and pro-tumoral functions, CAFs are an attractive target for E-ACTs. CAR T-cells targeting fibroblast activation protein (FAP) have been evaluated in syngeneic 4T1 TNBC mouse models with conflicting results. One study found that FAP-specific CAR T-cells demonstrated minimal anti-tumor activity and caused bone toxicities due to expression of FAP on bone marrow stromal cells [191]. Other preclinical studies of anti-FAP CAR T-cells derived from other mAb clones report significant anti-tumor activity in 4T1 [192] and other solid tumor models [193], with no on-target/off-tumor toxicities. Despite limited success in breast cancer thus far, methods for targeting CAFs to improve T-cell efficacy are being actively investigated. A recent study found that CAFs secrete high levels of IL-6, increasing PD-L1 expression in TNBC cells and inhibiting CAR T-cell efficacy. The authors recommend the exploration of methods to target the signaling pathways driving IL-6 and PD-L1 expression to improve the response to CAR T-cell therapy [80].

4.2.3. Endothelial Cells

Endothelial cells within the TME play a pivotal role in tumor angiogenesis through their expression of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptors (VEGFR) [194,195]. Tumor-associated endothelial cells also hinder T-cell extravasation by suppressing the required endothelial adhesion molecules [196]. In breast cancer, tumor cells can promote mesenchymal phenotypes in endothelial cells, which favor tumor proliferation, stemness, and invasiveness [197]. Due to their pivotal role in the breast TME, endothelial cells are a prime target for engineered T-cell therapies. CAR T-cells specific for tumor endothelial marker 8 (TEM8), a cell surface protein that functions in endothelial cell migration and invasion, are effective not only against TNBC cells but also against the associated tumor endothelium [198]. Furthermore, a ligand-based CAR utilizing VEGF-C as the antigen binding domain effectively targeted VEGFR-2 and VEGFR-3-positive breast cancer cells and the tubular structures formed by human umbilical vein endothelial cells (HUVECs), thus inhibiting angiogenesis [194]. Moderate anti-tumor effects were seen in a study utilizing VEGFR-2-specific CAR macrophages. However, the authors did not evaluate their impact on the associated endothelial cells [199].

4.2.4. Metabolic Conditions

Hypoxia and competition for metabolic fuels within the TME contribute to exacerbated immunosuppression and poor T-cell survival. In the hypoxic TME, tumor cells, immune cells, and other cellular components constantly compete for limited amounts of metabolic fuels and nutrients [200]. The lack of essential nutrients and low availability of oxygen force T-cells to adopt an anaerobic metabolism, hindering full T-cell activation. In addition, other cellular components of the TME can deplete amino acids essential for T-cell activation and proliferation, such as arginine and cysteine, and release reactive oxygen species that hinder T-cell signaling [201]. Tumor cells can produce high levels of metabolites like adenosine and lactate, leading to T-cell inhibition [202]. CAFs and mesenchymal cells can also produce toxic metabolites for T-cells [203], and it has been shown that selective depletion of those cell populations diminishes the immunosuppressive conditions and improves T-cell metabolic function [204,205,206].
Other strategies to combat the effects of these “chemical” immunosuppressors have been tested in preclinical breast cancer models. For example, adenosine exerts immunosuppressive effects in T-cells through the A2a receptor (A2aR) [207]. mRNA and protein analysis of breast tumor samples also revealed that A2aR expression is associated with aggressive phenotypes, poor survival, and immunosuppressive immune infiltrates [208]. CRISPR/Cas9 knockout of A2aR can increase the effector function of CAR T-cells without compromising their memory phenotype or persistence. A2aR knockout CAR T-cells mediated an enhanced therapeutic response in a HER2+ murine breast cancer model [209].

5. Persistence of Adoptively Transferred Engineered Cells

While E-ACTs have the potential to become a powerful component of the breast cancer treatment arsenal, success is limited in part due to a lack of cell persistence at the tumor site. Compared to cell therapies for hematologic malignancies, which encounter the cancer cells immediately upon entering the bloodstream, T-cells redirected against breast cancer must endure long enough to navigate to and penetrate the immunosuppressive TME. Only after this do they encounter their target antigen. Moreover, for successful tumor debulking and elimination, these cells must not only reach the tumor but also thrive and remain within it until the clearance of malignant cells. Several strategies have been utilized to support CAR T-cell persistence and expansion in breast cancer models (Figure 3).

5.1. Engineered Chimeric Receptors

One approach to improve T-cell persistence in suppressive breast cancer TME is the inclusion of engineered receptors that provide additional cytokine signaling or co-stimulation. This added support can help CAR T-cells resist inhibitory signals. Inverted cytokine receptors, for instance, provide proliferative signals in response to immunosuppressive cytokines in the TME, turning the tumor’s defenses against it. IL-4 is an inhibitory Th2 cytokine that has been shown to promote the survival of breast cancer cells and support Tregs [210,211,212]. An inverted cytokine receptor composed of the IL-4 receptor exodomain fused to the IL-7 receptor endodomain improved the persistence and anti-tumor activity of anti-MUC1 CAR T-cells against an IL-4-secreting breast cancer CDX model [53]. As the immunosuppressive TME typically contains low levels of the immunostimulatory cytokines necessary to maintain CAR T-cell activation, IL-7 signaling has also been provided as an engineered constitutively active receptor known as C7R [213]. C7R activates the IL-7 signaling axis without needing extracellular cytokines and enhances the anti-tumor activity of anti-AXL CAR T-cells in TNBC models [98,213]. Currently, three E-ACT clinical trials include C7R co-stimulation (NCT04099797, NCT03635632, NCT04664179).

5.2. Soluble Cytokine Production

CAR T-cells engineered to secrete specific cytokines are also known as TRUCK (T-cells Redirected towards Universal Cytokine Killing) CAR T-cells [187,214]. TRUCK CAR T-cells incorporating various soluble cytokines, including IL-15, IL-7, and IL-18, have been explored for breast cancer.

5.2.1. Interleukin-15 (IL-15)

While IL-15 is structurally similar to IL-2, it possesses certain functions in vivo that distinguish it from IL-2 and make it a desirable candidate for enhancing E-ACT efficacy [215]. Unlike IL-2, IL-15 does not affect Treg expansion. It regulates tumor-infiltrating lymphocyte numbers within the TME and plays a crucial role in T-cell activation, expansion, differentiation, and function [215,216,217]. In a recent preclinical study, IL-15 co-expression enhanced the persistence and anti-tumor activity of anti-EGFRvIII CAR T-cells in a murine breast cancer model [218]. These CAR T-cells also expressed CXCR2, which accelerated T-cell trafficking to the tumor site via chemokines expressed in the breast TME [218]. Additional studies in other solid tumor models also demonstrate that IL-15 co-expression is a powerful method to enhance T-cell proliferation and anti-tumor activity in the solid TME [216,217,219,220,221,222]. These encouraging results will hopefully inspire additional breast cancer studies and lead to clinical investigations.

5.2.2. Interleukin-7 (IL-7)

As with IL-15, IL-7 is a critical player in the expansion of naïve and memory T-cells and does not affect Tregs [223,224]. In preclinical breast cancer studies, IL-7 is often combined with chemokines such as CCL19 and CCL21 to improve the chemotaxis of CAR T cells and other immune cells to the tumor site. In combination with CCL21, IL-7 improved the anti-tumor activity of CLDN18.2-specific CAR T-cells in a syngeneic mouse model without lymphodepletion [225]. Another study found that anti-Folate Receptor α (FRα) CAR T-cells co-expressing IL-7 and CCL19 demonstrated superior T-cell infiltration [226]. The addition of CCL19 and CCL21 increased the infiltration of endogenous dendritic cells [225,226], while CCL21 also showed inhibition of tumor angiogenesis [225]. IL-7 is also being investigated in other solid tumor CAR T-cell models [227,228,229].

5.2.3. Interleukin-18 (IL-18)

IL-18 is a pleiotropic cytokine with various functions in different T-cell types. Most notably for CAR T-cell therapies, IL-18 induces IFNɣ production, decreases Tregs, and increases the expansion of CD8+ T-cells [230,231,232]. Ruixin and colleagues’ previously mentioned study of anti-EGFRvIII CAR T-cells with CXCR2 also included conditions with IL-18 co-expression (as opposed to IL-15). Similar to their results for IL-15, anti-EGFRvIII CAR T-cells co-expressing IL-18 had reduced expression of exhaustion markers and superior anti-tumor activity [218]. Additional studies in other solid tumor models have also produced promising results [231,233,234,235].

6. Cost of Autologous Therapy

One of the significant challenges of autologous E-ACT is the cost. Autologous E-ACT requires a lengthy and complex manufacturing process that must be carefully orchestrated and tailored to each patient. For example, current FDA-approved CAR T-cell therapies range from $373,000 to $475,000 for a single infusion [236,237,238]. Overall, the exorbitant costs of E-ACT can be improved by lower-cost manufacturing techniques and allogeneic, or “off-the-shelf,” therapies. Such techniques are beginning to be explored in the context of breast cancer.

6.1. Non-Viral Manufacturing Techniques

Genetic modifications for cellular immunotherapies are commonly introduced via viral transduction. While this method results in high transduction efficiencies, it is extremely labor intensive, has limited capacity for multigene insertions, and is expensive to produce clinically [239,240,241]. Non-viral manufacturing techniques, such as transposons and in vitro-transcribed (IVT) mRNA, have emerged as potential low-cost alternatives.

6.1.1. Transposon Systems

DNA transposons are mobile genetic elements excised from one part of the genome and integrated into another using a “cut and paste” mechanism mediated by a transposase enzyme [242]. Sleeping Beauty (SB) is one of the most widely used transposon systems for genetically modifying human cells. SB is a synthetic transposon derived from inactive transposon sequences in fish genomes [243,244]. The SB vector system comprises two components: the transposon DNA, consisting of the gene of interest flanked by inverted terminal repeats (ITRs), and the SB transposase [243,244]. The SB transposase recognizes the ITR sequences and transfers the transgene from the donor vector to the acceptor site in the genome [244]. Much of the preclinical validation of SB as a viable method for genetic manipulation in E-ACT is focused on CAR T-cell therapy for hematological malignancies and is well-reviewed by Moretti et al. [244].
Compared to viral vectors, the production of transposon plasmids under GMP conditions is significantly cheaper and faster. Using transposons also does not require the complex biohazard procedures associated with viral vector production [242]. Despite these advantages, few clinical trials evaluating E-ACT for breast cancer utilize transposon systems. As of July 2023, two clinical trials use the SB system for CAR/TCR T-cell therapy for breast cancer (NCT04102436, NCT05694364). These trials are in the recruiting phase, with no results posted.

6.1.2. In Vitro-Transcribed (IVT) mRNA

Another alternative to viral manufacturing techniques is IVT mRNA. While the process mirrors viral transduction, IVT mRNA is unique in that genetic modification is transient, allowing for enhanced safety and the ability to modulate expression levels [240,244,245,246]. After isolation and expansion of patient T-cells, IVT mRNA encoding the construct of interest, such as a CAR, TCR, or cytokine, is electroporated into the cells. After mRNA translation is confirmed, the cells are re-infused into the patient [245]. Early preclinical papers demonstrate that electroporation of TCR-encoding IVT mRNA into T-cells produces cytotoxic CTLs that specifically recognize the target peptide [247,248]. In recent years, IVT mRNA has been further optimized with ionizable lipid nanoparticle-mediated mRNA delivery for improved viability compared to electroporation, as well as the ability to reprogram T-cells in situ using IVT mRNA carried by polymeric nanoparticles targeted to cytotoxic T-cells [240,249,250].
Clinical trials have been initiated using IVT mRNA CAR T-cells for solid tumors, including breast cancer. A clinical trial evaluating the safety and feasibility of mRNA-transfected anti-c-Met CAR T-cells found that all patients tolerated a single intra-tumoral injection of 3 × 107 or 3 × 108 cells. However, no measurable clinical responses were observed (NCT01837602) [47]. The transient nature of mRNA expression may necessitate multiple infusions due to reduced CAR T-cell persistence in vivo [244]. For example, an early phase I clinical trial evaluating the same mRNA-transfected anti-c-Met CAR T-cells for breast cancer and melanoma patients planned to administer up to six doses of 1 × 108 modified T-cells over a short two-week period; however, this study was unfortunately terminated due to a halt in funding (NCT03060356). Safety concerns regarding the administration of multiple T-cell doses and the large amount of T-cell product required for repeated dosing raise questions regarding the feasibility of IVT mRNA-modified T-cells, and more trials are needed to fully evaluate their efficacy in various solid tumors, particularly breast cancer.

6.2. Allogeneic (“Off-the-Shelf”) Therapies

Allogeneic, or “off-the-shelf”, therapies are a desirable alternative to current autologous methods for breast cancer E-ACT. Not only are allogeneic therapies more cost-effective, as cell products can be banked for future use and administered upon request, but immune cells from healthy donors also have greater cellular fitness than patient immune cells that have been through multiple rounds of cytotoxic therapies [251,252]. One of the major concerns regarding allogeneic therapies is mitigating graft-versus-host disease (GvHD). GvHD occurs due to human leukocyte antigen (HLA) mismatches between the donor and the recipient [253]. The immunocompetent donor T-cells recognize the recipient as foreign, resulting in life-threatening cytotoxic activity that could seriously harm the patients [254]. Natural killer (NK) cells and gamma delta (ɣδ) T-cells are promising candidates for allogeneic therapies in breast cancer, as both can circumvent the need for HLA matching.

6.2.1. Natural Killer (NK) Cells

NK cells are innate cytotoxic immune cells that recognize targets in an antigen-independent manner and play a significant role in tumor surveillance [255,256]. The fact that NK cells do not require HLA matching makes them an attractive candidate for allogeneic CAR-NK cell therapies [256]. NK cells can be isolated from three primary sources: donor peripheral blood (PB), cord blood (CB), or differentiation from CB hematopoietic stem and progenitor cells (HSPCs) or induced pluripotent stem cells (iPSCs) [252]. Although PB is an easily accessible source of NK cells, issues with donor-to-donor variability and the limited number of NK cells in a single pheresis present challenges [252,256]. CB NK cells are present at higher numbers and can be easily expanded, however, donors cannot be used again as there is a finite amount of starting material; moreover, regulations regarding the use of CB vary among countries [252,253]. Conversely, iPSCs have immense proliferative potential, are easily genetically modified, and allow for a homogenous cell product [252,253,256]. In addition to donor-derived NK cells, numerous studies have utilized the NK-92 cell line as an alternative to primary NK cells. However, NK-92 cells have reduced anti-tumor potency due to the need for irradiation [257].
Preclinical studies of CAR-NK cells for breast cancer have primarily utilized modified NK-92 cells [41,258,259,260,261,262,263] or, less frequently, PB-derived NK cells [264]. Overall, NK-92-derived and PB-derived CAR-NK cells efficiently traffic to the tumor site [41] and exhibit specific anti-tumor cytotoxicity in preclinical models [258,260,261,262,264]. Despite their potential for allogeneic therapy, both registered CAR-NK cell trials for breast cancer utilize autologous cell products (NCT05686720, NCT02839954).

6.2.2. Gamma Delta (ɣδ) T-Cells

ɣδ T-cells constitute 1–5% of lymphocytes and primarily reside in epithelial tissues [251,265,266]. Most circulating ɣδ T-cells express a Vɣ9Vδ2 TCR specific for nonpeptide phosphoantigens without CD4 or CD8 coreceptors [267]. Despite their small numbers, ɣδ T-cells contribute to anti-tumor immunity through their co-expression of activating NK receptors and Toll-like receptors and their ability to lyse target cells [266]. Furthermore, ɣδ T-cells are an ideal candidate for allogeneic cell therapies because their TCR can recognize targets in an MHC-independent manner, minimizing GvHD risk [251].
Studies have demonstrated that ɣδ T-cells can target breast cancer cell lines both in vitro and in vivo [267,268,269]. We did not encounter any preclinical studies of CAR ɣδ T-cells that focus on breast cancer, however, a study using off-the-shelf anti-GPC3 CAR ɣδ T-cells controlled hepatocellular carcinoma tumor growth without evidence of GvHD [216]. One clinical trial was planned to assess allogeneic NKG2DL-targeting CAR ɣδ T-cells for various relapsed or refractory solid tumors, including TNBC, but the trial status is unknown (NCT04107142).

