Next Article in Journal
Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions
Next Article in Special Issue
The Antidepressant Drug Amitriptyline Affects Human SH-SY5Y Neuroblastoma Cell Proliferation and Modulates Autophagy
Previous Article in Journal
Recent Developments in the Role of Protein Tyrosine Phosphatase 1B (PTP1B) as a Regulator of Immune Cell Signalling in Health and Disease
Previous Article in Special Issue
Gene Expression Reprogramming by Citrate Supplementation Reduces HepG2 Cell Migration and Invasion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert “Cold” Tumors to “Hot” Tumors

by
Liucui Yang
,
Qingyi Hu
* and
Tao Huang
*
Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 7208; https://doi.org/10.3390/ijms25137208
Submission received: 15 May 2024 / Revised: 20 June 2024 / Accepted: 25 June 2024 / Published: 29 June 2024
(This article belongs to the Special Issue New Agents and Novel Drugs Use for the Oncological Diseases Treatment)

Abstract

:
Breast cancer characterized as “cold tumors” exhibit low levels of immune cell infiltration, which limits the efficacy of conventional immunotherapy. Recent studies have focused on strategies using nanotechnology combined with tumor microenvironment modulation to transform “cold tumors” into “hot tumors”. This approach involves the use of functionalized nanoparticles that target and modify the tumor microenvironment to promote the infiltration and activation of antitumor immune cells. By delivering immune activators or blocking immunosuppressive signals, these nanoparticles activate otherwise dormant immune responses, enhancing tumor immunogenicity and the therapeutic response. These strategies not only promise to increase the response rate of breast cancer patients to existing immunotherapies but also may pave new therapeutic avenues, providing a new direction for the immunotherapy of breast cancer.

1. Introduction

In the past decade (2010–2019), the incidence of breast cancer (BC) has gradually increased at a rate of 0.5% per year [1]. In 2020, female BC became the most commonly diagnosed cancer in the world for the first time, with an estimated number of new cases reaching 2.26 million [2]. By 2040, the global burden of BC is expected to increase to more than 3 million new cases per year [3]. Surgical treatment, radiotherapy and chemotherapy remain the basic methods for treating BC. For early-stage patients (stage I or II), surgical treatment can be combined with postoperative chemoradiotherapy; for stage III BC, patients mainly receive chemoradiotherapy, and a few receive mastectomy; for distant metastasis (stage IV) BC, patients only receive chemoradiotherapy, and hormone receptor (HR)-positive patients can choose endocrine therapy. The five-year relative survival rates for BC differ by stage. Stage I has a 99% survival rate, with the cancer confined solely to the breast. Stage II shows a survival rate of 93–97%, as the cancer spreads to adjacent lymph nodes. In Stage III, the rate is 72–86% when the cancer reaches distant lymph nodes or muscles. Stage IV, or metastatic BC, has a survival rate of about 28%, with cancer spreading to distant sites. These data emphasize the critical challenges in treating BC, especially late-stage (stage III or IV) [4].
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of BC that is associated with a poor prognosis, high recurrence and metastasis rates [5]. Neoadjuvant chemotherapy is the primary treatment option for early-stage TNBC due to the absence of available endocrine or targeted therapies [6]. However, neoadjuvant therapy achieved pathological complete remission (pCR) in 30–40% of TNBC patients. The remaining patients showed moderate response or drug resistance [7].
Conventional clinical interventions frequently result in the destruction of healthy tissues and increase the risk of tumor recurrence [8]. In contrast, immunotherapy is a more effective and specific treatment for tumor recognition by enhancing immune surveillance. As an innate immune “cold” tumor, in BC processes of occurrence and development, cancer cells reduce immunogenicity to evade the host immune surveillance, inhibit intratumoral infiltration and the activation of cytotoxic T lymphocytes (CTLs) [9,10]. On the other hand, there are a large number of tumors infiltrating immune cells in the immunosuppressive tumor microenvironment (TME) of BC, which promote tumor progression by promoting cancer cells proliferation, vascular formation and immunosuppression (Figure 1) [11,12]. Up until now, the results in clinical trials of immunotherapy have been disappointing. Nevertheless, the modulation of the immunosuppressive TME to convert “cold” immunosuppression tumors into “hot” immune-activating tumors represents a promising avenue for enhancing the efficacy of BC treatment [13].

2. Tumor Microenvironment, the “Soil” on Which Cancer Cells Depend for Survival

In the initial stages of cancer development, immune cells and cancer cells collaboratively establish the TME to foster tumor plasticity and heterogeneity. The recognition of the TME’s role in cancer emerged as Hanahan and Weinberg outlined ten biological hallmarks that characterize cancer complexity [14,15,16]. These include proliferative signaling, growth suppressor evasion, cell death resistance, angiogenesis, metastasis activation, replicative immortality, metabolic reprogramming, immune evasion, inflammation promotion and genome instability. This framework enhances our understanding of cancer and drives the development of targeted therapies, advancing oncology significantly.
In fact, the TME contributes to various aspects in promoting cancer occurrence, which include initiation, survival, growth, metastasis and immune evasion. The previous cancer researchers mainly focused on the tumor cell itself and a series of molecular events occurring within the tumor cell, limiting cancer research to the cancer cell itself, ignoring the role of the TME and the systemic regulation of the body in the occurrence and development of cancer, which is very one-sided.
Based on the different immune components, the TME is classified into “cold” and “hot” phenotypes [17]. Immune “cold” tumors are characterized by low tumor immunogenicity, defects in tumor antigen processing and presentation mechanisms, insufficient T cells infiltration and an immunosuppressive TME [18]. These features significantly impact the effectiveness of immunotherapies and the overall prognosis of cancer patients. Although some immune populations in TME have the potential to clear malignant cells, many factors in tumors can weaken this activity or reprogram cells to promote tumorigenesis. For example, dendritic cells (DCs) and macrophages are the most important antigen-presenting cells (APCs) in the TME. Meanwhile, tumor-associated macrophage (TAM) polarization, increased myeloid-derived suppressor cells (MDSCs) and DC maturation inhibition can exacerbate immunosuppressive TME that jointly coordinates the activation and function of antitumor CD8+ T cells [19,20,21].
In addition to the relative proportion, the location of immune cells is also associated with disease progression. For instance, in ductal carcinoma in situ (DCIS), focal dysplasia of the myoepithelial cell layer is associated with leukocyte enrichment, suggesting its role in promoting or sustaining local invasion [22]. Additionally, T cells rejection within the tumor niche may enhance resistance to immune checkpoint inhibitor (ICI) therapy in advanced stages of the disease [23,24].
Non-immune stromal cells, including fibroblasts, endothelial cells and pericytes, also exhibit unique gene/protein expression profiles distinct from their normal tissue counterparts and contribute to the characteristics of a “cold” TME. For example, cancer-associated fibroblasts (CAFs) can suppress T cells responses through various pathways. They directly inhibit T cells by expressing PD-L2 and FAS ligands and indirectly by secreting cytokines and recruiting immunosuppressive myeloid cells [25,26,27].
The physical and chemical properties of the TME, such as matrix stiffness, density, composition, pH value and interstitial fluid flow, are dysregulated, impacting cancer risk, tumor progression and treatment efficacy [28,29]. The hardness of most solid tumors, including BC, is significantly higher compared to surrounding normal tissues, and they exhibit a fibrotic state known as the “desmoplastic reaction” [30,31]. The alterations in the physical properties of the matrix further regulate the TME [32], such as higher levels of collagen and collagen crosslinking, which enhance pro-tumorigenic phenotypic macrophages [33,34,35]. The uncontrolled proliferation of cancer cells leads to poor hemoperfusion efficiency and increased glucose metabolism rates, forming an acidic TME. Decreased pH values are associated with tumor aggressiveness, angiogenesis, cell clock disruption and stemness, which also hinder CD8+ T cells reactivity [36,37,38,39,40].
Our understanding of the complex interaction between cancer and the immune system has shifted from the concept of “immune surveillance” to the concept of “immune editing”, which brings great hope for the treatment of malignant tumors by editorializing the immune microenvironment to monitor and control tumor development.

3. Strategies of Editing the Tumor Microenvironment for Breast Cancer Treatment: Nano-Based Approaches

3.1. T Cells

T cells, as essential effector cells, execute a vital function in antitumor immunity. The abundant infiltration of T cells is also a hallmark characteristic of “hot tumors”. Consequently, the strategies for recruiting and augmenting the activity of T cells within the TME have emerged as the central focus of researchers [41,42,43,44].
T cell bispecific antibodies (TCBs) are characterized by their dual antigen-binding domains derived from two distinct proteins [45]. Each domain is specifically designed to engage with different cellular targets—one domain targets T cells, while the other is directed against tumor cells. For instance, TCBs employ antibodies against CD3, a marker on T cells, in conjunction with antibodies that recognize specific tumor cell antigens. This design facilitates the redirection of CD3-positive T cells towards tumor cells, enabling a focused and potent immune-mediated attack on the cancer cells. Owing to the overexpression of human epidermal growth factor receptor 2 (HER2) in approximately 20% of primary invasive BCs, CD3/HER2 TCB is extensively employed in clinical studies. Nonetheless, severe cases of targeted toxicity have been observed in clinical trials (NCT00924287) [46,47,48]. To avoid side effects, attempts are being made to confine the selection of targeted antigens to those that are exclusively expressed in tumor cells, such as p95HER2, which present in merely 40% of HER2-positive BCs. Alternatively, the targeting of bispecific antibodies necessitates enhancement to avoid off-targeting [49].
TNBC demonstrates no overexpression of HER2, and CD3/TROP2 TCB (F7AK3) exhibits specific binding to both T cells and TROP2, which is overexpressed in TNBC cell lines such as MDA-MB-231, MDA-MB-468 and HCC1395. In animal studies, F7AK3 has been observed to facilitate a significant expansion of immune cells within tumors and the spleen, elevate IL-2 expression and reduce tumor size and growth rate (Table 1) [50].
The proliferation, survival and differentiation of T cells are primarily governed by interleukin (IL)-like cytokines, such as IL-2, IL-12 and IL-15. Initial attempts focused on augmenting T cells responses through the systemic administration of free cytokines, resulting in tumor rejection and an antitumor memory immune response, without inducing an increase in generating Tregs [71,72,73]. Yet, these approaches were soon abandoned due to their toxicity and limited half-times.
Nanoparticles (NPs) can deliver immune modulators directly to T cells, enhancing their activation and proliferation. The Enhanced Permeability and Retention (EPR) effect aids NP accumulation in tumor tissues. Functionalized NPs with specific ligands target T cells receptors for enhanced uptake [74]. A study demonstrated that immunostimulatory NPs reprogrammed dysfunctional APCs, leading to a significant activation of cytotoxic T cells via robust interferon-β production. This approach improved both short-term and long-term immune responses in breast cancer, particularly in the 4T1 mouse model [75].

3.2. Tumor-associated Macrophages, TAMs

Macrophages, as the most abundant immune cell type, are prevalent in various solid tumors, including BC. Tumor-infiltrating macrophages predominantly exhibit the M2 phenotype, instigating pro-tumor activities such as tumor progression, angiogenesis and metastasis, also known as tumor-associated macrophages (TAMs) [76,77,78,79,80,81,82,83]. Moreover, they are capable of secreting immunosuppressive cytokines and enzymes or presenting immune checkpoint proteins (notably PD-L1), which cause CD8+ T cells dysfunction, prompting in the generation of immunosuppressive TME (Figure 2) [84,85].
The initiation of M2 polarization signaling requires the binding of colony-stimulating factor (M-CSF) to the CSF1 receptor on macrophage surfaces. Additionally, the interaction between tumor cell CD47 and macrophage SIRPa inhibits the phagocytic function of the macrophages [86]. The development of tumors was effectively suppressed, and the population of M1-polarized macrophages was notably augmented in animal tests through the application of either small molecule inhibitors or nanoparticle-supported inhibitors. Nonetheless, the observed therapeutic outcome failed to meet the anticipated expectations [87]. NPs have emerged as a crucial element in facilitating multimodal therapy, enabling the delivery of potent drugs such as Doxorubicin and Paclitaxel to eradicate cancer cells and modulate macrophages (Table 2) [88,89,90].
Paclitaxel, for instance, has been demonstrated to reprogram M2-polarized macrophages into a M1-like phenotype via a TLR4-dependent pathway, thereby enhancing the antitumor immunity. In both in vitro and in vivo models, Paclitaxel not only inhibits tumor cell growth but also shifts the TAM profile from a M2-like to a M1-like phenotype. This process is potentially mediated through the activation of the TLR4 receptor and associated changes in gene expression and the release of proinflammatory cytokines [88]. Moreover, Paclitaxel can activate the cGAS-STING signaling pathway, prompting tumor cells to release soluble factors that induce M1-like polarization in macrophages [101]. In addition, a study on Paclitaxel demonstrated its ability to suppress the induction of M2 macrophages, suggesting a shift towards an M1-like profile even at lower concentrations [102].
However, numerous medications that influence macrophages also impact other immune cell populations. Thus, determining the authentic mechanism for macrophage-specific nano-immunotherapy remains a daunting task. Comprehensive immunoanalyses conducted at varying times within the tumor, lymph node/spleen and other relevant locations may provide new knowledge.

3.3. Dendritic Cells, DCs

DCs are crucial for generating long-lasting antitumor immunity by activating CTLs and NK cells (Figure 3) [103]. However, within the immunosuppressive TME, DCs often remain immature and tolerant [104]. DC vaccination can reverse this state, enhancing CTL and NK cell activation, increasing effector immune cell infiltration and reducing immunosuppressive cells like Tregs and MDSCs. Combining DC vaccination with therapies such as ICIs has shown synergistic effects, improving treatment outcomes and extending the progression-free survival in BC patients.
NPs enhance DC functions by delivering antigens or adjuvants (Table 3). They can penetrate dense tumor stroma and are functionalized with ligands targeting DC-specific receptors. In vitro methods such as culture medium incubation and electroporation can effectively introduce NPs into DCs. In vivo methods, like local injection and intravenous injection, rely on the natural immune surveillance function of DCs to uptake the NPs. The properties of NPs, such as size, shape, surface charge and coating, significantly influence their uptake efficiency by DCs. It is crucial to ensure that the DCs’ viability and functionality are not compromised during the loading process, as they need to retain their ability to process and present antigens to T cells to activate an immune response [105].
AIRISE-02, a NP loaded with CpG and siRNA targeting STAT3, enhances the immune response and inhibits the immunosuppressive TME. After intratumoral injection into tumors like melanoma and breast tumors, CpG stimulates local APCs, especially DCs, while si-STAT3 eliminates immunosuppressive pathways in cancer and bone marrow cells. The combined application of AIRISE-02 with ICIs has shown significantly better therapeutic effects than ICIs alone. Additionally, AIRISE-02 is effective in inhibiting tumor growth after intravenous injection and is currently entering the investigational new drug (IND) phase [114,115,116,117,118].
Overall, optimizing the TME and using DC vaccination with nanotechnology to activate antitumor immune responses are promising strategies for enhancing breast cancer therapies. Nanotechnology facilitates the targeted delivery of immunomodulatory agents, improving the maturation and function of dendritic cells within the TME.