7. Current Clinical Trials

7.1. Trends in E-ACT Trials for Breast Cancer

A search of clinicaltrials.gov using the keywords “breast cancer”, “CAR T-cells”, and “TCR T-cells” (as well as synonyms “CAR” and “TCR”) yielded a total of 49 unique trials (as of 24 July 2023). One additional trial was identified during the literature search for a total of 50 trials. A comprehensive list of these trials is found in Supplementary Table S1. Among these trials, 32 (64%) utilized CAR T-cells, 14 (26%) used TCR T-cells, and the remaining 4 (8%) utilized engineered cells other than traditional αβ T-cells (Figure 4A). The earliest E-ACT trials involving breast cancer were initiated in 2013 (NCT01837602, NCT01967823). The University of Pennsylvania began a study to test the safety and efficacy of intratumoral injections of anti-c-Met CAR T-cells in patients with metastatic breast cancer [47]. The first TCR T-cell trial involving breast cancer targeted NY-ESO-1 and was initiated at the National Cancer Institute (NCI). During the last decade, there has been a steady increase in the cumulative number of E-ACT trials, with distinct peaks around 2015/2016 and 2020 (Figure 4B). Among the E-ACT trials to date, 29 (58%) are in Phase I, 15 (30%) are in Phase I/Phase II, and only three (6%) are in Phase II (Figure 4C). No Phase III trials have been initiated, likely due to a lack of positive results that warrant a Phase III trial. As of 24 July 2023, 23 trials are recruiting (46%), six have been completed (12%), and a total of 18 have been terminated (6, 12%), withdrawn (3, 6%), suspended (2, 4%), or their status is unknown (7, 14%) (Figure 4D). Lymphodepletion is associated with the augmented function of adoptively transferred immune cells, as it expands tumor-reactive T-cells and suppresses endogenous Tregs [270]. Most current E-ACT trials for breast cancer include lymphodepletion in their protocols (48%), and only two trials (4%) have chosen to omit lymphodepletion for reasons not cited (Figure 4E).
A wide variety of antigens are targeted in these trials. Unsurprisingly, the most common CAR T-cell target for breast cancer is HER2 (25%). Patients’ tumors are only considered HER2+ if they have a HER2 immunohistochemistry (IHC) score of 3+ or an IHC score of 2+ with positive fluorescence in-situ hybridization (FISH) [3]. Anti-HER2 CAR T-cells are an attractive alternative for HER2-low breast cancers, i.e., breast cancers with a HER2 IHC score of 2+ without a positive FISH or a score of 1+. Other CAR T-cell targets being investigated include MUC1 (18.8%) and mesothelin (15.6%), both of which have demonstrated promising results in preclinical studies [51,53,57,271]. Compared to CAR T-cells, TCR T-cells have the advantage of targeting intracellular CGAs and neoantigens that would otherwise be inaccessible to CAR T-cells. CGAs and neoantigens also tend to have more restricted expression, minimizing potential on-target/off-tumor toxicities [32]. The most common TCR T-cell targets include NY-ESO-1 (36%), MAGE-A3 (14%), and KK-LC-1 (14%). A comprehensive list of antigen targets for both CAR T and TCR T trials can be found in Figure 4G. Unlike CAR T-cells, successful TCR T-cell treatment requires that the patient possesses compatible HLA alleles. Among TCR-T trials, HLA-A*02 was the most prevalent restricting HLA (50%), followed by HLA-A*01 (21.4%) (Figure 4F). Most E-ACT trials for breast cancer utilize autologous cell products, apart from two current trials evaluating allogeneic CAR T-cells (NCT05239143) and CAR ɣδ T-cells (NCT04107142). These “off-the-shelf” therapies will significantly reduce patient waiting times and minimize the labor required to manufacture a single dose of CAR T-cells. Furthermore, some trials are exploring engineered cell products derived from other cell types, including CAR NK cells (NCT02839954, NCT05686720) and CAR macrophages (NCT04660929) targeting MUC1, mesothelin, and HER2, respectively.
Most breast cancer E-ACT trial locations are in the United States (58%), followed by China (36%), Malaysia (2%), Israel (2%), and Japan (2%) (Figure 4H). Among the 29 E-ACT trials held in the United States, 41.4% are sponsored by academic institutions, 34.5% by industry, and the NIH sponsors 24.1%. Most trials opened at academic institutions are from the University of Pennsylvania (3 trials, 25%), followed by Memorial Sloan Kettering Cancer Center, Fred Hutchinson Cancer Center, and Baylor College of Medicine, each with two trials. (Figure 4I). Each of the trials sponsored by the industry is supported by a different company, as listed in Figure 4I. The NCI initiated all NIH trials. In the earlier years of breast cancer E-ACT trials (2013–2017), trial sponsorship primarily came from academic institutions and the NIH. Since 2018, however, the number of industry-sponsored trials has increased, which helps to accelerate the initiation of new trials.
Trial results were located via analysis of published manuscripts. We identified seven trials that have published results from breast cancer patients. Clinical results of four CAR and three TCR T-cell trials are detailed in Table 3. We provide a broad analysis of the results from these trials, with particular emphasis on their safety and feasibility. Of note, all seven trials are in the early phase and therefore have a relatively small sample size. Despite their small numbers, the results of these early trials are a crucial step toward the clinical establishment of these products and will guide the conduct and design of future trials.

7.2. Safety and Efficacy of CAR T-Cells for Breast Cancer

7.2.1. c-Met-Specific CAR T-Cells: A Safe and Moderately Effective Target

Two CAR T-cell trials with published results for breast cancer patients target c-Met. An initial phase 0/phase I study evaluated the safety and feasibility of intratumoral (I.T.) injections of autologous c-Met CAR T-cells in patients with metastatic breast cancer (four TNBC, two ER+HER2). Three patients received dose level 1 (DL1) at 3 × 107 cells, and three received DL2 at 3 × 108 cells. It was not specified whether lymphodepletion was used. No measurable clinical responses were observed, and all grade 3 serious adverse events (SAEs) were deemed unrelated to the study drug [47]. CAR mRNA was detected in the peripheral blood or tumor tissue in 5 of 6 (83.3%) patients, however, levels became Undetectable 24 h after I.T. injection. These initial results demonstrated the safety of autologous c-Met CAR T-cells. Earlier this year, the same group published results from an additional early phase I study evaluating the safety and efficacy of intravenous (I.V.) c-Met CAR T-cells. This study included four TNBC patients and three melanoma patients. Patients received six doses of 1 × 108 cells each over 14 days without lymphodepletion. Published study results observed 4 of 7 (57.1%) patients with stable disease (SD) and 3 of 7 (42.8%) with progressive disease (PD). Among the four TNBC patients, two had SD, and two showed PD. Of the seven total patients, six (85.7%) experienced grade 1/2 toxicity, and one (14.3%) experienced grade 1 cytokine release syndrome (CRS). No grade 3 or higher toxicity, neurotoxicity, or treatment discontinuation occurred [272]. Together, the results from these two trials demonstrate the safety and therapeutic potential of c-Met CAR T-cells. Additional trials and preclinical studies are warranted to fully evaluate their clinical efficacy.

7.2.2. ROR1-Specific CAR T-Cells: Initial Safety and Poor Intratumoral Persistence

One phase I study evaluated the safety, persistence, trafficking, and preliminary antitumor activity of ROR1-specific CAR T-cells in patients with ROR+ solid and hematologic malignancies. A total of 21 patients with various tumor types participated in this trial and were divided among three DLs with lymphodepletion. A subsequent manuscript describes the results of three metastatic TNBC patients and will be the focus of this analysis [273]. Among these patients, 1 of 3 (33.3%) progressed after treatment, while 2 of 3 (66.7%) had SD, with one patient achieving a partial response (PR) after advancing from DL2 to DL3. Only one TNBC patient experienced grade 1 CRS. As for CAR T-cell expansion and trafficking in mTNBC patients, 2 of 3 (66.7%) experienced a robust CAR T-cell expansion in their peripheral blood, with no toxicity to normal tissues. At peak expansion, however, these T-cells upregulated inhibitory receptors and lost the ability to produce crucial effector cytokines. Unfortunately, CAR T-cells did not accumulate or persist at the tumor site. The success of ROR1-specific CAR T-cells may require additional cytokine stimulation or co-stimulatory receptors. One such trial is currently recruiting and incorporates membrane-bound IL-15, intrinsic PD-1 blockade, and a kill switch for additional safety (NCT05694364).

7.2.3. Mesothelin-Specific CAR T-Cells: Emerging Results from an Ongoing Trial

A phase I/II trial evaluating regional delivery of mesothelin-specific CAR T-cells in patients with malignant pleural disease is ongoing (NCT02414269), and phase I results that include one breast cancer patient were recently published [274]. This patient received 3 × 105 CAR T-cells per kilogram intrapleurally via intervention radiology-guided imaging following lymphodepletion. No adverse events were noted, and the patient received multiple lines of therapy after CAR T-cells and survived 11 months. These preliminary results suggest that mesothelin is a safe target that warrants further clinical investigation.

7.3. Safety and Efficacy of TCR T-Cells for Breast Cancer

7.3.1. NY-ESO-1-Specific TCR T-Cells: Additional Clinical Data Needed

While NY-ESO-1 is by far the most clinically evaluated target for TCR T-cell therapy in breast cancer, few studies report results from breast cancer patients. However, one study published in 2022 provides a small window into the efficacy of NY-ESO-1 redirected T-cells in breast cancer. This phase I study tested NY-ESO-1-specific TCR T-cells in HLA-A*2:01 or HLA-A*2:06 positive patients, including one breast cancer patient treated with one dose of 5 × 108 cells with lymphodepletion (NCT02366546). NY-ESO-1 expression for this patient was between 5–25%, as determined by IHC. Although the patient did not develop CRS, the transferred T-cells did not expand well in peripheral blood, and the patient progressed soon after treatment and passed away at six months post-treatment [276]. However, three other patients enrolled in the study showed greater than 30% tumor regression, albeit with much higher NY-ESO-1 expression (>/=75%). The poor response of the breast cancer patient, therefore, is likely due to low antigen density. A larger breast cancer patient population is needed to assess the safety and efficacy of NY-ESO-1-specific TCR T-cells accurately.

7.3.2. MAGE-A3-Specific TCR T-Cells: Toxicity and No Evidence of Efficacy in Breast Cancer

Early reports of TCR T-cell trials targeting MAGE-A3 describe severe cardiac toxicity due to the cross-reactivity of the TCR to normal cardiac proteins [21]. A later study aimed to target MAGE-A3 in patients with metastatic cancer who were HLA-DPB1*04:01 positive (NCT02111850). The primary objective was to determine the maximum tolerated dose of TCR T-cells. Two breast cancer patients were enrolled in the high-dose cohort, receiving 7.8 × 1010 (plus three doses of IL-2) and 9 × 1010 (plus one dose of IL-2) cells with lymphodepletion. Of note, the patient treated with 9 × 1010 T-cells and one dose of IL-2 experienced grade 4 toxicities, including elevated ALT, AST, and creatinine. The patient later developed respiratory failure, requiring hospitalization. Both breast cancer patients exhibited no response (NR) to treatment [275]. A similar study was conducted in patients with HLA-A*01; however, it was terminated due to slow, insufficient accrual (NCT02153905). Preliminary results for only three participants are posted, and it cannot be determined if any are breast cancer patients.

7.3.3. Neoantigen-Specific TCR T-Cells: Promising Results from an Ongoing Trial

An ongoing phase II clinical trial recently published results from a chemo-refractory breast cancer patient treated with T-cells transduced with an HLA-A*02:01-restricted TCR specific for the p53 R175H mutation. TCR T-cells were administered following standard lymphodepletion with no IL-2. The patient also received one dose of pembrolizumab post-ACT. After infusion, the patient developed grade 3 acute CRS, which resolved following intubation, vasopressors, and steroid treatment. Despite initial distress, the patient exhibited a 55% decrease in tumor burden at 14 weeks post-treatment. By day 60, metastatic sites had decreased, and all detectable skin lesions had resolved. Infused R175H-TCR T-cells also persisted and developed into memory T-cells that could be detected four months post-treatment. While the patient initially had a PR, they progressed at six months post-treatment. New lesions were found to have lost expression of HLA-A*02:01, thereby allowing the tumor cells to escape R175H-TCR T-cell recognition [116]. While these results warrant further investigation into preventing tumor immune escape through HLA loss, the initial anti-tumor efficacy of R175H-TCR T-cells is very promising.

8. Conclusions

E-ACTs for breast cancer are rapidly evolving, with no shortage of targets to explore. While great strides have been made in the preclinical assessment of these novel therapies, including technologies to improve T-cell persistence and target the immunosuppressive TME, most preclinical models are limited in their ability to recapitulate the full scope of interactions between the breast microenvironment and patient immune system. Various studies have been performed in syngeneic mouse models, utilizing murine breast cancer lines in immunocompetent mice and allowing for a comprehensive study of E-ACTs in the context of a complete TME [61,162,185,192,199,218,273,277]. However, these studies require the use of fully murine receptors that cannot be immediately translated to clinical trials without humanization. Fully humanized mice with an intact immune system can be achieved through transplantation of human CD34+ hematopoietic stem/progenitor cells [278]. While humanized breast cancer models have been developed [279,280,281], further studies are needed to establish protocols for breast cancer E-ACT in these models.
Despite the limitations of preclinical models, we present a wealth of evidence supporting the efficacy of E-ACTs for breast cancer. The real test, however, lies in their clinical efficacy. Available phase I/II trial results for patients with breast cancer demonstrate the general safety of CAR T-cell therapies; however, we eagerly await the analysis of results from numerous ongoing and completed trials. Early trials of TCR T-cell therapies for breast cancer report varied results, with some therapies showing great promise and others resulting in severe toxicities. Overall, while there are 14 CAR/TCR trials listed as completed, terminated, or suspended, results regarding breast cancer patients from only seven trials could be located. Regardless of the success of the trial, all results must be published upon trial closure. The results of these trials will guide future research and foster advancements in the technology and safety of E-ACT. Given the steady rise in the number of clinical trials using E-ACT for breast cancer since 2013, we can anticipate an influx of results in the coming years that will influence the next generation of cellular therapies. In the meantime, new studies with “armored” next-generation engineered T-cells are emerging, promising improved T-cell performance and clinical outcomes. The continued efforts to understand and overcome the factors that limit the efficacy of these therapies in solid tumors will light the way toward success.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers16010124/s1, Table S1: Clinical Trials.

Author Contributions

L.K.S. and D.F.C. contributed to the article’s development, writing, and review. V.H. contributed to the conceptualization and review of the paper and the creation of the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Cancer Prevention and Research Institute of Texas (CPRIT) Scholar Award (RR170024), The V Foundation Translational Grant (T2021-018), The AACR and Victoria’s Secret Global Fund for Woman’s Cancers Career Development Award (993916), and the National Institute of General Medical Sciences of the National Institutes of Health (T32GM136554). The content is solely the responsibility of the authors and does not necessarily represent the official views of CPRIT, the V Foundation, AACR/Victoria’s Secret, or the National Institutes of Health.