3.4. Cancer-Associated Fibroblasts, CAFs

The stromal cells in the TME include vascular endothelial cells, pericytes, adipocytes, fibroblasts and bone marrow mesenchymal stromal cells. CAFs mediate fibroproliferative responses and serve as principal instigators of immunosuppressive microenvironments, leading to the failure of immunotherapy in solid tumors (Figure 4) [119,120]. The consumption of CAFs may enhance the efficacy of immunotherapy, which is currently under extensive investigation. Recent research has indicated that TGF-β inhibitors are capable of inhibiting the development of CAFs. For instance, the release of TGF-β inhibitors driven by heparanase (HPSE) at extracellular sites prevents tumor spreading by changing the TME, reducing CAFs activation and minimizing TGF-β production [121,122]. Inducing quiescence in CAFs through the use of angiotensin receptor blockers (ARBs) [123], such as valsartan, may prove to be a more effective strategy than depleting CAFs, which can potentially enhance tumor progression by promoting metastasis. The conjugation of Valsartan with a polyacetal polymer then precipitated into NPs, yielding the preparation of ARB NPs that were activated only in the TME. This approach effectively avoided the systemic adverse reactions induced by the ARB’s involvement in various physiological processes (Table 4).
Nanomedicines can modulate CAF activity and reduce the extracellular matrix (ECM) components to enhance the permeability of solid tumors to therapeutic agents and immune cells. At the tissue level, NPs are designed to penetrate the dense ECM. At the cellular level, they are functionalized with ligands that target fibroblast activation protein (FAP) on CAFs. This approach aims to compromise the tumor integrity and increase the treatment effectiveness, and the current research should focus on modulating CAF functions rather than depleting these cells to avoid increasing tumor aggressiveness.

3.5. Tumor-Associated Neutrophils, TANs

Neutrophils, known for their role in inflammation and immunity, display both tumor-promoting and tumor-suppressing functions. The antitumor N1 TANs enhance cytotoxic T cells responses, while the pro-tumor N2 TANs promote angiogenesis and suppress T cells. Targeting chemokines like CXCL1/2 can reduce pro-tumor neutrophils. Agents like TGF-β inhibitors can shift neutrophils to an antitumor phenotype, enhancing immunotherapy [131].
NPs loaded with TGF-β inhibitors can modulate neutrophils within tumors, enhancing local immune responses while minimizing systemic side effects. Furthermore, inhibiting neutrophil extracellular traps (NETs) with agents like DNase I improves PD-1 blockade immunotherapy in TNBC, enhancing CD8+ T cells infiltration and activity, thus overcoming resistance and achieving durable clinical responses [132,133].

3.6. ErbB/HER Signaling

In the molecular characteristics of BC, besides the amplification of the Her2/neu gene being associated with invasive BC and unfavorable prognosis, Her3 and Her4 belong to the ErbB receptor family and interact with Her2 to play a critical role in signal transduction, cell proliferation and survival in BC. Integrating these biomarkers into BC treatment strategies could lead to more precise and effective therapies [134].
Combining DC vaccination with agents targeting Her3 and Her4 may synergistically enhance antitumor immune responses, thereby improving outcomes. Bispecific antibodies targeting Her2 and Her3, or Her2 and Her4, are being developed to more effectively inhibit these signaling networks [135,136]. Furthermore, inhibitors of downstream pathways activated by Her3 and Her4, such as PI3K/Akt and MAPK, can be combined with these therapies to overcome resistance mechanisms [137].

3.7. Physical and Chemical Properties

The rapid proliferation of cancer cells causes glycolysis, resulting in an acidic pH at the tumor site. In the TME, the acidic pH allows the NPs to take advantage of the difference in pH between normal cells and cancer cells to target the tumor and achieve a controlled release of pH sensitivity, optimizing the biological distribution of the tumor site and reducing the therapeutic side effects [138,139]. Moreover, various activation technologies are used to enhance the efficacy of NPs conjugated with antitumor agents. For example, magnetic fields can induce temperature changes in lipids, facilitating magnetically triggered hyperthermia stimulation [140]. These magnetic liposomes have demonstrated significant antitumor activity against MDA-MB-231 cells [141].
In the TME, factors such as inadequate blood flow, elevated interstitial fluid pressure, heightened vascularity and interstitial obstruction have a pronounced impact on the permeability and passive targeting of drugs, inhibiting the antitumor efficacy. The enzyme-based NP drug delivery system uses different enzymes, including collagenase, hyaluronidase and matrix metalloproteinases, to degrade dense extracellular mechanisms [142,143]. This enhances the permeability of therapeutic drugs and NPs, enabling effective cancer targeted therapy by improving their access to tumor tissues. For example, AuNPs have shown the highest bioavailability and lowest cytotoxicity among various metals [144]. Gold NPs possess excellent biocompatibility and can be easily functionalized for targeted delivery, making them ideal for drug delivery and imaging applications [145].

4. Conclusions and Prospects

With the deepening of the overall understanding of tumors, it has been widely recognized that traditional oncogene therapy is insufficient to treat all patients. Combining or exclusively targeting immune cells and other components within the tumor microenvironment, by enhancing the body’s own immune response and promoting the antitumor effects of various components, can help improve the current treatment landscape and benefit the majority of cancer patients.
The targeted delivery of drugs to exert their effects is a crucial step in antitumor therapy. Efficiently delivering antitumor drugs to tumor tissues through the bloodstream has become a significant focus of new therapeutic strategies. However, the rapid clearance of nanomaterials by the reticuloendothelial system (RES) remains a critical challenge. Stealth nanocarriers, often modified with polyethylene glycol (PEG), aim to evade immune detection but still experience significant early clearance, leading to reduced efficacy. Pseudo-stealth nanocarriers, which exhibit partial stealth effects, offer some improvements but require further optimization. Future research should prioritize the engineering of nanoparticles to enhance their stealth properties, prolong the circulation time and improve bioavailability. Strategies such as optimizing surface coatings, exploring pathogen-mimetic approaches and adjusting physical characteristics like stiffness are essential to overcome RES-related challenges and maximize the therapeutic potential of nanomedicine in cancer treatment [146].
NP delivery systems offer several general advantages within the therapeutic landscape, which include (1) the co-delivery of multiple drugs: Different physicochemical properties of drugs can be accommodated by designing NPs with corresponding characteristics to achieve combination therapy [147,148]; (2) precise targeting: The EPR effect or ligand (e.g., antibodies or peptides) modification of NPs facilitates the accumulation of drug-loaded NPs in tumor tissues [149]; and (3) evasion of clearance: Modifying the external coating of NPs can reduce their recognition and clearance by the mononuclear phagocyte system (MPS) or enzymatic degradation. Additionally, NPs can reduce the required dosage of therapeutic agents, thereby decreasing the antitumor side effects and promoting immune responses. Immuno-oncological nanomedicine expands potential targets to the entire immune system or the overall ecosystem of the tumor host. During treatment, the synergistic work of each part of the innate and adaptive immunity influences the therapeutic efficacy. Increasing efforts are attempting to use combination therapies to mobilize different parts of the body, thereby improving treatment outcomes. Tumor nanotherapy combined with immunotherapy helps weaken the biological barrier effects of tumors and activates immune cells in small portions of the tumor or peripheral tissues to propagate the immune response, thereby more effectively enhancing targeted tumor cytotoxicity. Consequently, the demand for NPs in cancer treatment is increasing due to their exceptional performance in various aspects [150]. By integrating advanced genetic and immunotherapeutic strategies, NPs have the potential to achieve more efficient and precise tumor targeting and treatment, ultimately benefiting a broader range of cancer patients.
NPs are currently in various stages of development, with some already in clinical trials or approved for use. However, comprehensive studies on the toxicology or potential immune responses are still needed to ensure their safety and efficacy. This limitation is particularly evident in the evaluation of immunotherapy for BC, where there is a significant deficiency in tumor models. The most commonly used models are the 4T1 tumor cell model, spontaneous tumor models and patient-derived xenograft (PDX) models [151,152]. Despite their utility, these models have inherent limitations due to issues such as tumor heterogeneity and patient-specific differences, making it challenging to effectively demonstrate the safety and efficacy of nanotechnology-based immunotherapy. Therefore, while clinical advancements in NPs are promising, ongoing research and development are essential to address these challenges and optimize their application in cancer therapy.
Organoids, an emerging three-dimensional cell culture technology, have been shown to faithfully replicate the actual conditions of patients. BC organoids have been utilized for cocultures with various immune cells, creating a system that more accurately simulates the TME. This system is used to study cancer–immune interactions and evaluate the therapeutic efficacy. Research has demonstrated that NPs can effectively penetrate tumor 3D models, deliver antitumor drugs, activate immune cells and enhance the therapeutic efficacy [153].
Due to the heterogeneity (including intertumor and intratumor variations) of breast tumors, accurately predicting the targeting characteristics is challenging in clinical practice. Therefore, the modularity of enhanced therapeutic strategies can help fine tune formulations, allowing for formulation adjustments between individual patients in the treatment process to meet individualized needs.