Conflicts of Interest

V.H. has submitted a patent application for the TR2.41BB construct and does consulting for AstraZeneca. V.H. and L.K.S. have submitted a patent application for the use of anti-ZP4 monoclonal antibodies.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast Cancer. Nat. Rev. Dis. Prim. 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  3. Mercogliano, M.F.; Bruni, S.; Mauro, F.L.; Schillaci, R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers 2023, 15, 1987. [Google Scholar] [CrossRef] [PubMed]
  4. Soleja, M.; Raj, G.V.; Unni, N. An Evaluation of Fulvestrant for the Treatment of Metastatic Breast Cancer. Expert Opin. Pharmacother. 2019, 20, 1819–1829. [Google Scholar] [CrossRef] [PubMed]
  5. Rath, J.A.; Arber, C. Engineering Strategies to Enhance TCR-Based Adoptive T Cell Therapy. Cells 2020, 9, 1485. [Google Scholar] [CrossRef]
  6. Wei, F.; Cheng, X.X.; Xue, J.Z.; Xue, S.A. Emerging Strategies in TCR-Engineered T Cells. Front. Immunol. 2022, 13, 850358. [Google Scholar] [CrossRef]
  7. Yang, Y.H.; Liu, J.W.; Lu, C.; Wei, J.F. CAR-T Cell Therapy for Breast Cancer: From Basic Research to Clinical Application. Int. J. Biol. Sci. 2022, 18, 2609–2626. [Google Scholar] [CrossRef]
  8. Kirtane, K.; Elmariah, H.; Chung, C.H.; Abate-Daga, D. Adoptive Cellular Therapy in Solid Tumor Malignancies: Review of the Literature and Challenges Ahead. J. Immunother. Cancer 2021, 9, e002723. [Google Scholar] [CrossRef]
  9. Akatsuka, Y. TCR-Like CAR-T Cells Targeting MHC-Bound Minor Histocompatibility Antigens. Front. Immunol. 2020, 11, e002723. [Google Scholar] [CrossRef]
  10. Teppert, K.; Wang, X.; Anders, K.; Evaristo, C.; Lock, D.; Künkele, A. Joining Forces for Cancer Treatment: From “TCR versus CAR” to “TCR and CAR”. Int. J. Mol. Sci. 2022, 23, 14563. [Google Scholar] [CrossRef]
  11. Alnefaie, A.; Albogami, S.; Asiri, Y.; Ahmad, T.; Alotaibi, S.S.; Al-Sanea, M.M.; Althobaiti, H. Chimeric Antigen Receptor T-Cells: An Overview of Concepts, Applications, Limitations, and Proposed Solutions. Front. Bioeng. Biotechnol. 2022, 10, 797440. [Google Scholar] [CrossRef] [PubMed]
  12. Martinez, M.; Moon, E.K. CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment. Front. Immunol. 2019, 10, 128. [Google Scholar] [CrossRef] [PubMed]
  13. Flugel, C.L.; Majzner, R.G.; Krenciute, G.; Dotti, G.; Riddell, S.R.; Wagner, D.L.; Abou-el-Enein, M. Overcoming On-Target, off-Tumour Toxicity of CAR T Cell Therapy for Solid Tumours. Nat. Rev. Clin. Oncol. 2023, 20, 49–62. [Google Scholar] [CrossRef] [PubMed]
  14. Brocker, T.; Karjalainen, K. Signals through T Cell Receptor-ζ Chain Alone Are Insufficient to Prime Resting T Lymphocytes. J. Exp. Med. 1995, 181, 1653–1659. [Google Scholar] [CrossRef] [PubMed]
  15. Kershaw, M.H.; Westwood, J.A.; Parker, L.L.; Wang, G.; Eshhar, Z.; Mavroukakis, S.A.; White, D.E.; Wunderlich, J.R.; Canevari, S.; Rogers-Freezer, L.; et al. A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer. Clin. Cancer Res. 2006, 12, 6106–6115. [Google Scholar] [CrossRef] [PubMed]
  16. Sterner, R.C.; Sterner, R.M. CAR-T Cell Therapy: Current Limitations and Potential Strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef] [PubMed]
  17. Guedan, S.; Calderon, H.; Posey, A.D.; Maus, M.V. Engineering and Design of Chimeric Antigen Receptors. Mol. Ther. Methods Clin. Dev. 2019, 12, 145–156. [Google Scholar] [CrossRef]
  18. Chmielewski, M.; Abken, H. TRUCKs: The Fourth Generation of CARs. Expert Opin. Biol. Ther. 2015, 15, 1145–1154. [Google Scholar] [CrossRef]
  19. Chmielewski, M.; Abken, H. TRUCKS, the Fourth-generation CAR T Cells: Current Developments and Clinical Translation. Adv. Cell Gene Ther. 2020, 3, e84. [Google Scholar] [CrossRef]
  20. Tomasik, J.; Jasiński, M.; Basak, G.W. Next Generations of CAR-T Cells—New Therapeutic Opportunities in Hematology? Front. Immunol. 2022, 13, 1034707. [Google Scholar] [CrossRef]
  21. Shafer, P.; Kelly, L.M.; Hoyos, V. Cancer Therapy with TCR-Engineered T Cells: Current Strategies, Challenges, and Prospects. Front. Immunol. 2022, 13, 835762. [Google Scholar] [CrossRef] [PubMed]
  22. He, Q.; Jiang, X.; Zhou, X.; Weng, J. Targeting Cancers through TCR-Peptide/MHC Interactions. J. Hematol. Oncol. 2019, 12, 139. [Google Scholar] [CrossRef] [PubMed]
  23. Zhou, S.; Liu, S.; Zhao, L.; Sun, H.X. A Comprehensive Survey of Genomic Mutations in Breast Cancer Reveals Recurrent Neoantigens as Potential Therapeutic Targets. Front. Oncol. 2022, 12, 786438. [Google Scholar] [CrossRef] [PubMed]
  24. Brett, J.O.; Spring, L.M.; Bardia, A.; Wander, S.A. ESR1 Mutation as an Emerging Clinical Biomarker in Metastatic Hormone Receptor-Positive Breast Cancer. Breast Cancer Res. 2021, 23, 85. [Google Scholar] [CrossRef] [PubMed]
  25. Mikhael, J.; Fowler, J.; Shah, N. Chimeric Antigen Receptor T-Cell Therapies: Barriers and Solutions to Access. JCO Oncol. Pract. 2022, 18, 800–807. [Google Scholar] [CrossRef] [PubMed]
  26. Dees, S.; Ganesan, R.; Singh, S.; Grewal, I.S. Emerging CAR-T Cell Therapy for the Treatment of Triple-Negative Breast Cancer. Mol. Cancer Ther. 2020, 19, 2409–2421. [Google Scholar] [CrossRef] [PubMed]
  27. Leko, V.; Rosenberg, S.A. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020, 38, 454–472. [Google Scholar] [CrossRef]
  28. Vigneron, N. Human Tumor Antigens and Cancer Immunotherapy. BioMed Res. Int. 2015, 2015, 948501. [Google Scholar] [CrossRef]
  29. Liu, X.; Jiang, S.; Fang, C.; Yang, S.; Olalere, D.; Pequignot, E.C.; Cogdill, A.P.; Li, N.; Ramones, M.; Granda, B.; et al. Affinity-Tuned ErbB2 or EGFR Chimeric Antigen Receptor T Cells Exhibit an Increased Therapeutic Index against Tumors in Mice. Cancer Res. 2015, 75, 3596–3607. [Google Scholar] [CrossRef]
  30. Jakobsen, M.K.; Gjerstorff, M.F. CAR T-Cell Cancer Therapy Targeting Surface Cancer/Testis Antigens. Front. Immunol. 2020, 11, 1568. [Google Scholar] [CrossRef]
  31. Li, Y.; Li, J.; Wang, Y.; Zhang, Y.; Chu, J.; Sun, C.; Fu, Z.; Huang, Y.; Zhang, H.; Yuan, H.; et al. Roles of Cancer/Testis Antigens (CTAs) in Breast Cancer. Cancer Lett. 2017, 399, 64–73. [Google Scholar] [CrossRef] [PubMed]
  32. Akers, S.N.; Odunsi, K.; Karpf, A.R. Regulation of Cancer Germline Antigen Gene Expression: Implications for Cancer Immunotherapy. Future Oncol. 2010, 6, 717–732. [Google Scholar] [CrossRef] [PubMed]
  33. Feola, S.; Chiaro, J.; Martins, B.; Cerullo, V. Uncovering the Tumor Antigen Landscape: What to Know about the Discovery Process. Cancers 2020, 12, 1660. [Google Scholar] [CrossRef] [PubMed]
  34. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising Targets for Cancer Therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef] [PubMed]
  35. Chandran, S.S.; Ma, J.; Klatt, M.G.; Dündar, F.; Bandlamudi, C.; Razavi, P.; Wen, H.Y.; Weigelt, B.; Zumbo, P.; Fu, S.N.; et al. Immunogenicity and Therapeutic Targeting of a Public Neoantigen Derived from Mutated PIK3CA. Nat. Med. 2022, 28, 946–957. [Google Scholar] [CrossRef] [PubMed]
  36. Tóth, G.; Szöllősi, J.; Abken, H.; Vereb, G.; Szöőr, Á. A Small Number of HER2 Redirected CAR T Cells Significantly Improves Immune Response of Adoptively Transferred Mouse Lymphocytes against Human Breast Cancer Xenografts. Int. J. Mol. Sci. 2020, 21, 1039. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, H.; Peng, Y. Current Biological, Pathological and Clinical Landscape of HER2-Low Breast Cancer. Cancers 2023, 15, 126. [Google Scholar] [CrossRef]
  38. Zhang, P.F.; Huang, Y.; Liang, X.; Li, D.; Jiang, L.; Yang, X.; Zhu, M.; Gou, H.F.; Gong, Y.L.; Wei, Y.Q.; et al. Enhancement of the Antitumor Effect of HER2-Directed CAR-T Cells through Blocking Epithelial-Mesenchymal Transition in Tumor Cells. FASEB J. 2020, 34, 11185–11199. [Google Scholar] [CrossRef]
  39. Li, H.; Yuan, W.; Bin, S.; Wu, G.; Li, P.; Liu, M.; Yang, J.; Li, X.; Yang, K.; Gu, H. Overcome Trastuzumab Resistance of Breast Cancer Using Anti-HER2 Chimeric Antigen Receptor T Cells and PD1 Blockade. Am. J. Cancer Res. 2020, 10, 688–703. [Google Scholar]
  40. Globerson-Levin, A.; Waks, T.; Eshhar, Z. Elimination of Progressive Mammary Cancer by Repeated Administrations of Chimeric Antigen Receptor-Modified T Cells. Mol. Ther. 2014, 22, 1029–1038. [Google Scholar] [CrossRef]
  41. Schönfeld, K.; Sahm, C.; Zhang, C.; Naundorf, S.; Brendel, C.; Odendahl, M.; Nowakowska, P.; Bönig, H.; Köhl, U.; Kloess, S.; et al. Selective Inhibition of Tumor Growth by Clonal NK Cells Expressing an ErbB2/HER2-Specific Chimeric Antigen Receptor. Mol. Ther. 2015, 23, 330–338. [Google Scholar] [CrossRef] [PubMed]
  42. Szöőr, Á.; Tóth, G.; Zsebik, B.; Szabó, V.; Eshhar, Z.; Abken, H.; Vereb, G. Trastuzumab Derived HER2-Specific CARs for the Treatment of Trastuzumab-Resistant Breast Cancer: CAR T Cells Penetrate and Eradicate Tumors That Are Not Accessible to Antibodies. Cancer Lett. 2020, 484, 1–8. [Google Scholar] [CrossRef] [PubMed]
  43. Priceman, S.J.; Tilakawardane, D.; Jeang, B.; Aguilar, B.; Murad, J.P.; Park, A.K.; Chang, W.-C.; Ostberg, J.R.; Neman, J.; Jandial, R.; et al. Regional Delivery of Chimeric Antigen Receptor–Engineered T Cells Effectively Targets HER2+ Breast Cancer Metastasis to the Brain. Clin. Cancer Res. 2018, 24, 95–105. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, M.; Shi, H.; Liu, C.; Liu, J.; Liu, X.; Sun, Y. Construction and Evaluation of a Novel Humanized HER2-Specific Chimeric Receptor. Breast Cancer Res. 2014, 16, R61. [Google Scholar] [CrossRef] [PubMed]
  45. Jia, L.; Yang, X.; Tian, W.; Gou, S.; Huang, W.; Zhao, W. Increased Expression of C-Met Is Associated with Chemotherapy-Resistant Breast Cancer and Poor Clinical Outcome. Med. Sci. Monit. 2018, 24, 8239–8249. [Google Scholar] [CrossRef] [PubMed]
  46. Zhao, X.; Qu, J.; Hui, Y.; Zhang, H.; Sun, Y.; Liu, X.; Zhao, X.; Zhao, Z.; Yang, Q.; Wang, F.; et al. Clinicopathological and Prognostic Significance of C-Met Overexpression in Breast Cancer. Oncotarget 2017, 8, 56758–56767. [Google Scholar] [CrossRef] [PubMed]
  47. 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] [PubMed]
  48. Ho-Yen, C.M.; Jones, J.L.; Kermorgant, S. The Clinical and Functional Significance of C-Met in Breast Cancer: A Review. Breast Cancer Res. 2015, 17, 52. [Google Scholar] [CrossRef]
  49. Nath, S.; Mukherjee, P. MUC1: A Multifaceted Oncoprotein with a Key Role in Cancer Progression. Trends Mol. Med. 2014, 20, 332–342. [Google Scholar] [CrossRef]
  50. Rakha, E.A.; Boyce, R.W.G.; El-Rehim, D.A.; Kurien, T.; Green, A.R.; Paish, E.C.; Robertson, J.F.R.; Ellis, I.O. Expression of Mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and MUC6) and Their Prognostic Significance in Human Breast Cancer. Mod. Pathol. 2005, 18, 1295–1304. [Google Scholar] [CrossRef]
  51. Zhou, R.; Yazdanifar, M.; Roy, L.D.; Whilding, L.M.; Gavrill, A.; Maher, J.; Mukherjee, P. CAR T Cells Targeting the Tumor MUC1 Glycoprotein Reduce Triple-Negative Breast Cancer Growth. Front. Immunol. 2019, 10, 1149. [Google Scholar] [CrossRef] [PubMed]
  52. Nalawade, S.A.; Shafer, P.; Bajgain, P.; McKenna, M.K.; Ali, A.; Kelly, L.; Joubert, J.; Gottschalk, S.; Watanabe, N.; Leen, A.; et al. Selectively Targeting Myeloid-Derived Suppressor Cells through TRAIL Receptor 2 to Enhance the Efficacy of CAR T Cell Therapy for Treatment of Breast Cancer. J. Immunother. Cancer 2021, 9, e003237. [Google Scholar] [CrossRef] [PubMed]
  53. Bajgain, P.; Tawinwung, S.; D’Elia, L.; Sukumaran, S.; Watanabe, N.; Hoyos, V.; Lulla, P.; Brenner, M.K.; Leen, A.M.; Vera, J.F. CAR T Cell Therapy for Breast Cancer: Harnessing the Tumor Milieu to Drive T Cell Activation. J. Immunother. Cancer 2018, 6, 34. [Google Scholar] [CrossRef] [PubMed]
  54. Tozbikian, G.; Brogi, E.; Kadota, K.; Catalano, J.; Akram, M.; Patil, S.; Ho, A.Y.; Reis-Filho, J.S.; Weigelt, B.; Norton, L.; et al. Mesothelin Expression in Triple Negative Breast Carcinomas Correlates Significantly with Basal-like Phenotype, Distant Metastases and Decreased Survival. PLoS ONE 2014, 9, e114900. [Google Scholar] [CrossRef] [PubMed]
  55. Li, Y.R.; Xian, R.R.; Ziober, A.; Conejo-Garcia, J.; Perales-Puchalt, A.; June, C.H.; Zhang, P.J.; Tchou, J. Mesothelin Expression Is Associated with Poor Outcomes in Breast Cancer. Breast Cancer Res. Treat. 2014, 147, 675–684. [Google Scholar] [CrossRef]
  56. Suzuki, T.; Yamagishi, Y.; Einama, T.; Koiwai, T.; Yamasaki, T.; Fukumura-Koga, M.; Ishibashi, Y.; Takihata, Y.; Shiraishi, T.; Miyata, Y.; et al. Membrane Mesothelin Expression Positivity Is Associated with Poor Clinical Outcome of Luminal-Type Breast Cancer. Oncol. Lett. 2020, 20, 193. [Google Scholar] [CrossRef]
  57. Tchou, J.; Wang, L.-C.; Selven, B.; Zhang, H.; Conejo-Garcia, J.; Borghaei, H.; Kalos, M.; Vondeheide, R.H.; Albelda, S.M.; June, C.H.; et al. Mesothelin, a Novel Immunotherapy Target for Triple Negative Breast Cancer. Breast Cancer Res. Treat. 2012, 133, 799–804. [Google Scholar] [CrossRef]
  58. Yang, M.; Guan, T.; Chen, C.F.; He, L.F.; Wu, H.M.; Zhang, R.D.; Li, Y.; Lin, Y.C.; Zeng, H.; Wu, J.D. Mesothelin-Targeted CAR-NK Cells Derived From Induced Pluripotent Stem Cells Have a High Efficacy in Killing Triple-Negative Breast Cancer Cells as Shown in Several Preclinical Models. J. Immunother. 2023, 46, 285–294. [Google Scholar] [CrossRef]
  59. Li, Y.; Xiao, F.; Zhang, A.; Zhang, D.; Nie, W.; Xu, T.; Han, B.; Seth, P.; Wang, H.; Yang, Y.; et al. Oncolytic Adenovirus Targeting TGF-β Enhances Anti-Tumor Responses of Mesothelin-Targeted Chimeric Antigen Receptor T Cell Therapy against Breast Cancer. Cell. Immunol. 2020, 348, 104041. [Google Scholar] [CrossRef]
  60. Soysal, S.D.; Muenst, S.; Barbie, T.; Fleming, T.; Gao, F.; Spizzo, G.; Oertli, D.; Viehl, C.T.; Obermann, E.C.; Gillanders, W.E. EpCAM Expression Varies Significantly and Is Differentially Associated with Prognosis in the Luminal B HER2+, Basal-like, and HER2 Intrinsic Subtypes of Breast Cancer. Br. J. Cancer 2013, 108, 1480–1487. [Google Scholar] [CrossRef]
  61. Gao, G.; Liao, W.; Shu, P.; Ma, Q.; He, X.; Zhang, B.; Qin, D.; Wang, Y. Targeting Sphingosine 1-Phosphate Receptor 3 Inhibits T-Cell Exhaustion and Regulates Recruitment of Proinflammatory Macrophages to Improve Antitumor Efficacy of CAR-T Cells against Solid Tumor. J. Immunother. Cancer 2023, 11, e006343. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, Y.; McCloskey, J.E.; Yang, H.; Puc, J.; Alcaina, Y.; Vedvyas, Y.; Gomez Gallegos, A.A.; Ortiz-Sánchez, E.; de Stanchina, E.; Min, I.M.; et al. Bispecific CAR T Cells against EpCAM and Inducible ICAM-1 Overcome Antigen Heterogeneity and Generate Superior Antitumor Responses. Cancer Immunol. Res. 2021, 9, 1158–1174. [Google Scholar] [CrossRef] [PubMed]
  63. 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] [PubMed]
  64. Irmer, B.; Efing, J.; Reitnauer, L.E.; Angenendt, A.; Heinrichs, S.; Schubert, A.; Schulz, M.; Binder, C.; Tio, J.; Hansen, U.; et al. Extracellular Vesicle-Associated Tyrosine Kinase-like Orphan Receptors ROR1 and ROR2 Promote Breast Cancer Progression. Cell Commun. Signal. 2023, 21, 171. [Google Scholar] [CrossRef] [PubMed]
  65. Nadanaka, S.; Tamura, J.I.; Kitagawa, H. Chondroitin Sulfates Control Invasiveness of the Basal-Like Breast Cancer Cell Line MDA-MB-231 Through ROR1. Front. Oncol. 2022, 12, 914838. [Google Scholar] [CrossRef] [PubMed]
  66. Chien, H.P.; Ueng, S.H.; Chen, S.C.; Chang, Y.S.; Lin, Y.C.; Lo, Y.F.; Chang, H.K.; Chuang, W.Y.; Huang, Y.T.; Cheung, Y.C.; et al. Expression of ROR1 Has Prognostic Significance in Triple Negative Breast Cancer. Virchows Arch. 2016, 468, 589–595. [Google Scholar] [CrossRef] [PubMed]
  67. 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]
  68. Stüber, T.; Monjezi, R.; Wallstabe, L.; Kühnemundt, J.; Nietzer, S.L.; Dandekar, G.; Wöckel, A.; Einsele, H.; Wischhusen, J.; Hudecek, M. Inhibition of TGF-β-Receptor Signaling Augments the Antitumor Function of ROR1-Specific CAR T-Cells against Triple-Negative Breast Cancer. J. Immunother. Cancer 2020, 8, e000676. [Google Scholar] [CrossRef]
  69. Wallstabe, L.; Göttlich, C.; Nelke, L.C.; Kühnemundt, J.; Schwarz, T.; Nerreter, T.; Einsele, H.; Walles, H.; Dandekar, G.; Nietzer, S.L.; et al. ROR1-CAR T Cells Are Effective against Lung and Breast Cancer in Advanced Microphysiologic 3D Tumor Models. JCI Insight 2019, 4, e126345. [Google Scholar] [CrossRef]
  70. Shao, Y.; Sun, X.; He, Y.; Liu, C.; Liu, H. Elevated Levels of Serum Tumor Markers CEA and CA15-3 Are Prognostic Parameters for Different Molecular Subtypes of Breast Cancer. PLoS ONE 2015, 10, e0133830. [Google Scholar] [CrossRef]
  71. Guadagni, F.; Ferroni, P.; Carlini, S.; Mariotti, S.; Spila, A.; Aloe, S.; D’Alessandro, R.; Carone, M.D.; Cicchetti, A.; Ricciotti, A.; et al. A Re-Evaluation of Carcinoembryonic Antigen (CEA) as a Serum Marker for Breast Cancer: A Prospective Longitudinal Study. Clin. Cancer Res. 2001, 7, 2357–2362. [Google Scholar] [PubMed]
  72. De Kruijf, E.M.; Sajet, A.; Van Nes, J.G.H.; Putter, H.; Smit, V.T.; Eagle, R.A.; Jafferji, I.; Trowsdale, J.; Liefers, G.J.; Van De Velde, C.J.H.; et al. NKG2D Ligand Tumor Expression and Association with Clinical Outcome in Early Breast Cancer Patients: An Observational Study. BMC Cancer 2012, 12, 24. [Google Scholar] [CrossRef] [PubMed]
  73. Han, Y.; Xie, W.; Song, D.G.; Powell, D.J. 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]
  74. Hsu, N.C.; Nien, P.Y.; Yokoyama, K.K.; Chu, P.Y.; Hou, M.F. High Chondroitin Sulfate Proteoglycan 4 Expression Correlates with Poor Outcome in Patients with Breast Cancer. Biochem. Biophys. Res. Commun. 2013, 441, 514–518. [Google Scholar] [CrossRef] [PubMed]
  75. Geldres, C.; Savoldo, B.; Hoyos, V.; Caruana, I.; Zhang, M.; Yvon, E.; Del Vecchio, M.; Creighton, C.J.; Ittmann, M.; Ferrone, S.; et al. T Lymphocytes Redirected against the Chondroitin Sulfate Proteoglycan-4 Control the Growth of Multiple Solid Tumors Both in Vitro and in Vivo. Clin. Cancer Res. 2014, 20, 962–971. [Google Scholar] [CrossRef] [PubMed]
  76. Beard, R.E.; Zheng, Z.; Lagisetty, K.H.; Burns, W.R.; Tran, E.; Hewitt, S.M.; Abate-Daga, D.; Rosati, S.F.; Fine, H.A.; Ferrone, S.; et al. Multiple Chimeric Antigen Receptors Successfully Target Chondroitin Sulfate Proteoglycan 4 in Several Different Cancer Histologies and Cancer Stem Cells. J. Immunother. Cancer 2014, 2, 25. [Google Scholar] [CrossRef]
  77. O’Shannessy, D.J.; Somers, E.B.; Maltzman, J.; Smale, R.; Fu, Y.S. Folate Receptor Alpha (FRA) Expression in Breast Cancer: Identification of a New Molecular Subtype and Association with Triple Negative Disease. Springerplus 2012, 1, 22. [Google Scholar] [CrossRef]
  78. Hartmann, L.C.; Keeney, G.L.; Lingle, W.L.; Christianson, T.J.H.; Varghese, B.; Hillman, D.; Oberg, A.L.; Low, P.S. Folate Receptor Overexpression Is Associated with Poor Outcome in Breast Cancer. Int. J. Cancer 2007, 121, 938–942. [Google Scholar] [CrossRef]
  79. Song, D.; Ye, Q.; Poussin, M.; Chacon, J.A.; Figini, M.; Powell, D.J., Jr. Effective Adoptive Immunotherapy of Triple-Negative Breast Cancer by Folate Receptor-Alpha Redirected CAR T Cells Is Influenced by Surface Antigen Expression Level. J. Hematol. Oncol. 2016, 9, 56. [Google Scholar] [CrossRef]
  80. Chuangchot, N.; Jamjuntra, P.; Yangngam, S.; Luangwattananun, P.; Thongchot, S.; Junking, M.; Thuwajit, P.; Yenchitsomanus, P.T.; Thuwajit, C. Enhancement of PD-L1-Attenuated CAR-T Cell Function through Breast Cancer-Associated Fibroblasts-Derived IL-6 Signaling via STAT3/AKT Pathways. Breast Cancer Res. 2023, 25, 86. [Google Scholar] [CrossRef]
  81. Battula, V.L.; Shi, Y.; Evans, K.W.; Wang, R.Y.; Spaeth, E.L.; Jacamo, R.O.; Guerra, R.; Sahin, A.A.; Marini, F.C.; Hortobagyi, G.; et al. Ganglioside GD2 Identifies Breast Cancer Stem Cells and Promotes Tumorigenesis. J. Clin. Investig. 2012, 122, 2066–2078. [Google Scholar] [CrossRef] [PubMed]
  82. Shao, C.; Anand, V.; Andreeff, M.; Battula, V.L. Ganglioside GD2: A Novel Therapeutic Target in Triple-Negative Breast Cancer. Ann. N. Y. Acad. Sci. 2022, 1508, 35–53. [Google Scholar] [CrossRef] [PubMed]
  83. Zhong, E.; Brogi, E.; D’Alfonso, T.M.; Wen, H.; Frosina, D.; Cheung, N.K.; Jungbluth, A.A.; Ross, D.S. Expression Analysis of GD2 by Immunohistochemistry in Invasive Breast Carcinoma: Clinical and Pathologic Correlation. Appl. Immunohistochem. Mol. Morphol. 2022, 30, 113–118. [Google Scholar] [CrossRef] [PubMed]
  84. Seitz, C.M.; Schroeder, S.; Knopf, P.; Krahl, A.C.; Hau, J.; Schleicher, S.; Martella, M.; Quintanilla-Martinez, L.; Kneilling, M.; Pichler, B.; et al. GD2-Targeted Chimeric Antigen Receptor T Cells Prevent Metastasis Formation by Elimination of Breast Cancer Stem-like Cells. Oncoimmunology 2020, 9, 1683345. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, Y.; Zhou, Y.; Huang, K.H.; Li, Y.; Fang, X.; An, L.; Wang, F.; Chen, Q.; Zhang, Y.; Shi, A.; et al. EGFR-Specific CAR-T Cells Trigger Cell Lysis in EGFR-Positive TNBC. Aging 2019, 11, 11054–11072. [Google Scholar] [CrossRef] [PubMed]
  86. Xia, L.; Zheng, Z.Z.; Liu, J.Y.; Chen, Y.J.; Ding, J.C.; Xia, N.S.; Luo, W.X.; Liu, W. EGFR-Targeted CAR-T Cells Are Potent and Specific in Suppressing Triple-Negative Breast Cancer Both in Vitro and in Vivo. Clin. Transl. Immunol. 2020, 9, e1135. [Google Scholar] [CrossRef] [PubMed]
  87. Li, R.H.; Huang, W.H.; Wu, J.D.; Du, C.W.; Zhang, G.J. EGFR Expression Is Associated with Cytoplasmic Staining of CXCR4 and Predicts Poor Prognosis in Triple-Negative Breast Carcinomas. Oncol. Lett. 2017, 13, 695–703. [Google Scholar] [CrossRef]
  88. Park, H.S.; Jang, M.H.; Kim, E.J.; Kim, H.J.; Lee, H.J.; Kim, Y.J.; Kim, J.H.; Kang, E.; Kim, S.W.; Kim, I.A.; et al. High EGFR Gene Copy Number Predicts Poor Outcome in Triple-Negative Breast Cancer. Mod. Pathol. 2014, 27, 1212–1222. [Google Scholar] [CrossRef]
  89. Chen, M.; Wu, C.; Fu, Z.; Liu, S. ICAM1 Promotes Bone Metastasis via Integrin-Mediated TGF-β/EMT Signaling in Triple-Negative Breast Cancer. Cancer Sci. 2022, 113, 3751–3765. [Google Scholar] [CrossRef]
  90. Guo, P.; Huang, J.; Wang, L.; Jia, D.; Yang, J.; Dillon, D.A.; Zurakowski, D.; Mao, H.; Moses, M.A.; Auguste, D.T.; et al. ICAM-1 as a Molecular Target for Triple Negative Breast Cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 14710–14715. [Google Scholar] [CrossRef]
  91. Wei, H.; Wang, Z.; Kuang, Y.; Wu, Z.; Zhao, S.; Zhang, Z.; Li, H.; Zheng, M.; Zhang, N.; Long, C.; et al. Intercellular Adhesion Molecule-1 as Target for CAR-T-Cell Therapy of Triple-Negative Breast Cancer. Front. Immunol. 2020, 11, 573823. [Google Scholar] [CrossRef] [PubMed]
  92. Yang, P.; Yu, F.; Yao, Z.; Ding, X.; Xu, H.; Zhang, J. CD24 Is a Novel Target of Chimeric Antigen Receptor T Cells for the Treatment of Triple Negative Breast Cancer. Cancer Immunol. Immunother. 2023, 72, 3191–3202. [Google Scholar] [CrossRef] [PubMed]
  93. Baba, S.A.; Sun, Q.; Mugisha, S.; Labhsetwar, S.; Klemke, R.; Desgrosellier, J.S. Breast Cancer Stem Cells Tolerate Chromosomal Instability during Tumor Progression via C-Jun/AXL Stress Signaling. Heliyon 2023, 9, e20182. [Google Scholar] [CrossRef] [PubMed]
  94. Ji, J.; Ding, Y.; Kong, Y.; Fang, M.; Yu, X.; Lai, X.; Gu, Q. Triple-negative Breast Cancer Cells That Survive Ionizing Radiation Exhibit an Axl-dependent Aggressive Radioresistant Phenotype. Exp. Ther. Med. 2023, 26, 448. [Google Scholar] [CrossRef] [PubMed]
  95. Gjerdrum, C.; Tiron, C.; Høiby, T.; Stefansson, I.; Haugen, H.; Sandal, T.; Collett, K.; Li, S.; McCormack, E.; Gjertsen, B.T.; et al. Axl Is an Essential Epithelial-to-Mesenchymal Transition-Induced Regulator of Breast Cancer Metastasis and Patient Survival. Proc. Natl. Acad. Sci. USA 2010, 107, 1124–1129. [Google Scholar] [CrossRef] [PubMed]
  96. Sun, X.; Chen, H.; You, S.; Tian, Z.; Wang, Z.; Liu, F.; Hu, W.; Zhang, H.; Zhang, G.; Zhao, H.; et al. AXL Upregulates C-Myc Expression through AKT and ERK Signaling Pathways in Breast Cancers. Mol. Clin. Oncol. 2023, 18, 22. [Google Scholar] [CrossRef] [PubMed]
  97. Wei, J.; Sun, H.; Zhang, A.; Wu, X.; Li, Y.; Liu, J.; Duan, Y.; Xiao, F.; Wang, H.; Lv, M.; et al. A Novel AXL Chimeric Antigen Receptor Endows T Cells with Anti-Tumor Effects against Triple Negative Breast Cancers. Cell. Immunol. 2018, 331, 49–58. [Google Scholar] [CrossRef]
  98. Zhao, Z.; Li, Y.; Liu, W.; Li, X. Engineered IL-7 Receptor Enhances the Therapeutic Effect of AXL-CAR-T Cells on Triple-Negative Breast Cancer. BioMed Res. Int. 2020, 2020, 4795171. [Google Scholar] [CrossRef]
  99. Tessari, A.; Pilla, L.; Silvia, D.; Duca, M.; Paolini, B.; Carcangiu, M.L.; Mariani, L.; de Braud, F.G.; Cresta, S. Expression of NY-ESO-1, MAGE-A3, PRAME and WT1 in Different Subgroups of Breast Cancer: An Indication to Immunotherapy? Breast 2018, 42, 68–73. [Google Scholar] [CrossRef]
  100. Raghavendra, A.; Kalita-de Croft, P.; Vargas, A.C.; Smart, C.E.; Simpson, P.T.; Saunus, J.M.; Lakhani, S.R. Expression of MAGE-A and NY-ESO-1 Cancer/Testis Antigens Is Enriched in Triple-Negative Invasive Breast Cancers. Histopathology 2018, 73, 68–80. [Google Scholar] [CrossRef]
  101. Liu, X.; Xu, Y.; Xiong, W.; Yin, B.; Huang, Y.; Chu, J.; Xing, C.; Qian, C.; Du, Y.; Duan, T.; et al. Development of a TCR-like Antibody and Chimeric Antigen Receptor against NY-ESO-1/HLA-A2 for Cancer Immunotherapy. J. Immunother. Cancer 2022, 10, e004035. [Google Scholar] [CrossRef] [PubMed]
  102. Ademuyiwa, F.O.; Bshara, W.; Attwood, K.; Morrison, C.; Edge, S.B.; Ambrosone, C.B.; O’Connor, T.L.; Levine, E.G.; Miliotto, A.; Ritter, E.; et al. NY-ESO-1 Cancer Testis Antigen Demonstrates High Immunogenicity in Triple Negative Breast Cancer. PLoS ONE 2012, 7, e38783. [Google Scholar] [CrossRef]
  103. Oh, C.; Kim, H.-R.; Oh, S.; Ko, J.Y.; Kim, Y.; Kang, K.; Yang, Y.; Kim, J.; Park, J.H.; Roe, J.-S.; et al. Epigenetic Upregulation of MAGE-A Isoforms Promotes Breast Cancer Cell Aggressiveness. Cancers 2021, 13, 3176. [Google Scholar] [CrossRef] [PubMed]
  104. Chinnasamy, N.; Wargo, J.A.; Yu, Z.; Rao, M.; Frankel, T.L.; Riley, J.P.; Hong, J.J.; Parkhurst, M.R.; Feldman, S.A.; Schrump, D.S.; et al. A TCR Targeting the HLA-A*0201–Restricted Epitope of MAGE-A3 Recognizes Multiple Epitopes of the MAGE-A Antigen Superfamily in Several Types of Cancer. J. Immunol. 2011, 186, 685–696. [Google Scholar] [CrossRef] [PubMed]
  105. Matković, B.; Juretić, A.; Spagnoli, G.C.; Šeparović, V.; Gamulin, M.; Šeparović, R.; Šarić, N.; Bašić-Koretić, M.; Novosel, I.; Krušlin, B. Expression of MAGE-A and NY-ESO-1 Cancer/Testis Antigens in Medullary Breast Cancer: Retrospective Immunohistochemical Study. Croat. Med. J. 2011, 52, 171–177. [Google Scholar] [CrossRef] [PubMed]
  106. Bu, J.; Zhang, Y.; Wu, S.; Li, H.; Sun, L.; Liu, Y.; Zhu, X.; Qiao, X.; Ma, Q.; Liu, C.; et al. KK-LC-1 as a Therapeutic Target to Eliminate ALDH+ Stem Cells in Triple Negative Breast Cancer. Nat. Commun. 2023, 14, 2602. [Google Scholar] [CrossRef] [PubMed]
  107. Zhu, X.; Bu, J.; Zhu, T.; Jiang, Y. Targeting KK-LC-1 Inhibits Malignant Biological Behaviors of Triple-Negative Breast Cancer. J. Transl. Med. 2023, 21, 184. [Google Scholar] [CrossRef]
  108. Kondo, Y.; Fukuyama, T.; Yamamura, R.; Futawatari, N.; Ichiki, Y.; Tanaka, Y.; Nishi, Y.; Takahashi, Y.; Yamazaki, H.; Kobayashi, N.; et al. Detection of KK-LC-1 Protein, a Cancer/Testis Antigen, in Patients with Breast Cancer. Anticancer Res. 2018, 38, 5923–5928. [Google Scholar] [CrossRef]
  109. Keraite, I.; Alvarez-Garcia, V.; Garcia-Murillas, I.; Beaney, M.; Turner, N.C.; Bartos, C.; Oikonomidou, O.; Kersaudy-Kerhoas, M.; Leslie, N.R. PIK3CA Mutation Enrichment and Quantitation from Blood and Tissue. Sci. Rep. 2020, 10, 17082. [Google Scholar] [CrossRef]
  110. Martínez-Saéz, O.; Chic, N.; Pascual, T.; Adamo, B.; Vidal, M.; González-Farré, B.; Sanfeliu, E.; Schettini, F.; Conte, B.; Brasó-Maristany, F.; et al. Frequency and Spectrum of PIK3CA Somatic Mutations in Breast Cancer. Breast Cancer Res. 2020, 22, 45. [Google Scholar] [CrossRef]
  111. Reinhardt, K.; Stückrath, K.; Hartung, C.; Kaufhold, S.; Uleer, C.; Hanf, V.; Lantzsch, T.; Peschel, S.; John, J.; Pöhler, M.; et al. PIK3CA-Mutations in Breast Cancer. Breast Cancer Res. Treat. 2022, 196, 483–493. [Google Scholar] [CrossRef] [PubMed]
  112. Forbes, S.A.; Bindal, N.; Bamford, S.; Cole, C.; Kok, C.Y.; Beare, D.; Jia, M.; Shepherd, R.; Leung, K.; Menzies, A.; et al. COSMIC: Mining Complete Cancer Genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011, 39, 945–950. [Google Scholar] [CrossRef] [PubMed]