Author Contributions

Conceptualization, L.Y., Q.H. and T.H.; writing—original draft preparation, L.Y. and Q.H.; writing—review and editing, Q.H. and T.H.; supervision, Q.H. and T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

All figures were created with BioRender.com (accessed on 3 March 2024).

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interests.

References

  1. Giaquinto, A.N.; Sung, H.; Miller, K.D.; Kramer, J.L.; Newman, L.A.; Minihan, A.; Jemal, A.; Siegel, R.L. Breast Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 524–541. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Arnold, M.; Morgan, E.; Rumgay, H.; Mafra, A.; Singh, D.; Laversanne, M.; Vignat, J.; Gralow, J.R.; Cardoso, F.; Siesling, S.; et al. Current and future burden of breast cancer: Global statistics for 2020 and 2040. Breast 2022, 66, 15–23. [Google Scholar] [CrossRef] [PubMed]
  4. Miller, K.D.; Nogueira, L.; Devasia, T.; Mariotto, A.B.; Yabroff, K.R.; Jemal, A.; Kramer, J.; Siegel, R.L. Cancer treatment and survivorship statistics, 2022. CA Cancer J. Clin. 2022, 72, 409–436. [Google Scholar] [CrossRef] [PubMed]
  5. Geyer, F.C.; Pareja, F.; Weigelt, B.; Rakha, E.; Ellis, I.O.; Schnitt, S.J.; Reis-Filho, J.S. The Spectrum of Triple-Negative Breast Disease: High- and Low-Grade Lesions. Am. J. Pathol. 2017, 187, 2139–2151. [Google Scholar] [CrossRef]
  6. Lee, J.S.; Yost, S.E.; Yuan, Y. Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges. Cancers 2020, 12, 1404. [Google Scholar] [CrossRef]
  7. Sharma, P.; López-Tarruella, S.; García-Saenz, J.A.; Khan, Q.J.; Gómez, H.L.; Prat, A.; Moreno, F.; Jerez-Gilarranz, Y.; Barnadas, A.; Picornell, A.C.; et al. Pathological Response and Survival in Triple-Negative Breast Cancer Following Neoadjuvant Carboplatin plus Docetaxel. Clin. Cancer Res. 2018, 24, 5820–5829. [Google Scholar] [CrossRef] [PubMed]
  8. Marra, A.; Viale, G.; Curigliano, G. Recent advances in triple negative breast cancer: The immunotherapy era. BMC Med. 2019, 17, 90. [Google Scholar] [CrossRef]
  9. Rui, R.; Zhou, L.; He, S. Cancer immunotherapies: Advances and bottlenecks. Front. Immunol. 2023, 14, 1212476. [Google Scholar] [CrossRef]
  10. Law, A.M.; Lim, E.; Ormandy, C.J.; Gallego-Ortega, D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr. Relat. Cancer 2017, 24, R123–R144. [Google Scholar] [CrossRef]
  11. Deepak, K.G.K.; Vempati, R.; Nagaraju, G.P.; Dasari, V.R.; Nagini, S.; Rao, D.N.; Malla, R.R. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020, 153, 104683. [Google Scholar] [CrossRef] [PubMed]
  12. Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef]
  13. Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386. [Google Scholar] [CrossRef] [PubMed]
  14. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  15. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  16. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  17. Galon, J.; Bruni, D. Approaches to treat immune hot altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
  18. Chen, Q.; Sun, T.; Jiang, C. Recent Advancements in Nanomedicine for ‘Cold’ Tumor Immunotherapy. Nanomicro Lett. 2021, 13, 92. [Google Scholar] [CrossRef]
  19. Broz, M.L.; Krummel, M.F. The emerging understanding of myeloid cells as partners and targets in tumor rejection. Cancer Immunol. Res. 2015, 3, 313–319. [Google Scholar] [CrossRef]
  20. DeVito, N.C.; Plebanek, M.P.; Theivanthiran, B.; Hanks, B.A. Role of Tumor-Mediated Dendritic Cell Tolerization in Immune Evasion. Front. Immunol. 2019, 10, 2876. [Google Scholar] [CrossRef]
  21. Corzo, C.A.; Condamine, T.; Lu, L.; Cotter, M.J.; Youn, J.I.; Cheng, P.; Cho, H.I.; Celis, E.; Quiceno, D.G.; Padhya, T.; et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 2010, 207, 2439–2453. [Google Scholar] [CrossRef] [PubMed]
  22. Gil Del Alcazar, C.R.; Huh, S.J.; Ekram, M.B.; Trinh, A.; Liu, L.L.; Beca, F.; Zi, X.; Kwak, M.; Bergholtz, H.; Su, Y.; et al. Immune Escape in Breast Cancer During In Situ to Invasive Carcinoma Transition. Cancer Discov. 2017, 7, 1098–1115. [Google Scholar] [CrossRef]
  23. Jerby-Arnon, L.; Shah, P.; Cuoco, M.S.; Rodman, C.; Su, M.J.; Melms, J.C.; Leeson, R.; Kanodia, A.; Mei, S.; Lin, J.R.; et al. A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade. Cell 2018, 175, 984–997. [Google Scholar] [CrossRef] [PubMed]
  24. Jayaprakash, P.; Ai, M.; Liu, A.; Budhani, P.; Bartkowiak, T.; Sheng, J.; Ager, C.; Nicholas, C.; Jaiswal, A.R.; Sun, Y.; et al. Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy. J. Clin. Investig. 2018, 128, 5137–5149. [Google Scholar] [CrossRef] [PubMed]
  25. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+ T Cells to protect tumour cells. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef]
  26. Kato, T.; Noma, K.; Ohara, T.; Kashima, H.; Katsura, Y.; Sato, H.; Komoto, S.; Katsube, R.; Ninomiya, T.; Tazawa, H.; et al. Cancer-Associated Fibroblasts Affect Intratumoral CD8+ and FoxP3+ T Cells Via IL6 in the Tumor Microenvironment. Clin. Cancer Res. 2018, 24, 4820–4833. [Google Scholar] [CrossRef]
  27. Kumar, V.; Donthireddy, L.; Marvel, D.; Condamine, T.; Wang, F.; Lavilla-Alonso, S.; Hashimoto, A.; Vonteddu, P.; Behera, R.; Goins, M.A.; et al. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017, 32, 654–668. [Google Scholar] [CrossRef] [PubMed]
  28. Butcher, D.T.; Alliston, T.; Weaver, V.M. A tense situation: Forcing tumour progression. Nat. Rev. Cancer 2009, 9, 108–122. [Google Scholar] [CrossRef] [PubMed]
  29. Emon, B.; Bauer, J.; Jain, Y.; Jung, B.; Saif, T. Biophysics of Tumor Microenvironment and Cancer Metastasis—A Mini Review. Comput. Struct. Biotechnol. J. 2018, 16, 279–287. [Google Scholar] [CrossRef]
  30. Lu, P.; Weaver, V.M.; Werb, Z. The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol. 2012, 196, 395–406. [Google Scholar] [CrossRef]
  31. Samani, A.; Zubovits, J.; Plewes, D. Elastic moduli of normal and pathological human breast tissues: An inversion-technique-based investigation of 169 samples. Phys. Med. Biol. 2007, 52, 1565–1576. [Google Scholar] [CrossRef] [PubMed]
  32. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef] [PubMed]
  33. Pickup, M.W.; Mouw, J.K.; Weaver, V.M. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014, 15, 1243–1253. [Google Scholar] [CrossRef] [PubMed]
  34. Van Goethem, E.; Poincloux, R.; Gauffre, F.; Maridonneau-Parini, I.; Le Cabec, V. Matrix architecture dictates three-dimensional migration modes of human macrophages: Differential involvement of proteases and podosome-like structures. J. Immunol. 2010, 184, 1049–1061. [Google Scholar] [CrossRef] [PubMed]
  35. McWhorter, F.Y.; Davis, C.T.; Liu, W.F. Physical and mechanical regulation of macrophage phenotype and function. Cell. Mol. Life Sci. 2015, 72, 1303–1316. [Google Scholar] [CrossRef] [PubMed]
  36. Estrella, V.; Chen, T.; Lloyd, M.; Wojtkowiak, J.; Cornnell, H.H.; Ibrahim-Hashim, A.; Bailey, K.; Balagurunathan, Y.; Rothberg, J.M.; Sloane, B.F.; et al. Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 2013, 73, 1524–1535. [Google Scholar] [CrossRef]
  37. Shi, Q.; Le, X.; Wang, B.; Abbruzzese, J.L.; Xiong, Q.; He, Y.; Xie, K. Regulation of vascular endothelial growth factor expression by acidosis in human cancer cells. Oncogene 2001, 20, 3751–3756. [Google Scholar] [CrossRef]
  38. Walton, Z.E.; Patel, C.H.; Brooks, R.C.; Yu, Y.; Ibrahim-Hashim, A.; Riddle, M.; Porcu, A.; Jiang, T.; Ecker, B.L.; Tameire, F.; et al. Acid Suspends the Circadian Clock in Hypoxia through Inhibition of mTOR. Cell 2018, 174, 72–87. [Google Scholar] [CrossRef]
  39. Shah, L.; Latif, A.; Williams, K.J.; Tirella, A. Role of stiffness and physico-chemical properties of tumour microenvironment on breast cancer cell stemness. Acta Biomater. 2022, 152, 273–289. [Google Scholar] [CrossRef]
  40. Erra Díaz, F.; Dantas, E.; Geffner, J. Unravelling the Interplay between Extracellular Acidosis and Immune Cells. Mediators Inflamm. 2018, 2018, 1218297. [Google Scholar] [CrossRef]
  41. Pantelidou, C.; Sonzogni, O.; De Oliveria Taveira, M.; Mehta, A.K.; Kothari, A.; Wang, D.; Visal, T.; Li, M.K.; Pinto, J.; Castrillon, J.A.; et al. PARP Inhibitor Efficacy Depends on CD8+ T-cell Recruitment via Intratumoral STING Pathway Activation in BRCA-Deficient Models of Triple-Negative Breast Cancer. Cancer Discov. 2019, 9, 722–737. [Google Scholar] [CrossRef] [PubMed]
  42. Disis, M.L.; Dang, Y.; Coveler, A.L.; Childs, J.S.; Higgins, D.M.; Liu, Y.; Zhou, J.; Mackay, S.; Salazar, L.G. A Phase I/II Trial of HER2 Vaccine-Primed Autologous T-Cell Infusions in Patients with Treatment Refractory HER2-Overexpressing Breast Cancer. Clin. Cancer Res. 2023, 29, 3362–3371. [Google Scholar] [CrossRef] [PubMed]
  43. Chaib, M.; Sipe, L.M.; Yarbro, J.R.; Bohm, M.S.; Counts, B.R.; Tanveer, U.; Pingili, A.K.; Daria, D.; Marion, T.N.; Carson, J.A.; et al. PKC agonism restricts innate immune suppression promotes antigen cross-presentation and synergizes with agonistic CD40 antibody therapy to activate CD8+ T cells in breast cancer. Cancer Lett. 2022, 531, 98–108. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, Y.N.; Lee, H.H.; Jiang, Z.; Chan, L.C.; Hortobagyi, G.N.; Yu, D.; Hung, M.C. Ribonuclease 1 Enhances Antitumor Immunity against Breast Cancer by Boosting T cell Activation. Int. J. Biol. Sci. 2023, 19, 2957–2973. [Google Scholar] [CrossRef] [PubMed]
  45. Torres, E.T.R.; Emens, L.A. Emerging combination immunotherapy strategies for breast cancer: Dual immune checkpoint modulation antibody-drug conjugates and bispecific antibodies. Breast Cancer Res. Treat. 2022, 191, 291–302. [Google Scholar] [CrossRef] [PubMed]
  46. Rius Ruiz, I.; Vicario, R.; Morancho, B.; Morales, C.B.; Arenas, E.J.; Herter, S.; Freimoser-Grundschober, A.; Somandin, J.; Sam, J.; Ast, O.; et al. p95HER2-T cell bispecific antibody for breast cancer treatment. Sci. Transl. Med. 2018, 10, eaat1445. [Google Scholar] [CrossRef] [PubMed]
  47. Zhu, Y.; Liu, K.; Wang, K.; Zhu, H. Treatment-related adverse events of antibody-drug conjugates in clinical trials: A systematic review and meta-analysis. Cancer 2023, 129, 283–295. [Google Scholar] [CrossRef] [PubMed]
  48. Copeland-Halperin, R.S.; Liu, J.E.; Yu, A.F. Cardiotoxicity of HER2-targeted therapies. Curr. Opin. Cardiol. 2019, 34, 451–458. [Google Scholar] [CrossRef] [PubMed]
  49. Scaltriti, M.; Rojo, F.; Ocaña, A.; Anido, J.; Guzman, M.; Cortes, J.; Di Cosimo, S.; Matias-Guiu, X.; Ramon y Cajal, S.; Arribas, J.; et al. Expression of p95HER2 a truncated form of the HER2 receptor and response to anti-HER2 therapies in breast cancer. J. Natl. Cancer Inst. 2007, 99, 628–638. [Google Scholar] [CrossRef]
  50. Liu, H.; Bai, L.; Huang, L.; Ning, N.; Li, L.; Li, Y.; Dong, X.; Du, Q.; Xia, M.; Chen, Y.; et al. Bispecific antibody targeting TROP2xCD3 suppresses tumor growth of triple negative breast cancer. J. Immunother. Cancer 2021, 9, e003468. [Google Scholar] [CrossRef]