  113. Muller, P.A.J.; Caswell, P.T.; Doyle, B.; Iwanicki, M.P.; Tan, E.H.; Karim, S.; Lukashchuk, N.; Gillespie, D.A.; Ludwig, R.L.; Gosselin, P.; et al. Mutant P53 Drives Invasion by Promoting Integrin Recycling. Cell 2009, 139, 1327–1341. [Google Scholar] [CrossRef] [PubMed]
  114. Capaci, V.; Bascetta, L.; Fantuz, M.; Beznoussenko, G.V.; Sommaggio, R.; Cancila, V.; Bisso, A.; Campaner, E.; Mironov, A.A.; Wiśniewski, J.R.; et al. Mutant P53 Induces Golgi Tubulo-Vesiculation Driving a Prometastatic Secretome. Nat. Commun. 2020, 11, 3945. [Google Scholar] [CrossRef] [PubMed]
  115. Matsuda, N.; Lim, B.; Wang, Y.; Krishnamurthy, S.; Woodward, W.; Alvarez, R.H.; Lucci, A.; Valero, V.; Reuben, J.M.; Meric-Bernstam, F.; et al. Identification of Frequent Somatic Mutations in Inflammatory Breast Cancer. Breast Cancer Res. Treat. 2017, 163, 263–272. [Google Scholar] [CrossRef] [PubMed]
  116. Kim, S.P.; Vale, N.R.; Zacharakis, N.; Krishna, S.; Yu, Z.; Gasmi, B.; Gartner, J.J.; Sindiri, S.; Malekzadeh, P.; Deniger, D.C.; et al. Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-Cell Receptor-Engineered T Cells Targeting Common P53 Neoantigens in Human Solid Tumors. Cancer Immunol. Res. 2022, 10, 932–946. [Google Scholar] [CrossRef] [PubMed]
  117. Walerych, D.; Napoli, M.; Collavin, L.; Del Sal, G. The Rebel Angel: Mutant P53 as the Driving Oncogene in Breast Cancer. Carcinogenesis 2012, 33, 2007–2017. [Google Scholar] [CrossRef]
  118. Chen, N.; Li, X.; Chintala, N.K.; Tano, Z.E.; Adusumilli, P.S. Driving CARs on the Uneven Road of Antigen Heterogeneity in Solid Tumors. Curr. Opin. Immunol. 2018, 51, 103–110. [Google Scholar] [CrossRef]
  119. Hou, Y.; Nitta, H.; Li, Z. HER2 Intratumoral Heterogeneity in Breast Cancer, an Evolving Concept. Cancers 2023, 15, 2664. [Google Scholar] [CrossRef]
  120. Anurathapan, U.; Chan, R.C.; Hindi, H.F.; Mucharla, R.; Bajgain, P.; Hayes, B.C.; Fisher, W.E.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; et al. Kinetics of Tumor Destruction by Chimeric Antigen Receptor-Modified T Cells. Mol. Ther. 2014, 22, 623–633. [Google Scholar] [CrossRef]
  121. Kailayangiri, S.; Altvater, B.; Lesch, S.; Balbach, S.; Göttlich, C.; Kühnemundt, J.; Mikesch, J.-H.; Schelhaas, S.; Jamitzky, S.; Meltzer, J.; et al. EZH2 Inhibition in Ewing Sarcoma Upregulates GD2 Expression for Targeting with Gene-Modified T Cells. Mol. Ther. 2019, 27, 933–946. [Google Scholar] [CrossRef] [PubMed]
  122. Driouk, L.; Gicobi, J.; Kamihara, Y.; Rutherford, K.; Dranoff, G.; Ritz, J.; Baumeister, S.H.C. Chimeric Antigen Receptor T Cells Targeting NKG2D-Ligands Show Robust Efficacy Against Acute Myeloid Leukemia and T-Cell Acute Lymphoblastic Leukemia. Blood 2019, 134, 1930. [Google Scholar] [CrossRef]
  123. Ruella, M.; Barrett, D.M.; Kenderian, S.S.; Shestova, O.; Hofmann, T.J.; Perazzelli, J.; Klichinsky, M.; Aikawa, V.; Nazimuddin, F.; Kozlowski, M.; et al. Dual CD19 and CD123 Targeting Prevents Antigen-Loss Relapses after CD19-Directed Immunotherapies. J. Clin. Investig. 2016, 126, 3814–3826. [Google Scholar] [CrossRef] [PubMed]
  124. Fousek, K.; Watanabe, J.; Joseph, S.K.; George, A.; An, X.; Byrd, T.T.; Morris, J.S.; Luong, A.; Martínez-Paniagua, M.A.; Sanber, K.; et al. CAR T-Cells That Target Acute B-Lineage Leukemia Irrespective of CD19 Expression. Leukemia 2021, 35, 75–89. [Google Scholar] [CrossRef]
  125. Wilkie, S.; Van Schalkwyk, M.C.I.; Hobbs, S.; Davies, D.M.; Van Der Stegen, S.J.C.; Pereira, A.C.P.; Burbridge, S.E.; Box, C.; Eccles, S.A.; Maher, J. Dual Targeting of ErbB2 and MUC1 in Breast Cancer Using Chimeric Antigen Receptors Engineered to Provide Complementary Signaling. J. Clin. Immunol. 2012, 32, 1059–1070. [Google Scholar] [CrossRef] [PubMed]
  126. Guo, Z.S.; Hong, J.A.; Irvine, K.R.; Chen, G.A.; Spiess, P.J.; Liu, Y.; Zeng, G.; Wunderlich, J.R.; Nguyen, D.M.; Restifo, N.P.; et al. De Novo Induction of a Cancer/Testis Antigen by 5-Aza-2′-Deoxycytidine Augments Adoptive Immunotherapy in a Murine Tumor Model. Cancer Res. 2006, 66, 1105–1113. [Google Scholar] [CrossRef] [PubMed]
  127. Tousley, A.M.; Rotiroti, M.C.; Labanieh, L.; Rysavy, L.W.; Kim, W.J.; Lareau, C.; Sotillo, E.; Weber, E.W.; Rietberg, S.P.; Dalton, G.N.; et al. Co-Opting Signalling Molecules Enables Logic-Gated Control of CAR T Cells. Nature 2023, 615, 507–516. [Google Scholar] [CrossRef]
  128. Soysal, S.D.; Tzankov, A.; Muenst, S.E. Role of the Tumor Microenvironment in Breast Cancer. Pathobiology 2015, 82, 142–152. [Google Scholar] [CrossRef]
  129. Li, J.J.; Tsang, J.Y.; Tse, G.M. Tumor Microenvironment in Breast Cancer—Updates on Therapeutic Implications and Pathologic Assessment. Cancers 2021, 13, 4233. [Google Scholar] [CrossRef]
  130. Wu, S.Z.; Al-Eryani, G.; Roden, D.L.; Junankar, S.; Harvey, K.; Andersson, A.; Thennavan, A.; Wang, C.; Torpy, J.R.; Bartonicek, N.; et al. A Single-Cell and Spatially Resolved Atlas of Human Breast Cancers. Nat. Genet. 2021, 53, 1334–1347. [Google Scholar] [CrossRef]
  131. Tan, Z.; Kan, C.; Sun, M.; Yang, F.; Wong, M.; Wang, S.; Zheng, H. Mapping Breast Cancer Microenvironment Through Single-Cell Omics. Front. Immunol. 2022, 13, 868813. [Google Scholar] [CrossRef] [PubMed]
  132. Xie, J.; Deng, W.; Deng, X.; Liang, J.Y.; Tang, Y.; Huang, J.; Tang, H.; Zou, Y.; Zhou, H.; Xie, X. Single-Cell Histone Chaperones Patterns Guide Intercellular Communication of Tumor Microenvironment That Contribute to Breast Cancer Metastases. Cancer Cell Int. 2023, 23, 311. [Google Scholar] [CrossRef] [PubMed]
  133. Ma, C.; Yang, C.; Peng, A.; Sun, T.; Ji, X.; Mi, J.; Wei, L.; Shen, S.; Feng, Q. Pan-Cancer Spatially Resolved Single-Cell Analysis Reveals the Crosstalk between Cancer-Associated Fibroblasts and Tumor Microenvironment. Mol. Cancer 2023, 22, 170. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, W.; Xu, M.; Xu, S.; Guan, Q.; Geng, S.; Wang, J.; Wei, W.; Xu, H.; Liu, Y.; Meng, Y.; et al. Single-Cell RNA Reveals a Tumorigenic Microenvironment in the Interface Zone of Human Breast Tumors. Breast Cancer Res. 2023, 25, 100. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, Y.; Zhen, F.; Sun, Y.; Han, B.; Wang, H.; Zhang, Y.; Zhang, H.; Hu, J. Single-Cell RNA Sequencing Reveals Small Extracellular Vesicles Derived from Malignant Cells That Contribute to Angiogenesis in Human Breast Cancers. J. Transl. Med. 2023, 21, 570. [Google Scholar] [CrossRef] [PubMed]
  136. Hou, P.; Luo, Y.; Wu, N. TCL1A+ B Cells Predict Prognosis in Triple-Negative Breast Cancer through Integrative Analysis of Single-Cell and Bulk Transcriptomic Data. Open Life Sci. 2023, 18, 20220707. [Google Scholar] [CrossRef] [PubMed]
  137. Watanabe, M.A.E.; Oda, J.M.M.; Amarante, M.K.; Cesar Voltarelli, J. Regulatory T Cells and Breast Cancer: Implications for Immunopathogenesis. Cancer Metastasis Rev. 2010, 29, 569–579. [Google Scholar] [CrossRef]
  138. Hashemi, V.; Maleki, L.A.; Esmaily, M.; Masjedi, A.; Ghalamfarsa, G.; Namdar, A.; Yousefi, M.; Yousefi, B.; Jadidi-Niaragh, F. Regulatory T Cells in Breast Cancer as a Potent Anti-Cancer Therapeutic Target. Int. Immunopharmacol. 2020, 78, 106087. [Google Scholar] [CrossRef]
  139. Kos, K.; De Visser, K.E. The Multifaceted Role of Regulatory T Cells in Breast Cancer. Annu. Rev. Cancer Biol. 2020, 5, 291–310. [Google Scholar] [CrossRef]
  140. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T Cells in Cancer Immunosuppression—Implications for Anticancer Therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
  141. Lucca, L.E.; Dominguez-Villar, M. Modulation of Regulatory T Cell Function and Stability by Co-Inhibitory Receptors. Nat. Rev. Immunol. 2020, 20, 680–693. [Google Scholar] [CrossRef] [PubMed]
  142. Bohling, S.D.; Allison, K.H. Immunosuppressive Regulatory T Cells Are Associated with Aggressive Breast Cancer Phenotypes: A Potential Therapeutic Target. Mod. Pathol. 2008, 21, 1527–1532. [Google Scholar] [CrossRef] [PubMed]
  143. Stenström, J.; Hedenfalk, I.; Hagerling, C. Regulatory T Lymphocyte Infiltration in Metastatic Breast Cancer—An Independent Prognostic Factor That Changes with Tumor Progression. Breast Cancer Res. 2021, 23, 27. [Google Scholar] [CrossRef] [PubMed]
  144. Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity 2016, 45, 1122–1134. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, B.J.; Zhao, J.W.; Zhang, D.H.; Zheng, A.H.; Wu, G.Q. Immunotherapy of Cancer by Targeting Regulatory T Cells. Int. Immunopharmacol. 2022, 104, 108469. [Google Scholar] [CrossRef]
  146. Tanaka, A.; Sakaguchi, S. Regulatory T Cells in Cancer Immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef]
  147. Taylor, N.A.; Vick, S.C.; Iglesia, M.D.; Brickey, W.J.; Midkiff, B.R.; McKinnon, K.P.; Reisdorf, S.; Anders, C.K.; Carey, L.A.; Parker, J.S.; et al. Treg Depletion Potentiates Checkpoint Inhibition in Claudin-Low Breast Cancer. J. Clin. Investig. 2017, 127, 3472–3483. [Google Scholar] [CrossRef]
  148. Terranova-Barberio, M.; Thomas, S.; Ali, N.; Pawlowska, N.; Park, J.; Krings, G.; Rosenblum, M.D.; Budillon, A.; Munster, P.N. HDAC Inhibition Potentiates Immunotherapy in Triple Negative Breast Cancer. Oncotarget 2017, 8, 114156–114172. [Google Scholar] [CrossRef]
  149. Ge, Y.; Domschke, C.; Stoiber, N.; Schott, S.; Heil, J.; Rom, J.; Blumenstein, M.; Thum, J.; Sohn, C.; Schneeweiss, A.; et al. Metronomic Cyclophosphamide Treatment in Metastasized Breast Cancer Patients: Immunological Effects and Clinical Outcome. Cancer Immunol. Immunother. 2012, 61, 353–362. [Google Scholar] [CrossRef]
  150. Generali, D.; Bates, G.; Berruti, A.; Brizzi, M.P.; Campo, L.; Bonardi, S.; Bersiga, A.; Allevi, G.; Milani, M.; Aguggini, S.; et al. Immunomodulation of FOXP3+ Regulatory T Cells by the Aromatase Inhibitor Letrozole in Breast Cancer Patients. Clin. Cancer Res. 2009, 15, 1046–1051. [Google Scholar] [CrossRef]
  151. Rech, A.J.; Mick, R.; Recio, A.; DeMichele, A.; Tweed, C.K.; Fox, K.R.; Domchek, S.M.; Vonderheide, R.H. Phase I Study of Anti-CD25 Mab Daclizumab to Deplete Regulatory T Cells Prior to Telomerase/Survivin Peptide Vaccination in Patients (Pts) with Metastatic Breast Cancer (MBC) (Meeting Abstract). J. Clin. Oncol. 2010, 28, 2508. [Google Scholar] [CrossRef]
  152. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [PubMed]
  153. Law, A.M.K.; Valdes-Mora, F.; Gallego-Ortega, D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells 2020, 9, 561. [Google Scholar] [CrossRef] [PubMed]
  154. Cassetta, L.; Baekkevold, E.S.; Brandau, S.; Bujko, A.; Cassatella, M.A.; Dorhoi, A.; Krieg, C.; Lin, A.; Loré, K.; Marini, O.; et al. Deciphering Myeloid-Derived Suppressor Cells: Isolation and Markers in Humans, Mice and Non-Human Primates. Cancer Immunol. Immunother. 2019, 68, 687–697. [Google Scholar] [CrossRef] [PubMed]
  155. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for Myeloid-Derived Suppressor Cell Nomenclature and Characterization Standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed]
  156. De Cicco, P.; Ercolano, G.; Ianaro, A. The New Era of Cancer Immunotherapy: Targeting Myeloid-Derived Suppressor Cells to Overcome Immune Evasion. Front. Immunol. 2020, 11, 1680. [Google Scholar] [CrossRef] [PubMed]
  157. Bergenfelz, C.; Larsson, A.M.; Von Stedingk, K.; Gruvberger-Saal, S.; Aaltonen, K.; Jansson, S.; Jernström, H.; Janols, H.; Wullt, M.; Bredberg, A.; et al. Systemic Monocytic-MDSCs Are Generated from Monocytes and Correlate with Disease Progression in Breast Cancer Patients. PLoS ONE 2015, 10, e0127028. [Google Scholar] [CrossRef]
  158. Bergenfelz, C.; Roxå, A.; Mehmeti, M.; Leandersson, K.; Larsson, A.M. Clinical Relevance of Systemic Monocytic-MDSCs in Patients with Metastatic Breast Cancer. Cancer Immunol. Immunother. 2020, 69, 435–448. [Google Scholar] [CrossRef]
  159. Di, S.; Zhou, M.; Pan, Z.; Sun, R.; Chen, M.; Jiang, H.; Shi, B.; Luo, H.; Li, Z. Combined Adjuvant of Poly I:C Improves Antitumor Effects of CAR-T Cells. Front. Oncol. 2019, 9, 241. [Google Scholar] [CrossRef]
  160. Bauer, R.; Udonta, F.; Wroblewski, M.; Ben-Batalla, I.; Santos, I.M.; Taverna, F.; Kuhlencord, M.; Gensch, V.; Päsler, S.; Vinckier, S.; et al. Blockade of Myeloid-Derived Suppressor Cell Expansion with All-Trans Retinoic Acid Increases the Efficacy of Antiangiogenic Therapy. Cancer Res. 2018, 78, 3220–3232. [Google Scholar] [CrossRef]
  161. Christmas, B.J.; Rafie, C.I.; Hopkins, A.C.; Scott, B.A.; Ma, H.S.; Cruz, K.A.; Woolman, S.; Armstrong, T.D.; Connolly, R.M.; Azad, N.A.; et al. Entinostat Converts Immune-Resistant Breast and Pancreatic Cancers into Checkpoint-Responsive Tumors by Reprogramming Tumor-Infiltrating MDSCs. Cancer Immunol. Res. 2018, 6, 1561–1577. [Google Scholar] [CrossRef] [PubMed]
  162. Sun, R.; Luo, H.; Su, J.; Di, S.; Zhou, M.; Shi, B.; Sun, Y.; Du, G.; Zhang, H.; Jiang, H.; et al. Olaparib Suppresses MDSC Recruitment via SDF1α/CXCR4 Axis to Improve the Anti-Tumor Efficacy of CAR-T Cells on Breast Cancer in Mice. Mol. Ther. 2021, 29, 60–74. [Google Scholar] [CrossRef] [PubMed]
  163. Condamine, T.; Kumar, V.; Ramachandran, I.R.; Youn, J.I.; Celis, E.; Finnberg, N.; El-Deiry, W.S.; Winograd, R.; Vonderheide, R.H.; English, N.R.; et al. ER Stress Regulates Myeloid-Derived Suppressor Cell Fate through TRAIL-R-Mediated Apoptosis. J. Clin. Investig. 2014, 124, 2626–2639. [Google Scholar] [CrossRef] [PubMed]
  164. Dominguez, G.A.; Condamine, T.; Mony, S.; Hashimoto, A.; Wang, F.; Liu, Q.; Forero, A.; Bendell, J.; Witt, R.; Hockstein, N.; et al. Selective Targeting of Myeloid-Derived Suppressor Cells in Cancer Patients Using DS-8273a, an Agonistic TRAIL-R2 Antibody. Clin. Cancer Res. 2017, 23, 2942–2950. [Google Scholar] [CrossRef] [PubMed]
  165. Cha, Y.J.; Koo, J.S. Role of Tumor-Associated Myeloid Cells in Breast Cancer. Cells 2020, 9, 1785. [Google Scholar] [CrossRef] [PubMed]
  166. Cendrowicz, E.; Sas, Z.; Bremer, E.; Rygiel, T.P. The Role of Macrophages in Cancer Development and Therapy. Cancers 2021, 13, 1946. [Google Scholar] [CrossRef] [PubMed]
  167. Bozorgi, A.; Bozorgi, M.; Khazaei, M. Immunotherapy and Immunoengineering for Breast Cancer; a Comprehensive Insight into CAR-T Cell Therapy Advancements, Challenges and Prospects. Cell. Oncol. 2022, 45, 755–777. [Google Scholar] [CrossRef]
  168. Zhang, Y.; Cheng, S.; Zhang, M.; Zhen, L.; Pang, D.; Zhang, Q.; Li, Z. High-Infiltration of Tumor-Associated Macrophages Predicts Unfavorable Clinical Outcome for Node-Negative Breast Cancer. PLoS ONE 2013, 8, e76147. [Google Scholar] [CrossRef]
  169. Mehta, A.K.; Kadel, S.; Townsend, M.G.; Oliwa, M.; Guerriero, J.L. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front. Immunol. 2021, 12, 643771. [Google Scholar] [CrossRef]
  170. Ye, X.; Li, Y.; Stawicki, S.; Couto, S.; Eastham-Anderson, J.; Kallop, D.; Weimer, R.; Wu, Y.; Pei, L. An Anti-Axl Monoclonal Antibody Attenuates Xenograft Tumor Growth and Enhances the Effect of Multiple Anticancer Therapies. Oncogene 2010, 29, 5254–5264. [Google Scholar] [CrossRef]
  171. Cha, J.H.; Chan, L.C.; Wang, Y.N.; Chu, Y.Y.; Wang, C.H.; Lee, H.H.; Xia, W.; Shyu, W.C.; Liu, S.P.; Yao, J.; et al. Ephrin Receptor A10 Monoclonal Antibodies and the Derived Chimeric Antigen Receptor T Cells Exert an Antitumor Response in Mouse Models of Triple-Negative Breast Cancer. J. Biol. Chem. 2022, 298, 101817. [Google Scholar] [CrossRef] [PubMed]
  172. Lin, E.Y.; Nguyen, A.V.; Russell, R.G.; Pollard, J.W. Colony-Stimulating Factor 1 Promotes Progression of Mammary Tumors to Malignancy. J. Exp. Med. 2001, 193, 727–739. [Google Scholar] [CrossRef] [PubMed]
  173. Goswami, S.; Sahai, E.; Wyckoff, J.B.; Cammer, M.; Cox, D.; Pixley, F.J.; Stanley, E.R.; Segall, J.E.; Condeelis, J.S. Macrophages Promote the Invasion of Breast Carcinoma Cells via a Colony-Stimulating Factor-1/Epidermal Growth Factor Paracrine Loop. Cancer Res. 2005, 65, 5278–5283. [Google Scholar] [CrossRef] [PubMed]
  174. Strachan, D.C.; Ruffell, B.; Oei, Y.; Bissell, M.J.; Coussens, L.M.; Pryer, N.; Daniel, D. CSF1R Inhibition Delays Cervical and Mammary Tumor Growth in Murine Models by Attenuating the Turnover of Tumor-Associated Macrophages and Enhancing Infiltration by CD8+ T Cells. Oncoimmunology 2013, 2, e26968. [Google Scholar] [CrossRef]
  175. Gomez-Roca, C.A.; Italiano, A.; Le Tourneau, C.; Cassier, P.A.; Toulmonde, M.; D’Angelo, S.P.; Campone, M.; Weber, K.L.; Loirat, D.; Cannarile, M.A.; et al. Phase i Study of Emactuzumab Single Agent or in Combination with Paclitaxel in Patients with Advanced/Metastatic Solid Tumors Reveals Depletion of Immunosuppressive M2-like Macrophages. Ann. Oncol. 2019, 30, 1381–1392. [Google Scholar] [CrossRef]
  176. Guerriero, J.L.; Sotayo, A.; Ponichtera, H.E.; Castrillon, J.A.; Pourzia, A.L.; Schad, S.; Johnson, S.F.; Carrasco, R.D.; Lazo, S.; Bronson, R.T.; et al. Class IIa HDAC Inhibition Reduces Breast Tumours and Metastases through Anti-Tumour Macrophages. Nature 2017, 543, 428–432. [Google Scholar] [CrossRef]
  177. Georgoudaki, A.M.; Prokopec, K.E.; Boura, V.F.; Hellqvist, E.; Sohn, S.; Östling, J.; Dahan, R.; Harris, R.A.; Rantalainen, M.; Klevebring, D.; et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016, 15, 2000–2011. [Google Scholar] [CrossRef]
  178. Insua-Rodríguez, J.; Oskarsson, T. The Extracellular Matrix in Breast Cancer. Adv. Drug Deliv. Rev. 2016, 97, 41–55. [Google Scholar] [CrossRef]
  179. Oskarsson, T. Extracellular Matrix Components in Breast Cancer Progression and Metastasis. Breast 2013, 22, S66–S72. [Google Scholar] [CrossRef]
  180. Papanicolaou, M.; Parker, A.L.; Yam, M.; Filipe, E.C.; Wu, S.Z.; Chitty, J.L.; Wyllie, K.; Tran, E.; Mok, E.; Nadalini, A.; et al. Temporal Profiling of the Breast Tumour Microenvironment Reveals Collagen XII as a Driver of Metastasis. Nat. Commun. 2022, 13, 4587. [Google Scholar] [CrossRef]
  181. Acerbi, I.; Cassereau, L.; Dean, I.; Shi, Q.; Au, A.; Park, C.; Chen, Y.Y.; Liphardt, J.; Hwang, E.S.; Weaver, V.M. Human Breast Cancer Invasion and Aggression Correlates with ECM Stiffening and Immune Cell Infiltration. Integr. Biol. 2015, 7, 1120–1134. [Google Scholar] [CrossRef] [PubMed]
  182. Jena, M.K.; Janjanam, J. Role of Extracellular Matrix in Breast Cancer Development: A Brief Update. F1000Research 2018, 7, 274. [Google Scholar] [CrossRef] [PubMed]
  183. Sun, X.; Wu, B.; Chiang, H.-C.; Deng, H.; Zhang, X.; Xiong, W.; Liu, J.; Rozeboom, A.M.; Harris, B.T.; Blommaert, E.; et al. Tumour DDR1 Promotes Collagen Fibre Alignment to Instigate Immune Exclusion. Nature 2021, 599, 673–678. [Google Scholar] [CrossRef] [PubMed]
  184. Zhong, X.; Zhang, W.; Sun, T. DDR1 Promotes Breast Tumor Growth by Suppressing Antitumor Immunity. Oncol. Rep. 2019, 42, 2844–2854. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, W.; Liu, L.; Su, H.F.; Liu, Q.; Shen, J.; Dai, H.; Zheng, W.; Lu, Y.; Zhang, W.; Bei, Y.; et al. Chimeric Antigen Receptor Macrophage Therapy for Breast Tumours Mediated by Targeting the Tumour Extracellular Matrix. Br. J. Cancer 2019, 121, 837–845. [Google Scholar] [CrossRef]
  186. Hu, D.; Li, Z.; Zheng, B.; Lin, X.; Pan, Y.; Gong, P.; Zhuo, W.; Hu, Y.; Chen, C.; Chen, L.; et al. Cancer-Associated Fibroblasts in Breast Cancer: Challenges and Opportunities. Cancer Commun. 2022, 42, 401–434. [Google Scholar] [CrossRef]
  187. Bughda, R.; Dimou, P.; D’Souza, R.R.; Klampatsa, A. Fibroblast Activation Protein (FAP)-Targeted CAR-T Cells: Launching an Attack on Tumor Stroma. ImmunoTargets Ther. 2021, 10, 313–323. [Google Scholar] [CrossRef]
  188. Elwakeel, E.; Weigert, A. Breast Cancer Cafs: Spectrum of Phenotypes and Promising Targeting Avenues. Int. J. Mol. Sci. 2021, 22, 11636. [Google Scholar] [CrossRef]
  189. Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal Fibroblasts Present in Invasive Human Breast Carcinomas Promote Tumor Growth and Angiogenesis through Elevated SDF-1/CXCL12 Secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef]
  190. Chen, B.; Sang, Y.; Song, X.; Zhang, D.; Wang, L.; Zhao, W.; Liang, Y.; Zhang, N.; Yang, Q. Exosomal MiR-500a-5p Derived from Cancer-Associated Fibroblasts Promotes Breast Cancer Cell Proliferation and Metastasis through Targeting USP28. Theranostics 2021, 11, 3932–3947. [Google Scholar] [CrossRef]
  191. Tran, E.; Chinnasamy, D.; Yu, Z.; Morgan, R.A.; Lee, C.C.R.; Restifo, N.P.; Rosenberg, S.A. Immune Targeting of Fibroblast Activation Protein Triggers Recognition of Multipotent Bone Marrow Stromal Cells and Cachexia. J. Exp. Med. 2013, 210, 1065–1068. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, L.-C.S.; Lo, A.; Scholler, J.; Sun, J.; Majumdar, R.S.; Kapoor, V.; Antzis, M.; Cotner, C.E.; Johnson, L.A.; Durham, A.C.; et al. Targeting Fibroblast Activation Protein in Tumor Stroma with Chimeric Antigen Receptor T Cells Can Inhibit Tumor Growth and Augment Host Immunity without Severe Toxicity. Cancer Immunol. Res. 2014, 2, 154–166. [Google Scholar] [CrossRef] [PubMed]
  193. Kakarla, S.; Chow, K.K.H.; Mata, M.; Shaffer, D.R.; Song, X.T.; Wu, M.F.; Liu, H.; Wang, L.L.; Rowley, D.R.; Pfizenmaier, K.; et al. Antitumor Effects of Chimeric Receptor Engineered Human T Cells Directed to Tumor Stroma. Mol. Ther. 2013, 21, 1611–1620. [Google Scholar] [CrossRef] [PubMed]
  194. Xing, H.; Yang, X.; Xu, Y.; Tang, K.; Tian, Z.; Chen, Z.; Zhang, Y.; Xue, Z.; Rao, Q.; Wang, M.; et al. Anti-Tumor Effects of Vascular Endothelial Growth Factor/Vascular Endothelial Growth Factor Receptor Binding Domain-Modified Chimeric Antigen Receptor T Cells. Cytotherapy 2021, 23, 810–819. [Google Scholar] [CrossRef] [PubMed]
  195. Schmitt, F.C.; Longatto Filho, A.; Lopes, J.M. Angiogenesis and Breast Cancer. J. Oncol. 2010, 2010, 576384. [Google Scholar] [CrossRef]
  196. Akbari, P.; Katsarou, A.; Daghighian, R.; van Mil, L.W.H.G.; Huijbers, E.J.M.; Griffioen, A.W.; van Beijnum, J.R. Directing CAR T Cells towards the Tumor Vasculature for the Treatment of Solid Tumors. Biochim. Biophys. Acta-Rev. Cancer 2022, 1877, 188701. [Google Scholar] [CrossRef]
  197. Ghiabi, P.; Jiang, J.; Pasquier, J.; Maleki, M.; Abu-Kaoud, N.; Halabi, N.; Guerrouahen, B.S.; Rafii, S.; Rafii, A. Breast Cancer Cells Promote a Notch-Dependent Mesenchymal Phenotype in Endothelial Cells Participating to a pro-Tumoral Niche. J. Transl. Med. 2015, 13, 27. [Google Scholar] [CrossRef]
  198. Byrd, T.T.; Fousek, K.; Pignata, A.; Szot, C.; Samaha, H.; Seaman, S.; Dobrolecki, L.; Salsman, V.S.; Oo, H.Z.; Bielamowicz, K.; et al. TEM8/ANTXR1-Specific CAR T Cells as a Targeted Therapy for Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 489–500. [Google Scholar] [CrossRef]
  199. Duan, Z.; Li, Z.; Wang, Z.; Chen, C.; Luo, Y. Chimeric Antigen Receptor Macrophages Activated through TLR4 or IFN-γ Receptors Suppress Breast Cancer Growth by Targeting VEGFR2. Cancer Immunol. Immunother. 2023, 72, 3243–3257. [Google Scholar] [CrossRef]
  200. Li, Y.; Zhao, L.; Li, X.F. Hypoxia and the Tumor Microenvironment. Technol. Cancer Res. Treat. 2021, 20, 15330338211036304. [Google Scholar] [CrossRef]
  201. Schurich, A.; Magalhaes, I.; Mattsson, J. Metabolic Regulation of CAR T Cell Function by the Hypoxic Microenvironment in Solid Tumors. Immunotherapy 2019, 11, 335–345. [Google Scholar] [CrossRef] [PubMed]
  202. Buck, M.D.; Sowell, R.T.; Kaech, S.M.; Pearce, E.L. Metabolic Instruction of Immunity. Cell 2017, 169, 570–586. [Google Scholar] [CrossRef] [PubMed]
  203. Lourdes Mora-García, M.; García-Rocha, R.; Morales-Ramírez, O.; Montesinos, J.J.; Weiss-Steider, B.; Hernández-Montes, J.; Ávila-Ibarra, L.R.; Don-López, C.A.; Velasco-Velázquez, M.A.; Gutiérrez-Serrano, V.; et al. Mesenchymal Stromal Cells Derived from Cervical Cancer Produce High Amounts of Adenosine to Suppress Cytotoxic T Lymphocyte Functions. J. Transl. Med. 2016, 14, 302. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, Y.; Ertl, H.C.J. Depletion of FAP+ Cells Reduces Immunosuppressive Cells and Improves Metabolism and Functions CD8+T Cells within Tumors. Oncotarget 2016, 7, 23282–23299. [Google Scholar] [CrossRef] [PubMed]
  205. Kraman, M.; Bambrough, P.J.; Arnold, J.N.; Roberts, E.W.; Magiera, L.; Jones, J.O.; Gopinathan, A.; Tuveson, D.A.; Fearon, D.T. Suppression of Antitumor Immunity by Stromal Cells Expressing Fibroblast Activation Protein–α. Science 2010, 330, 827–830. [Google Scholar] [CrossRef]
  206. Lo, A.; Wang, L.-C.S.; Scholler, J.; Monslow, J.; Avery, D.; Newick, K.; O’Brien, S.; Evans, R.A.; Bajor, D.J.; Clendenin, C.; et al. Tumor-Promoting Desmoplasia Is Disrupted by Depleting FAP-Expressing Stromal Cells. Cancer Res. 2015, 75, 2800–2810. [Google Scholar] [CrossRef]
  207. Leone, R.D.; Sun, I.M.; Oh, M.H.; Sun, I.H.; Wen, J.; Englert, J.; Powell, J.D. Inhibition of the Adenosine A2a Receptor Modulates Expression of T Cell Coinhibitory Receptors and Improves Effector Function for Enhanced Checkpoint Blockade and ACT in Murine Cancer Models. Cancer Immunol. Immunother. 2018, 67, 1271–1284. [Google Scholar] [CrossRef]
  208. Zohair, B.; Chraa, D.; Rezouki, I.; Benthami, H.; Razzouki, I.; Elkarroumi, M.; Olive, D.; Karkouri, M.; Badou, A. The Immune Checkpoint Adenosine 2A Receptor Is Associated with Aggressive Clinical Outcomes and Reflects an Immunosuppressive Tumor Microenvironment in Human Breast Cancer. Front. Immunol. 2023, 14, 1201632. [Google Scholar] [CrossRef]
  209. Giuffrida, L.; Sek, K.; Henderson, M.A.; Lai, J.; Chen, A.X.Y.; Meyran, D.; Todd, K.L.; Petley, E.V.; Mardiana, S.; Mølck, C.; et al. CRISPR/Cas9 Mediated Deletion of the Adenosine A2A Receptor Enhances CAR T Cell Efficacy. Nat. Commun. 2021, 12, 3236. [Google Scholar] [CrossRef]
  210. Leen, A.M.; Sukumaran, S.; Watanabe, N.; Mohammed, S.; Keirnan, J.; Yanagisawa, R.; Anurathapan, U.; Rendon, D.; Heslop, H.E.; Rooney, C.M.; et al. Reversal of Tumor Immune Inhibition Using a Chimeric Cytokine Receptor. Mol. Ther. 2014, 22, 1211–1220. [Google Scholar] [CrossRef]
  211. Conticello, C.; Pedini, F.; Zeuner, A.; Patti, M.; Zerilli, M.; Stassi, G.; Messina, A.; Peschle, C.; De Maria, R. IL-4 Protects Tumor Cells from Anti-CD95 and Chemotherapeutic Agents via Up-Regulation of Antiapoptotic Proteins. J. Immunol. 2004, 172, 5467–5477. [Google Scholar] [CrossRef] [PubMed]
  212. Yang, W.-C.; Hwang, Y.-S.; Chen, Y.-Y.; Liu, C.-L.; Shen, C.-N.; Hong, W.-H.; Lo, S.-M.; Shen, C.-R. Interleukin-4 Supports the Suppressive Immune Responses Elicited by Regulatory T Cells. Front. Immunol. 2017, 8, 1508. [Google Scholar] [CrossRef] [PubMed]
  213. Shum, T.; Omer, B.; Tashiro, H.; Kruse, R.L.; Wagner, D.L.; Parikh, K.; Yi, Z.; Sauer, T.; Liu, D.; Parihar, R.; et al. Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells. Cancer Discov. 2017, 7, 1238–1247. [Google Scholar] [CrossRef] [PubMed]
  214. Hawkins, E.R.; D’Souza, R.R.; Klampatsa, A. Armored CAR T-Cells: The Next Chapter in T-Cell Cancer Immunotherapy. Biol. Targets Ther. 2021, 15, 95–105. [Google Scholar] [CrossRef]
  215. Zhou, Y.; Husman, T.; Cen, X.; Tsao, T.; Brown, J.; Bajpai, A.; Li, M.; Zhou, K.; Yang, L. Interleukin 15 in Cell-Based Cancer Immunotherapy. Int. J. Mol. Sci. 2022, 23, 7311. [Google Scholar] [CrossRef] [PubMed]
  216. Makkouk, A.; Yang, X.C.; Barca, T.; Lucas, A.; Turkoz, M.; Wong, J.T.S.; Nishimoto, K.P.; Brodey, M.M.; Tabrizizad, M.; Gundurao, S.R.Y.; et al. Off-the-Shelf Vδ 1 Gamma Delta T Cells Engineered with Glypican-3 (GPC-3)-Specific Chimeric Antigen Receptor (CAR) and Soluble IL-15 Display Robust Antitumor Efficacy against Hepatocellular Carcinoma. J. Immunother. Cancer 2021, 9, e003441. [Google Scholar] [CrossRef]
  217. Batra, S.A.; Rathi, P.; Guo, L.; Courtney, A.N.; Fleurence, J.; Balzeau, J.; Shaik, R.S.; Nguyen, T.P.; Wu, M.F.; Bulsara, S.; et al. Glypican-3-Specific CAR T Cells Coexpressing IL15 and IL21 Have Superior Expansion and Antitumor Activity against Hepatocellular Carcinoma. Cancer Immunol. Res. 2020, 8, 309–320. [Google Scholar] [CrossRef]
  218. Ruixin, S.; Yifan, L.; Chuanlong, W.; Min, Z.; Hong, L.; Guoxiu, D.; Zhengyang, L.; Yansha, S.; Yiwei, D.; Jingwen, S.; et al. Expressing IL-15/IL-18 and CXCR2 Improve Infiltration and Survival of EGFRvIII-Targeting CAR-T Cells in Breast Cancer. Biochem. Pharmacol. 2023, 212, 115536. [Google Scholar] [CrossRef]
  219. Chen, Y.; Sun, C.; Landoni, E.; Metelitsa, L.; Dotti, G.; Savoldo, B. Eradication of Neuroblastoma by T Cells Redirected with an Optimized GD2-Specific Chimeric Antigen Receptor and Interleukin-15. Clin. Cancer Res. 2019, 25, 2915–2924. [Google Scholar] [CrossRef]
  220. Gargett, T.; Ebert, L.M.; Truong, N.T.H.; Kollis, P.M.; Sedivakova, K.; Yu, W.; Yeo, E.C.F.; Wittwer, N.L.; Gliddon, B.L.; Tea, M.N.; et al. GD2-Targeting CAR-T Cells Enhanced by Transgenic IL-15 Expression Are an Effective and Clinically Feasible Therapy for Glioblastoma. J. Immunother. Cancer 2022, 10, e005187. [Google Scholar] [CrossRef]