  51. Mittal, D.; Vijayan, D.; Neijssen, J.; Kreijtz, J.; Habraken, M.M.J.M.; Van Eenennaam, H.; Van Elsas, A.; Smyth, M.J. Blockade of ErbB2 and PD-L1 using a bispecific antibody to improve targeted anti-ErbB2 therapy. Oncoimmunology 2019, 8, e1648171. [Google Scholar] [CrossRef] [PubMed]
  52. Cheng, Y.A.; Wu, T.H.; Wang, Y.M.; Cheng, T.L.; Chen, I.J.; Lu, Y.C.; Chuang, K.H.; Wang, C.K.; Chen, C.Y.; Lin, R.A.; et al. Humanized bispecific antibody (mPEG × HER2) rapidly confers PEGylated nanoparticles tumor specificity for multimodality imaging in breast cancer. J. Nanobiotechnol. 2020, 18, 118. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, B.; Shi, J.; Shi, X.; Xu, X.; Gao, L.; Li, S.; Liu, M.; Gao, M.; Jin, S.; Zhou, J.; et al. Development and evaluation of a human CD47/HER2 bispecific antibody for Trastuzumab-resistant breast cancer immunotherapy. Drug Resist. Updates 2024, 74, 101068. [Google Scholar] [CrossRef] [PubMed]
  54. de Vries Schultink, A.H.M.; Doornbos, R.P.; Bakker, A.B.H.; Bol, K.; Throsby, M.; Geuijen, C.; Maussang, D.; Schellens, J.H.M.; Beijnen, J.H.; Huitema, A.D.R. Translational PK-PD modeling analysis of MCLA-128 a HER2/HER3 bispecific monoclonal antibody to predict clinical efficacious exposure and dose. Investig. New Drugs 2018, 36, 1006–1015. [Google Scholar] [CrossRef] [PubMed]
  55. Rau, A.; Lieb, W.S.; Seifert, O.; Honer, J.; Birnstock, D.; Richter, F.; Aschmoneit, N.; Olayioye, M.A.; Kontermann, R.E. Inhibition of Tumor Cell Growth and Cancer Stem Cell Expansion by a Bispecific Antibody Targeting EGFR and HER3. Mol. Cancer Ther. 2020, 19, 1474–1485. [Google Scholar] [CrossRef] [PubMed]
  56. Greiling, T.M.; Dehner, C.; Chen, X.; Hughes, K.; Iñiguez, A.J.; Boccitto, M.; Ruiz, D.Z.; Renfroe, S.C.; Vieira, S.M.; Ruff, W.E.; et al. Commensal orthologs of the human autoantigen Ro60 as triggers of autoimmunity in lupus. Sci. Transl. Med. 2018, 10, eaan2306. [Google Scholar] [CrossRef] [PubMed]
  57. Chang, C.H.; Wang, Y.; Li, R.; Rossi, D.L.; Liu, D.; Rossi, E.A.; Cardillo, T.M.; Goldenberg, D.M. Combination Therapy with Bispecific Antibodies and PD-1 Blockade Enhances the Antitumor Potency of T Cells. Cancer Res. 2017, 77, 5384–5394. [Google Scholar] [CrossRef] [PubMed]
  58. Kamada, H.; Taki, S.; Nagano, K.; Inoue, M.; Ando, D.; Mukai, Y.; Higashisaka, K.; Yoshioka, Y.; Tsutsumi, Y.; Tsunoda, S. Generation and characterization of a bispecific diabody targeting both EPH receptor A10 and CD3. Biochem. Biophys. Res. Commun. 2015, 456, 908–912. [Google Scholar] [CrossRef]
  59. Burges, A.; Wimberger, P.; Kümper, C.; Gorbounova, V.; Sommer, H.; Schmalfeldt, B.; Pfisterer, J.; Lichinitser, M.; Makhson, A.; Moiseyenko, V.; et al. Effective relief of malignant ascites in patients with advanced ovarian cancer by a trifunctional anti-EpCAM x anti-CD3 antibody: A phase I/II study. Clin. Cancer Res. 2007, 13, 3899–3905. [Google Scholar] [CrossRef]
  60. Hong, R.; Zhou, Y.; Tian, X.; Wang, L.; Wu, X. Selective inhibition of IDO1 D-1-methyl-tryptophan (D-1MT) effectively increased EpCAM/CD3-bispecific BiTE antibody MT110 efficacy against IDO1hi breast cancer via enhancing immune cells activity. Int. Immunopharmacol. 2018, 54, 118–124. [Google Scholar] [CrossRef]
  61. Fisher, T.S.; Hooper, A.T.; Lucas, J.; Clark, T.H.; Rohner, A.K.; Peano, B.; Elliott, M.W.; Tsaparikos, K.; Wang, H.; Golas, J.; et al. A CD3-bispecific molecule targeting P-cadherin demonstrates T cell-mediated regression of established solid tumors in mice. Cancer Immunol. Immunother. 2018, 67, 247–259. [Google Scholar] [CrossRef] [PubMed]
  62. Cheng, Q.; Shi, X.; Han, M.; Smbatyan, G.; Lenz, H.J.; Zhang, Y. Reprogramming Exosomes as Nanoscale Controllers of Cellular Immunity. J. Am. Chem. Soc. 2018, 140, 16413–16417. [Google Scholar] [CrossRef] [PubMed]
  63. Tang, J.; Li, B.; Howard, C.B.; Mahler, S.M.; Thurecht, K.J.; Wu, Y.; Huang, L.; Xu, Z.P. Multifunctional lipid-coated calcium phosphate nanoplatforms for complete inhibition of large triple negative breast cancer via targeted combined therapy. Biomaterials 2019, 216, 119232. [Google Scholar] [CrossRef] [PubMed]
  64. Fu, W.; Lei, C.; Yu, Y.; Liu, S.; Li, T.; Lin, F.; Fan, X.; Shen, Y.; Ding, M.; Tang, Y.; et al. EGFR/Notch Antagonists Enhance the Response to Inhibitors of the PI3K-Akt Pathway by Decreasing Tumor-Initiating Cell Frequency. Clin. Cancer Res. 2019, 25, 2835–2847. [Google Scholar] [CrossRef] [PubMed]
  65. Del Bano, J.; Florès-Florès, R.; Josselin, E.; Goubard, A.; Ganier, L.; Castellano, R.; Chames, P.; Baty, D.; Kerfelec, B. A Bispecific Antibody-Based Approach for Targeting Mesothelin in Triple Negative Breast Cancer. Front. Immunol. 2019, 10, 1593. [Google Scholar] [CrossRef] [PubMed]
  66. Li, Y.; Zhou, C.; Li, J.; Liu, J.; Lin, L.; Li, L.; Cao, D.; Li, Q.; Wang, Z. Single domain based bispecific antibody Muc1-Bi-1 and its humanized form Muc1-Bi-2 induce potent cancer cell killing in muc1 positive tumor cells. PLoS ONE 2018, 13, e0191024. [Google Scholar] [CrossRef] [PubMed]
  67. Berezhnoy, A.; Sumrow, B.J.; Stahl, K.; Shah, K.; Liu, D.; Li, J.; Hao, S.S.; De Costa, A.; Kaul, S.; Bendell, J.; et al. Development and Preliminary Clinical Activity of PD-1-Guided CTLA-4 Blocking Bispecific DART Molecule. Cell Rep. Med. 2020, 1, 100163. [Google Scholar] [CrossRef] [PubMed]
  68. Kelly, K.; Manitz, J.; Patel, M.R.; D’Angelo, S.P.; Apolo, A.B.; Rajan, A.; Kasturi, V.; Speit, I.; Bajars, M.; Warth, J.; et al. Efficacy and immune-related adverse event associations in avelumab-treated patients. J. Immunother. Cancer 2020, 8, e001427. [Google Scholar] [CrossRef] [PubMed]
  69. Strauss, J.; Heery, C.R.; Schlom, J.; Madan, R.A.; Cao, L.; Kang, Z.; Lamping, E.; Marté, J.L.; Donahue, R.N.; Grenga, I.; et al. Phase I Trial of M7824 (MSB0011359C) a Bifunctional Fusion Protein Targeting PD-L1 and TGFβ in Advanced Solid Tumors. Clin. Cancer Res. 2018, 24, 1287–1295. [Google Scholar] [CrossRef]
  70. Strauss, J.; Gatti-Mays, M.E.; Cho, B.C.; Hill, A.; Salas, S.; McClay, E.; Redman, J.M.; Sater, H.A.; Donahue, R.N.; Jochems, C.; et al. Bintrafusp alfa a bifunctional fusion protein targeting TGF-β and PD-L1 in patients with human papillomavirus-associated malignancies. J. Immunother. Cancer 2020, 8, e001395. [Google Scholar] [CrossRef]
  71. Xie, Y.Q.; Arik, H.; Wei, L.; Zheng, Y.; Suh, H.; Irvine, D.J.; Tang, L. Redox-responsive interleukin-2 nanogel specifically and safely promotes the proliferation and memory precursor differentiation of tumor-reactive T-cells. Biomater. Sci. 2019, 7, 1345–1357. [Google Scholar] [CrossRef] [PubMed]
  72. Xue, D.; Moon, B.; Liao, J.; Guo, J.; Zou, Z.; Han, Y.; Cao, S.; Wang, Y.; Fu, Y.X.; Peng, H. A tumor-specific pro-IL-12 activates preexisting cytotoxic T cells to control established tumors. Sci. Immunol. 2022, 7, eabi6899. [Google Scholar] [CrossRef] [PubMed]
  73. Tang, L.; Zheng, Y.; Melo, M.B.; Mabardi, L.; Castaño, A.P.; Xie, Y.Q.; Li, N.; Kudchodkar, S.B.; Wong, H.C.; Jeng, E.K.; et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 2018, 36, 707–716. [Google Scholar] [CrossRef] [PubMed]
  74. Li, J.; Kataoka, K. Chemo-physical Strategies to Advance the in Vivo Functionality of Targeted Nanomedicine: The Next Generation. J. Am. Chem. Soc. 2021, 143, 538–559. [Google Scholar] [CrossRef] [PubMed]
  75. Covarrubias, G.; Moon, T.J.; Loutrianakis, G.; Sims, H.M.; Umapathy, M.P.; Lorkowski, M.E.; Bielecki, P.A.; Wiese, M.L.; Atukorale, P.U.; Karathanasis, E. Comparison of the uptake of untargeted and targeted immunostimulatory nanoparticles by immune cells in the microenvironment of metastatic breast cancer. J. Mater. Chem. B 2022, 10, 224–235. [Google Scholar] [CrossRef]
  76. Qiu, X.; Zhao, T.; Luo, R.; Qiu, R.; Li, Z. Tumor-Associated Macrophages: Key Players in Triple-Negative Breast Cancer. Front. Oncol. 2022, 12, 772615. [Google Scholar] [CrossRef]
  77. Lu, H.; Clauser, K.R.; Tam, W.L.; Fröse, J.; Ye, X.; Eaton, E.N.; Reinhardt, F.; Donnenberg, V.S.; Bhargava, R.; Carr, S.A.; et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat. Cell Biol. 2014, 16, 1105–1117. [Google Scholar] [CrossRef]
  78. Yang, J.; Liao, D.; Chen, C.; Liu, Y.; Chuang, T.H.; Xiang, R.; Markowitz, D.; Reisfeld, R.A.; Luo, Y. Tumor-associated macrophages regulate murine breast cancer stem cells through a novel paracrine EGFR/Stat3/Sox-2 signaling pathway. Stem Cells 2013, 31, 248–258. [Google Scholar] [CrossRef]
  79. Fu, L.Q.; Du, W.L.; Cai, M.H.; Yao, J.Y.; Zhao, Y.Y.; Mou, X.Z. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 2020, 353, 104119. [Google Scholar] [CrossRef]
  80. Chen, D.; Zhang, X.; Li, Z.; Zhu, B. Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics 2021, 11, 1016–1030. [Google Scholar] [CrossRef]
  81. Komohara, Y.; Jinushi, M.; Takeya, M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 2014, 105, 1–8. [Google Scholar] [CrossRef] [PubMed]
  82. Kitamura, T.; Qian, B.Z.; Soong, D.; Cassetta, L.; Noy, R.; Sugano, G.; Kato, Y.; Li, J.; Pollard, J.W. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J. Exp. Med. 2015, 212, 1043–1059. [Google Scholar] [CrossRef]
  83. Nie, Y.; Huang, H.; Guo, M.; Chen, J.; Wu, W.; Li, W.; Xu, X.; Lin, X.; Fu, W.; Yao, Y.; et al. Breast Phyllodes Tumors Recruit and Repolarize Tumor-Associated Macrophages via Secreting CCL5 to Promote Malignant Progression Which Can Be Inhibited by CCR5 Inhibition Therapy. Clin. Cancer Res. 2019, 25, 3873–3886. [Google Scholar] [CrossRef]
  84. Oh, S.A.; Li, M.O. TGF-β: Guardian of T cell function. J. Immunol. 2013, 191, 3973–3979. [Google Scholar] [CrossRef] [PubMed]
  85. Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.T.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef] [PubMed]
  86. Ramesh, A.; Kumar, S.; Nandi, D.; Kulkarni, A. CSF1R- and SHP2-Inhibitor-Loaded Nanoparticles Enhance Cytotoxic Activity and Phagocytosis in Tumor-Associated Macrophages. Adv. Mater. 2019, 31, 1904364. [Google Scholar] [CrossRef]
  87. Molgora, M.; Colonna, M. Turning enemies into allies-reprogramming tumor-associated macrophages for cancer therapy. Med 2021, 2, 666–681. [Google Scholar] [CrossRef]
  88. Wanderley, C.W.; Colón, D.F.; Luiz, J.P.M.; Oliveira, F.F.; Viacava, P.R.; Leite, C.A.; Pereira, J.A.; Silva, C.M.; Silva, C.R.; Silva, R.L.; et al. Paclitaxel Reduces Tumor Growth by Reprogramming Tumor-Associated Macrophages to an M1 Profile in a TLR4-Dependent Manner. Cancer Res. 2018, 78, 5891–5900. [Google Scholar] [CrossRef]
  89. Xie, R.; Ruan, S.; Liu, J.; Qin, L.; Yang, C.; Tong, F.; Lei, T.; Shevtsov, M.; Gao, H.; Qin, Y. Furin-instructed aggregated gold nanoparticles for re-educating tumor associated macrophages and overcoming breast cancer chemoresistance. Biomaterials 2021, 275, 120891. [Google Scholar] [CrossRef]