  221. Lanitis, E.; Rota, G.; Kosti, P.; Ronet, C.; Spill, A.; Seijo, B.; Romero, P.; Dangaj, D.; Coukos, G.; Irving, M. Optimized Gene Engineering of Murine CAR-T Cells Reveals the Beneficial Effects of IL-15 Coexpression. J. Exp. Med. 2021, 218, e20192203. [Google Scholar] [CrossRef] [PubMed]
  222. Krenciute, G.; Prinzing, B.L.; Yi, Z.; Wu, M.-F.; Liu, H.; Dotti, G.; Balyasnikova, I.V.; Gottschalk, S. Transgenic Expression of IL15 Improves Antiglioma Activity of IL13Rα2-CAR T Cells but Results in Antigen Loss Variants. Cancer Immunol. Res. 2017, 5, 571–581. [Google Scholar] [CrossRef] [PubMed]
  223. Perna, S.K.; Pagliara, D.; Mahendravada, A.; Liu, H.; Brenner, M.K.; Savoldo, B.; Dotti, G. Interleukin-7 Mediates Selective Expansion of Tumor-Redirected Cytotoxic T Lymphocytes (CTLs) without Enhancement of Regulatory T-Cell Inhibition. Clin. Cancer Res. 2014, 20, 131–139. [Google Scholar] [CrossRef] [PubMed]
  224. Bradley, L.M.; Haynes, L.; Swain, S.L. IL-7: Maintaining T-Cell Memory and Achieving Homeostasis. Trends Immunol. 2005, 26, 172–176. [Google Scholar] [CrossRef] [PubMed]
  225. Luo, H.; Su, J.; Sun, R.; Sun, Y.; Wang, Y.; Dong, Y.; Shi, B.; Jiang, H.; Li, Z. Coexpression of IL7 and CCL21 Increases Efficacy of CAR-T Cells in Solid Tumors without Requiring Preconditioned Lymphodepletion. Clin. Cancer Res. 2020, 26, 5494–5505. [Google Scholar] [CrossRef] [PubMed]
  226. Ye, X.; Deng, X.; Wen, J.; Li, Y.; Zhang, M.; Cai, Z.; Liu, G.; Wang, H.; Cai, J. Folate Receptor-Alpha Targeted 7x19 CAR-Γδ T Suppressed Triple-Negative Breast Cancer Xenograft Model in Mice. J. Oncol. 2022, 2022, 2112898. [Google Scholar] [CrossRef] [PubMed]
  227. Swan, S.L.; Mehta, N.; Ilich, E.; Shen, S.H.; Wilkinson, D.S.; Anderson, A.R.; Segura, T.; Sanchez-Perez, L.; Sampson, J.H.; Bellamkonda, R.V. IL7 and IL7 Flt3L Co-Expressing CAR T Cells Improve Therapeutic Efficacy in Mouse EGFRvIII Heterogeneous Glioblastoma. Front. Immunol. 2023, 14, 1085547. [Google Scholar] [CrossRef]
  228. Li, G.; Zhang, Q.; Han, Z.; Zhu, Y.; Shen, H.; Liu, Z.; Zhou, Z.; Ding, W.; Han, S.; He, J.; et al. IL-7 and CCR2b Co-Expression-Mediated Enhanced CAR-T Survival and Infiltration in Solid Tumors. Front. Oncol. 2021, 11, 734593. [Google Scholar] [CrossRef]
  229. Xiong, X.; Xi, J.; Liu, Q.; Wang, C.; Jiang, Z.; Yue, S.; Shi, L.; Rong, Y. Co-expression of IL-7 and PH20 Promote Anti-GPC3 CAR-T Tumour Suppressor Activity in Vivo and in Vitro. Liver Int. 2021, 41, 1033–1043. [Google Scholar] [CrossRef]
  230. Yasuda, K.; Nakanishi, K.; Tsutsui, H. Interleukin-18 in Health and Disease. Int. J. Mol. Sci. 2019, 20, 649. [Google Scholar] [CrossRef]
  231. Hu, B.; Ren, J.; Luo, Y.; Keith, B.; Young, R.M.; Scholler, J.; Zhao, Y.; June, C.H. Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep. 2017, 20, 3025–3033. [Google Scholar] [CrossRef] [PubMed]
  232. Carroll, R.G.; Carpenito, C.; Shan, X.; Danet-Desnoyers, G.; Liu, R.; Jiang, S.; Albelda, S.M.; Golovina, T.; Coukos, G.; Riley, J.L.; et al. Distinct Effects of IL-18 on the Engraftment and Function of Human Effector CD8 T Cells and Regulatory T Cells. PLoS ONE 2008, 3, e3289. [Google Scholar] [CrossRef] [PubMed]
  233. Jaspers, J.E.; Khan, J.F.; Godfrey, W.D.; Lopez, A.V.; Ciampricotti, M.; Rudin, C.M.; Brentjens, R.J. IL-18-Secreting CAR T Cells Targeting DLL3 Are Highly Effective in Small Cell Lung Cancer Models. J. Clin. Investig. 2023, 133, e166028. [Google Scholar] [CrossRef] [PubMed]
  234. Chmielewski, M.; Abken, H. CAR T Cells Releasing IL-18 Convert to T-Bethigh FoxO1low Effectors That Exhibit Augmented Activity against Advanced Solid Tumors. Cell Rep. 2017, 21, 3205–3219. [Google Scholar] [CrossRef] [PubMed]
  235. Glienke, W.; Dragon, A.C.; Zimmermann, K.; Martyniszyn-Eiben, A.; Mertens, M.; Abken, H.; Rossig, C.; Altvater, B.; Aleksandrova, K.; Arseniev, L.; et al. GMP-Compliant Manufacturing of TRUCKs: CAR T Cells Targeting GD2 and Releasing Inducible IL-18. Front. Immunol. 2022, 13, 839783. [Google Scholar] [CrossRef] [PubMed]
  236. Choi, G.; Shin, G.; Bae, S.J. Price and Prejudice? The Value of Chimeric Antigen Receptor (CAR) T-Cell Therapy. Int. J. Environ. Res. Public Health 2022, 19, 12366. [Google Scholar] [CrossRef] [PubMed]
  237. Fiorenza, S.; Ritchie, D.S.; Ramsey, S.D.; Turtle, C.J.; Roth, J.A. Value and Affordability of CAR T-Cell Therapy in the United States. Bone Marrow Transplant. 2020, 55, 1706–1715. [Google Scholar] [CrossRef]
  238. Hernandez, I.; Prasad, V.; Gellad, W.F. Total Costs of Chimeric Antigen Receptor T-Cell Immunotherapy. JAMA Oncol. 2018, 4, 994. [Google Scholar] [CrossRef]
  239. Vormittag, P.; Gunn, R.; Ghorashian, S.; Veraitch, F.S. A Guide to Manufacturing CAR T Cell Therapies. Curr. Opin. Biotechnol. 2018, 53, 164–181. [Google Scholar] [CrossRef]
  240. Billingsley, M.M.; Singh, N.; Ravikumar, P.; Zhang, R.; June, C.H.; Mitchell, M.J. Ionizable Lipid Nanoparticle-Mediated MRNA Delivery for Human CAR T Cell Engineering. Nano Lett. 2020, 20, 1578–1589. [Google Scholar] [CrossRef]
  241. Nakazawa, Y.; Huye, L.E.; Dotti, G.; Foster, A.E.; Vera, J.F.; Manuri, P.R.; June, C.H.; Rooney, C.M.; Wilson, M.H. Optimization of the PiggyBac Transposon System for the Sustained Genetic Modification of Human T Lymphocytes. J. Immunother. 2009, 32, 826–836. [Google Scholar] [CrossRef] [PubMed]
  242. Vargas, J.E.; Chicaybam, L.; Stein, R.T.; Tanuri, A.; Delgado-Cañedo, A.; Bonamino, M.H. Retroviral Vectors and Transposons for Stable Gene Therapy: Advances, Current Challenges and Perspectives. J. Transl. Med. 2016, 14, 288. [Google Scholar] [CrossRef] [PubMed]
  243. Ivics, Z.N.; Hackett, P.B.; Plasterk, R.H.; Izsvá, Z. Molecular Reconstruction of Sleeping Beauty, a Tc1-like Transposon from Fish, and Its Transposition in Human Cells. Cell 1997, 91, 501–510. [Google Scholar] [CrossRef] [PubMed]
  244. Moretti, A.; Ponzo, M.; Nicolette, C.A.; Tcherepanova, I.Y.; Biondi, A.; Magnani, C.F. The Past, Present, and Future of Non-Viral CAR T Cells. Front. Immunol. 2022, 13, 867013. [Google Scholar] [CrossRef] [PubMed]
  245. Foster, J.B.; Barrett, D.M.; Karikó, K. The Emerging Role of In Vitro-Transcribed MRNA in Adoptive T Cell Immunotherapy. Mol. Ther. 2019, 27, 747–756. [Google Scholar] [CrossRef] [PubMed]
  246. Soundara Rajan, T.; Gugliandolo, A.; Bramanti, P.; Mazzon, E. In Vitro-Transcribed MRNA Chimeric Antigen Receptor T Cell (IVT MRNA CAR T) Therapy in Hematologic and Solid Tumor Management: A Preclinical Update. Int. J. Mol. Sci. 2020, 21, 6514. [Google Scholar] [CrossRef] [PubMed]
  247. Schaft, N.; Dörrie, J.; Müller, I.; Beck, V.; Baumann, S.; Schunder, T.; Kämpgen, E.; Schuler, G. A New Way to Generate Cytolytic Tumor-Specific T Cells: Electroporation of RNA Coding for a T Cell Receptor into T Lymphocytes. Cancer Immunol. Immunother. 2006, 55, 1132–1141. [Google Scholar] [CrossRef]
  248. Zhao, Y.; Zheng, Z.; Cohen, C.J.; Gattinoni, L.; Palmer, D.C.; Restifo, N.P.; Rosenberg, S.A.; Morgan, R.A. High-Efficiency Transfection of Primary Human and Mouse T Lymphocytes Using RNA Electroporation. Mol. Ther. 2006, 13, 151–159. [Google Scholar] [CrossRef]
  249. Billingsley, M.M.; Hamilton, A.G.; Mai, D.; Patel, S.K.; Swingle, K.L.; Sheppard, N.C.; June, C.H.; Mitchell, M.J. Orthogonal Design of Experiments for Optimization of Lipid Nanoparticles for MRNA Engineering of CAR T Cells. Nano Lett. 2022, 22, 533–542. [Google Scholar] [CrossRef]
  250. Parayath, N.N.; Stephan, S.B.; Koehne, A.L.; Nelson, P.S.; Stephan, M.T. In Vitro-Transcribed Antigen Receptor MRNA Nanocarriers for Transient Expression in Circulating T Cells in Vivo. Nat. Commun. 2020, 11, 6080. [Google Scholar] [CrossRef]
  251. Caldwell, K.J.; Gottschalk, S.; Talleur, A.C. Allogeneic CAR Cell Therapy—More Than a Pipe Dream. Front. Immunol. 2021, 11, 618427. [Google Scholar] [CrossRef] [PubMed]
  252. Berrien-Elliott, M.M.; Jacobs, M.T.; Fehniger, T.A. Allogeneic Natural Killer Cell Therapy. Blood 2023, 141, 856–868. [Google Scholar] [CrossRef] [PubMed]
  253. Morgan, M.A.; Büning, H.; Sauer, M.; Schambach, A. Use of Cell and Genome Modification Technologies to Generate Improved “Off-the-Shelf” CAR T and CAR NK Cells. Front. Immunol. 2020, 11, 1965. [Google Scholar] [CrossRef] [PubMed]
  254. Furukawa, Y.; Hamano, Y.; Shirane, S.; Kinoshita, S.; Azusawa, Y.; Ando, J.; Nakauchi, H.; Ando, M. Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using IPSC Technology and Gene Editing. Cells 2022, 11, 269. [Google Scholar] [CrossRef] [PubMed]
  255. Saetersmoen, M.L.; Hammer, Q.; Valamehr, B.; Kaufman, D.S.; Malmberg, K.J. Off-the-Shelf Cell Therapy with Induced Pluripotent Stem Cell-Derived Natural Killer Cells. Semin. Immunopathol. 2019, 41, 59–68. [Google Scholar] [CrossRef] [PubMed]
  256. Heipertz, E.L.; Zynda, E.R.; Stav-Noraas, T.E.; Hungler, A.D.; Boucher, S.E.; Kaur, N.; Vemuri, M.C. Current Perspectives on “Off-The-Shelf” Allogeneic NK and CAR-NK Cell Therapies. Front. Immunol. 2021, 12, 732135. [Google Scholar] [CrossRef] [PubMed]
  257. Daher, M.; Melo Garcia, L.; Li, Y.; Rezvani, K. CAR-NK Cells: The next Wave of Cellular Therapy for Cancer. Clin. Transl. Immunol. 2021, 10, e1274. [Google Scholar] [CrossRef]
  258. Liu, H.; Yang, B.; Sun, T.; Lin, L.; Hu, Y.; Deng, M.; Yang, J.; Liu, T.; Li, J.; Sun, S.; et al. Specific Growth Inhibition of ErbB2-Expressing Human Breast Cancer Cells by Genetically Modified NK-92 Cells. Oncol. Rep. 2015, 33, 95–102. [Google Scholar] [CrossRef]
  259. Chen, X.; Han, J.; Chu, J.; Zhang, L.; Zhang, J.; Chen, C.; Chen, L.; Wang, Y.; Wang, H.; Yi, L.; et al. A Combinational Therapy of EGFR-CAR NK Cells and Oncolytic Herpes Simplex Virus 1 for Breast Cancer Brain Metastases. Oncotarget 2016, 7, 27764–27777. [Google Scholar] [CrossRef]
  260. Hu, Z. Tissue Factor as a New Target for CAR-NK Cell Immunotherapy of Triple-Negative Breast Cancer. Sci. Rep. 2020, 10, 2815. [Google Scholar] [CrossRef]
  261. Uherek, C.; Tonn, T.; Uherek, B.; Becker, S.; Schnierle, B.; Klingemann, H.G.; Wels, W. Retargeting of Natural Killer-Cell Cytolytic Activity to ErbB2-Expressing Cancer Cells Results in Efficient and Selective Tumor Cell Destruction. Blood 2002, 100, 1265–1273. [Google Scholar] [CrossRef] [PubMed]
  262. Sahm, C.; Schönfeld, K.; Wels, W.S. Expression of IL-15 in NK Cells Results in Rapid Enrichment and Selective Cytotoxicity of Gene-Modified Effectors That Carry a Tumor-Specific Antigen Receptor. Cancer Immunol. Immunother. 2012, 61, 1451–1461. [Google Scholar] [CrossRef] [PubMed]
  263. Lin, Y.-Z.; Lee, C.-C.; Cho, D.-Y.; Wang, Y.-L.; Chen, C.-Y.; Weng, C.-Y.; Chiu, S.-C.; Hung, M.-C.; Wang, S.-C. Suppression of Breast Cancer Cells Resistant to a Pure Anti-Estrogen with CAR-Transduced Natural Killer Cells. Am. J. Cancer Res. 2021, 11, 4455–4469. [Google Scholar] [PubMed]
  264. Liu, Y.; Zhou, Y.; Huang, K.H.; Fang, X.; Li, Y.; Wang, F.; An, L.; Chen, Q.; Zhang, Y.; Shi, A.; et al. Targeting Epidermal Growth Factor-Overexpressing Triple-Negative Breast Cancer by Natural Killer Cells Expressing a Specific Chimeric Antigen Receptor. Cell Prolif. 2020, 53, e12858. [Google Scholar] [CrossRef] [PubMed]
  265. Depil, S.; Duchateau, P.; Grupp, S.A.; Mufti, G.; Poirot, L. ‘Off-the-Shelf’ Allogeneic CAR T Cells: Development and Challenges. Nat. Rev. Drug Discov. 2020, 19, 185–199. [Google Scholar] [CrossRef] [PubMed]
  266. Deng, J.; Yin, H. Gamma Delta (Γδ) T Cells in Cancer Immunotherapy; Where It Comes from, Where It Will Go? Eur. J. Pharmacol. 2022, 919, 174803. [Google Scholar] [CrossRef] [PubMed]
  267. Capietto, A.-H.; Martinet, L.; Fournié, J.-J. Stimulated Γδ T Cells Increase the In Vivo Efficacy of Trastuzumab in HER-2+ Breast Cancer. J. Immunol. 2011, 187, 1031–1038. [Google Scholar] [CrossRef]
  268. Dhar, S.; Chiplunkar, S.V. Lysis of Aminobisphosphonate-Sensitized MCF-7 Breast Tumor Cells by Vγ9Vδ2 T Cells. Cancer Immun. 2010, 10, 1–10. [Google Scholar]
  269. Chen, H.; Joalland, N.; Bridgeman, J.S.; Alchami, F.S.; Jarry, U.; Khan, M.W.A.; Piggott, L.; Shanneik, Y.; Li, J.; Herold, M.J.; et al. Synergistic Targeting of Breast Cancer Stem-like Cells by Human Γδ T Cells and CD8+ T Cells. Immunol. Cell Biol. 2017, 95, 620–629. [Google Scholar] [CrossRef]
  270. Klebanoff, C.A.; Khong, H.T.; Antony, P.A.; Palmer, D.C.; Restifo, N.P. Sinks, Suppressors and Antigen Presenters: How Lymphodepletion Enhances T Cell-Mediated Tumor Immunotherapy. Trends Immunol. 2005, 26, 111–117. [Google Scholar] [CrossRef]
  271. Hu, W.; Zi, Z.; Jin, Y.; Li, G.; Shao, K.; Cai, Q.; Ma, X.; Wei, F. CRISPR/Cas9-Mediated PD-1 Disruption Enhances Human Mesothelin-Targeted CAR T Cell Effector Functions. Cancer Immunol. Immunother. 2019, 68, 365–377. [Google Scholar] [CrossRef] [PubMed]
  272. 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] [PubMed]
  273. Srivastava, S.; Furlan, S.N.; Jaeger-Ruckstuhl, C.A.; Sarvothama, M.; Berger, C.; Smythe, K.S.; Garrison, S.M.; Specht, J.M.; Lee, S.M.; Amezquita, R.A.; et al. Immunogenic Chemotherapy Enhances Recruitment of CAR-T Cells to Lung Tumors and Improves Antitumor Efficacy When Combined with Checkpoint Blockade. Cancer Cell 2021, 39, 193–208.e10. [Google Scholar] [CrossRef] [PubMed]
  274. Adusumilli, P.S.; Zauderer, M.G.; Rivière, I.; Solomon, S.B.; Rusch, V.W.; O’Cearbhaill, R.E.; Zhu, A.; Cheema, W.; Chintala, N.K.; Halton, E.; et al. A Phase I Trial of Regional Mesothelin-Targeted CAR T-Cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab. Cancer Discov. 2021, 11, 2748–2763. [Google Scholar] [CrossRef] [PubMed]
  275. Lu, Y.C.; Parker, L.L.; Lu, T.; Zheng, Z.; Toomey, M.A.; White, D.E.; Yao, X.; Li, Y.F.; Robbins, P.F.; Feldman, S.A.; et al. Treatment of Patients with Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3. J. Clin. Oncol. 2017, 35, 3322–3329. [Google Scholar] [CrossRef] [PubMed]
  276. Ishihara, M.; Kitano, S.; Kageyama, S.; Miyahara, Y.; Yamamoto, N.; Kato, H.; Mishima, H.; Hattori, H.; Funakoshi, T.; Kojima, T.; et al. NY-ESO-1-Specific Redirected T Cells with Endogenous TCR Knockdown Mediate Tumor Response and Cytokine Release Syndrome. J. Immunother. Cancer 2022, 10, e003811. [Google Scholar] [CrossRef] [PubMed]
  277. Srivastava, S.; Salter, A.I.; Liggitt, D.; Yechan-Gunja, S.; Sarvothama, M.; Cooper, K.; Smythe, K.S.; Dudakov, J.A.; Pierce, R.H.; Rader, C.; et al. Logic-Gated ROR1 Chimeric Antigen Receptor Expression Rescues T Cell-Mediated Toxicity to Normal Tissues and Enables Selective Tumor Targeting. Cancer Cell 2019, 35, 489–503.e8. [Google Scholar] [CrossRef]
  278. Mhaidly, R.; Verhoeyen, E. Humanized Mice Are Precious Tools for Preclinical Evaluation of Car t and Car Nk Cell Therapies. Cancers 2020, 12, 1915. [Google Scholar] [CrossRef]
  279. Wang, M.; Yao, L.C.; Cheng, M.; Cai, D.; Martinek, J.; Pan, C.X.; Shi, W.; Ma, A.H.; De Vere White, R.W.; Airhart, S.; et al. Humanized Mice in Studying Efficacy and Mechanisms of PD-1-Targeted Cancer Immunotherapy. FASEB J. 2018, 32, 1537–1549. [Google Scholar] [CrossRef]
  280. Scherer, S.D.; Riggio, A.I.; Haroun, F.; DeRose, Y.S.; Ekiz, H.A.; Fujita, M.; Toner, J.; Zhao, L.; Li, Z.; Oesterreich, S.; et al. An Immune-Humanized Patient-Derived Xenograft Model of Estrogen-Independent, Hormone Receptor Positive Metastatic Breast Cancer. Breast Cancer Res. 2021, 23, 100. [Google Scholar] [CrossRef]
  281. Capasso, A.; Lang, J.; Pitts, T.M.; Jordan, K.R.; Lieu, C.H.; Davis, S.L.; Diamond, J.R.; Kopetz, S.; Barbee, J.; Peterson, J.; et al. Characterization of Immune Responses to Anti-PD-1 Mono and Combination Immunotherapy in Hematopoietic Humanized Mice Implanted with Tumor Xenografts. J. Immunother. Cancer 2019, 7, 37. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structural components of CARs vs. E-TCRs. (A) Illustrates the binding of monoclonal antibodies to their respective surface antigen molecule via their variable heavy and light chains. (B) Illustrates the physiologic activation of non-engineered T-cells, which rely on the binding of the variable regions of their α and β chains to a peptide-MHC complex in the surface of the antigen-presenting cell and requires the other CD3ε/ɣ/δ/ζ subunits to signal. For complete activation, the T-cell also receives signaling from co-stimulatory receptors (in this case, CD28). (C) Shows on the right a second-generation CAR, which combines the antigen-recognition components of a mAb with the activation and co-stimulatory signals of a TCR. CARs, therefore, can recognize antigens in an MHC-independent manner. On the left, we show E-TCRs, which function in the same manner as the native TCR, requiring MHC-peptide recognition and the presence of the CD3ε/ɣ/δ/ζ subunits to signal, and lack inclusion of co-stimulation.
Figure 1. Structural components of CARs vs. E-TCRs. (A) Illustrates the binding of monoclonal antibodies to their respective surface antigen molecule via their variable heavy and light chains. (B) Illustrates the physiologic activation of non-engineered T-cells, which rely on the binding of the variable regions of their α and β chains to a peptide-MHC complex in the surface of the antigen-presenting cell and requires the other CD3ε/ɣ/δ/ζ subunits to signal. For complete activation, the T-cell also receives signaling from co-stimulatory receptors (in this case, CD28). (C) Shows on the right a second-generation CAR, which combines the antigen-recognition components of a mAb with the activation and co-stimulatory signals of a TCR. CARs, therefore, can recognize antigens in an MHC-independent manner. On the left, we show E-TCRs, which function in the same manner as the native TCR, requiring MHC-peptide recognition and the presence of the CD3ε/ɣ/δ/ζ subunits to signal, and lack inclusion of co-stimulation.
Cancers 16 00124 g001
Figure 2. Challenges faced by E-ACTs in the treatment of solid malignancies. From right to left, E-ACTs must traffic and extravasate to the tumor site, infiltrate the ECM, and persist in the immunosuppressive tumor microenvironment before finally encountering their target. Each level presents its own set of unique challenges. All the illustrated cellular components of the TME (Stromal cells, endothelial cells, M2 macrophages, monocytic and granulocytic myeloid-derived suppressor cells, regulatory T-cells, and tumor cells) each play their role in the inhibition of T-cell effector functions.
Figure 2. Challenges faced by E-ACTs in the treatment of solid malignancies. From right to left, E-ACTs must traffic and extravasate to the tumor site, infiltrate the ECM, and persist in the immunosuppressive tumor microenvironment before finally encountering their target. Each level presents its own set of unique challenges. All the illustrated cellular components of the TME (Stromal cells, endothelial cells, M2 macrophages, monocytic and granulocytic myeloid-derived suppressor cells, regulatory T-cells, and tumor cells) each play their role in the inhibition of T-cell effector functions.
Cancers 16 00124 g002
Figure 3. Strategies to further engineer T cells to improve their efficacy. Methods that have been explored to increase the persistence and functionality of CAR T-cells include (A) Incorporating a cytokine transgene allowing additional production of stimulatory cytokines (IL-15, IL-7, and IL-18), (B) Constitutively active IL-7 receptor which signals without the need for IL-7 cytokine binding, (C) engineering switch receptors that turn an inhibitory signal into a positive stimulating one, such as an IL-4 receptor with an IL-7 signaling domain, (D) knockout of receptors that transmit inhibitory signals to T-cells, such as the A2aR receptor, which inhibits T-cells in the presence of adenosine, and (E) the costimulatory TR2.4-1BB receptor that induces activation of TRAIL-R2, thereby leading to apoptosis of MDSCs while delivering a co-stimulatory signal through the 4-1BB endodomain.
Figure 3. Strategies to further engineer T cells to improve their efficacy. Methods that have been explored to increase the persistence and functionality of CAR T-cells include (A) Incorporating a cytokine transgene allowing additional production of stimulatory cytokines (IL-15, IL-7, and IL-18), (B) Constitutively active IL-7 receptor which signals without the need for IL-7 cytokine binding, (C) engineering switch receptors that turn an inhibitory signal into a positive stimulating one, such as an IL-4 receptor with an IL-7 signaling domain, (D) knockout of receptors that transmit inhibitory signals to T-cells, such as the A2aR receptor, which inhibits T-cells in the presence of adenosine, and (E) the costimulatory TR2.4-1BB receptor that induces activation of TRAIL-R2, thereby leading to apoptosis of MDSCs while delivering a co-stimulatory signal through the 4-1BB endodomain.
Cancers 16 00124 g003
Figure 4. Current trends of E-ACT clinical trials for breast cancer. E-ACT trials registered in clinicaltrials.gov were assessed as of 24 July 2023. (A) Types of E-ACT trials. (B) The number of new E-ACT trials initiated each year and the cumulative number of registered E-ACT trials by year. (C) Phases of the 50 E-ACT trials. (D) Clinical status of the 50 E-ACT trials. (E) Use of lymphodepletion in E-ACT trials. (F) Restricting HLAs of the 14 TCR T trials. (G) Frequency of targets in E-ACT trials. (H) Locations where E-ACT trials have been conducted by country. (I) Primary sponsors of the 29 E-ACT trials conducted in the United States.
Figure 4. Current trends of E-ACT clinical trials for breast cancer. E-ACT trials registered in clinicaltrials.gov were assessed as of 24 July 2023. (A) Types of E-ACT trials. (B) The number of new E-ACT trials initiated each year and the cumulative number of registered E-ACT trials by year. (C) Phases of the 50 E-ACT trials. (D) Clinical status of the 50 E-ACT trials. (E) Use of lymphodepletion in E-ACT trials. (F) Restricting HLAs of the 14 TCR T trials. (G) Frequency of targets in E-ACT trials. (H) Locations where E-ACT trials have been conducted by country. (I) Primary sponsors of the 29 E-ACT trials conducted in the United States.
Cancers 16 00124 g004
Table 1. Comparison of TCR and CAR T-cell constructs.
Table 1. Comparison of TCR and CAR T-cell constructs.
TCR TCAR T
ConstructsMinimally engineered TCRFully synthetic receptor
MHC RestrictionDependentIndependent
Affinity and SensitivityLower affinity,
higher sensitivity
Higher affinity,
lower sensitivity
Antigens RecognizedPeptides presented within the MHC molecule (proteins)Cell surface proteins/molecules
Origin of AntigensIntra-/ExtracellularCell surface
Co-stimulatory MoleculesEndogenous CD28, 4-1BBLinked to scFv (CD28, 4-1BB in combination with CD3ζ
Probability of CRSLowerHigher
References[8,9,10]
Table 2. Common breast cancer TAA, CGA, and TSA targets.
Table 2. Common breast cancer TAA, CGA, and TSA targets.
Type of AntigenTargetPrognostic/Clinical AssociationExpression in Breast CancerTranslational StatusRef.
TAAHER2Overexpression promotes tumor proliferation, migration, and survivalHER2+ (overexpression): ~20%|HER2-low: ~45–55%Preclinical studies
Clinical trials
[3,29,36,37,38,39,40,41,42,43,44]
c-Met (HGFR)Chemotherapy resistance|Poor survival|Increased tumor migration, invasion, and proliferation~50% of breast cancerPreclinical studies
Clinical trials
[45,46,47,48]
MUC1Hypo-glycosylated in tumor cells|Associated with tumor invasion, metastasis, and angiogenesis>90% of breast cancerPreclinical studies
Clinical trials
[49,50,51,52,53]
MesothelinMetastasis|Decreased survival67% of TNBCPreclinical studies
Clinical trials
[54,55,56,57,58,59]
EpCAMWorse overall survival (all cases)|Unfavorable prognosis (basal-like/luminal B HER2+)|Favorable prognosis (HER2+)65% ER−|43% ER+|54% HER2+|47% HER2-Preclinical studies
Clinical trials
[60,61,62]
ROR1Aggressive disease|Tumor cell growth and survival~40% of breast cancer
22–57% of TNBC
Preclinical studies
Clinical trials
[63,64,65,66,67,68,69]
CEAHigher tumor burden|Poor overall survivalElevated serum levels in 10.9–16.7% of patientsClinical trials[70,71]
NKG2DLInduced by malignant transformation of cells|May result in favorable outcomesMIC-AB: 50%|ULBP-1: 90%|ULBP-2: 99%|ULBP-3: 100%|ULBP-4: 26%|ULBP-5: 90%Preclinical studies
Clinical trials
[72,73]
CSPG4Disease recurrence|Poor overall survival|Tumor migration, invasion, angiogenesis, and metastasis77% of breast cancerPreclinical studies[74,75,76]
FRαPoor outcomes (early recurrence)30% of breast cancer|70–80% of stage IV metastatic TNBCPreclinical studies[77,78,79,80]
Ganglioside GD2Stem cell marker|Tumorigenesis and migration35% of breast cancerPreclinical studies
Clinical trials
[81,82,83,84]
EGFRPoor prognosis|Poor disease-free survival (high EGFR copy number)61.2–64% of TNBCPreclinical studies
Clinical trials
[85,86,87,88]
ICAM-1Promotes bone metastasis|Aggressive phenotype|Metastasis|Poor prognosisOverexpressed in TNBC (% not specified)Preclinical studies[62,89,90,91]
CD24Advanced stage|Shorter survival|Resistance to chemotherapyHighest expression seen in HER2+ and TNBC samples (% not specified)Preclinical studies[92]
AXLTolerance of chromosomal instability|Therapy resistance|Reduced survival|Supports EMT and metastasisOverexpressed (% not specified)Preclinical studies[93,94,95,96,97,98]
CGANY-ESO-1High humoral immune response|No association with overall survival or progression-free survival17–28.6% TNBC|12.5% HER2+Preclinical studies
Clinical trials
[99,100,101,102]
MAGE-A3Worse prognosis|Reduced overall survival~10–15% of breast cancerPreclinical studies
Clinical trials
[103,104]
MAGE-A1Lower overall survival~6% of breast cancerClinical trials[105]
KK-LC-1TNBC cell stemness|Poor survival|Malignant cell behavior75% of TNBCClinical trials[106,107,108]
TSAPIK3CA H1047LCell transformation|Tumor proliferation|Resistance to apoptosis|Detected in tumors with favorable characteristicsPIK3CA mutations: 30–40% of breast cancer|~4% of PIK3CA mutations are H1047LPreclinical studies[34,109,110,111,112]
TP53 R175HCell migration/invasion through enhanced EGFR activation|Supports tumor microenvironment|Poor survivalTP53 mutations: ~30% of breast cancer, 50% of inflammatory breast cancer|TP53 R175H: 7% of breast cancerPreclinical studies
Clinical trials
[113,114,115,116,117]
Table 3. Available breast cancer E-ACT trial results.
Table 3. Available breast cancer E-ACT trial results.
YearTrial IDTargetTotal # PtsCommentsPhaseResponsesAdverse EffectsRef.
CAR2017NCT01837602c-Met6Intratumoral administration; mRNA electroporated CAR T-cells; Tumors resected two days later0/IClinical response was not measuredAll grade 3 SAEs were deemed unrelated to the study drug[47]
2023NCT03060356c-Met7mRNA electroporated CAR T-cells; Up to six infusions of CAR T-cells without LD; CAR T not found in tumor biopsy; 4 TNBC patients.I4/7 = 57.1% SDNo grade 3 or higher toxicity[272]
2021NCT02706392ROR121CAR T-cells were seen in tumor biopsy; CAR T-cells upregulated inhibitory receptors and lost the ability to produce effector cytokines; 3 TNBC patientsI2/21 = 9.5% SDAll patients noted as experiencing adverse events[273]
2021NCT02414269Mesothelin27LD, Intrapleural administration, +Pembrolizumab; 1 BC patientI/II56% SD; ORR: 12.5% (PR); BC patient did not respondNo adverse events were noted[274]
TCR2017NCT02111850MAGE-A317LD and high-dose IL-2 were given;
2 BC patients
I/IIORR: 23.5%
5.9% (CR), 17.6% (PR)
BC patients did not respond
Transient G3 transaminitis (2 pts)[275]
2022NCT01967823NY-ESO-19LD; 1 BC patientIORR: 33% (PR)
BC patient did not respond
1 Grade 3 Lung injury
3 CRS
[276]
2022NCT03412877p53 R175H1LD; Pembrolizumab given on day 16 after TCR T-cellsII55% decrease in tumor burden. Progressed six months post-treatment due to loss of HLA expressionGrade 3 acute CRS, resolved[116]
LD = lymphodepletion, ORR = objective response rate, SD = stable disease, CR = complete response, SAE = serious adverse event, CRS = cytokine release syndrome.
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

Chamorro, D.F.; Somes, L.K.; Hoyos, V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers 2024, 16, 124. https://doi.org/10.3390/cancers16010124

AMA Style

Chamorro DF, Somes LK, Hoyos V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers. 2024; 16(1):124. https://doi.org/10.3390/cancers16010124

Chicago/Turabian Style

Chamorro, Diego F., Lauren K. Somes, and Valentina Hoyos. 2024. "Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential" Cancers 16, no. 1: 124. https://doi.org/10.3390/cancers16010124

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