  90. Esser, A.K.; Ross, M.H.; Fontana, F.; Su, X.; Gabay, A.; Fox, G.C.; Xu, Y.; Xiang, J.; Schmieder, A.H.; Yang, X.; et al. Nanotherapy delivery of c-myc inhibitor targets Pro-tumor Macrophages and preserves Antitumor Macrophages in Breast Cancer. Theranostics 2020, 10, 7510–7526. [Google Scholar] [CrossRef]
  91. Deng, C.; Zhang, Q.; Jia, M.; Zhao, J.; Sun, X.; Gong, T.; Zhang, Z. Tumors and Their Microenvironment Dual-Targeting Chemotherapy with Local Immune Adjuvant Therapy for Effective Antitumor Immunity against Breast Cancer. Adv. Sci. 2019, 6, 1801868. [Google Scholar] [CrossRef]
  92. Niu, M.; Valdes, S.; Naguib, Y.W.; Hursting, S.D.; Cui, Z. Tumor-Associated Macrophage-Mediated Targeted Therapy of Triple-Negative Breast Cancer. Mol. Pharm. 2016, 13, 1833–1842. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, T.; Lip, H.; He, C.; Cai, P.; Wang, Z.; Henderson, J.T.; Rauth, A.M.; Wu, X.Y. Multitargeted Nanoparticles Deliver Synergistic Drugs across the Blood-Brain Barrier to Brain Metastases of Triple Negative Breast Cancer Cells and Tumor-Associated Macrophages. Adv. Healthc. Mater. 2019, 8, 1900543. [Google Scholar] [CrossRef] [PubMed]
  94. Pawar, V.K.; Singh, Y.; Sharma, K.; Shrivastav, A.; Sharma, A.; Singh, A.; Meher, J.G.; Singh, P.; Raval, K.; Bora, H.K.; et al. Doxorubicin Hydrochloride Loaded Zymosan-Polyethylenimine Biopolymeric Nanoparticles for Dual ‘Chemoimmunotherapeutic’ Intervention in Breast Cancer. Pharm. Res. 2017, 34, 1857–1871. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, M.; Yan, L.; Kim, J.A. Modulating mammary tumor growth metastasis and immunosuppression by siRNA-induced MIF reduction in tumor microenvironment. Cancer Gene Ther. 2015, 22, 463–474. [Google Scholar] [CrossRef]
  96. Shen, S.; Zhang, Y.; Chen, K.G.; Luo, Y.L.; Wang, J. Cationic Polymeric Nanoparticle Delivering CCR2 siRNA to Inflammatory Monocytes for Tumor Microenvironment Modification and Cancer Therapy. Mol. Pharm. 2018, 15, 3642–3653. [Google Scholar] [CrossRef] [PubMed]
  97. Song, Y.; Tang, C.; Yin, C. Combination antitumor immunotherapy with VEGF and PIGF siRNA via systemic delivery of multifunctionalized nanoparticles to tumor-associated macrophages and breast cancer cells. Biomaterials 2018, 185, 117–132. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, X.; Tian, W.; Cai, X.; Wang, X.; Dang, W.; Tang, H.; Cao, H.; Wang, L.; Chen, T. Hydrazinocurcumin Encapsuled nanoparticles ‘re-educate’ tumor-associated macrophages and exhibit anti-tumor effects on breast cancer following STAT3 suppression. PLoS ONE 2013, 8, e65896. [Google Scholar] [CrossRef]
  99. Chong, L.; Jiang, Y.W.; Wang, D.; Chang, P.; Xu, K.; Li, J. Targeting and repolarizing M2-like tumor-associated macrophage-mediated MR imaging and tumor immunotherapy by biomimetic nanoparticles. J. Nanobiotechnol. 2023, 21, 401. [Google Scholar] [CrossRef]
  100. Cai, X.J.; Wang, Z.; Cao, J.W.; Ni, J.J.; Xu, Y.Y.; Yao, J.; Xu, H.; Liu, F.; Yang, G.Y. Anti-angiogenic and anti-tumor effects of metronomic use of novel liposomal zoledronic acid depletes tumor-associated macrophages in triple negative breast cancer. Oncotarget 2017, 8, 84248–84257. [Google Scholar] [CrossRef]
  101. Hu, Y.; Manasrah, B.K.; McGregor, S.M.; Lera, R.F.; Norman, R.X.; Tucker, J.B.; Scribano, C.M.; Yan, R.E.; Humayun, M.; Wisinski, K.B.; et al. Paclitaxel Induces Micronucleation and Activates Pro-Inflammatory cGAS-STING Signaling in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2021, 20, 2553–2567. [Google Scholar] [CrossRef] [PubMed]
  102. Kim, S.H.; Kim, S.J.; Park, J.; Joe, Y.; Lee, S.E.; Saeidi, S.; Zhong, X.; Kim, S.H.; Park, S.A.; Na, H.K.; et al. Reprograming of Tumor-Associated Macrophages in Breast Tumor-Bearing Mice under Chemotherapy by Targeting Heme Oxygenase-1. Antioxidants 2021, 10, 470. [Google Scholar] [CrossRef]
  103. Gardner, A.; de Mingo Pulido, Á.; Ruffell, B. Dendritic Cells and Their Role in Immunotherapy. Front. Immunol. 2020, 11, 924. [Google Scholar] [CrossRef] [PubMed]
  104. Reddy, S.T.; Swartz, M.A.; Hubbell, J.A. Targeting dendritic cells with biomaterials: Developing the next generation of vaccines. Trends Immunol. 2006, 27, 573–579. [Google Scholar] [CrossRef]
  105. Szpor, J.; Streb, J.; Glajcar, A.; Frączek, P.; Winiarska, A.; Tyrak, K.E.; Basta, P.; Okoń, K.; Jach, R.; Hodorowicz-Zaniewska, D. Dendritic Cells Are Associated with Prognosis and Survival in Breast Cancer. Diagnostics 2021, 11, 702. [Google Scholar] [CrossRef]
  106. Zhang, X.; Wu, F.; Men, K.; Huang, R.; Zhou, B.; Zhang, R.; Zou, R.; Yang, L. Modified Fe3O4 Magnetic Nanoparticle Delivery of CpG Inhibits Tumor Growth and Spontaneous Pulmonary Metastases to Enhance Immunotherapy. Nanoscale Res. Lett. 2018, 13, 240. [Google Scholar] [CrossRef]
  107. He, Y.; Wang, M.; Li, X.; Yu, T.; Gao, X. Targeted MIP-3β plasmid nanoparticles induce dendritic cell maturation and inhibit M2 macrophage polarisation to suppress cancer growth. Biomaterials 2020, 249, 120046. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, S.; Pang, G.; Chen, C.; Qin, J.; Yu, H.; Liu, Y.; Zhang, X.; Song, Z.; Zhao, J.; Wang, F.; et al. Effective cancer immunotherapy by Ganoderma lucidum polysaccharide-gold nanocomposites through dendritic cell activation and memory T cell response. Carbohydr. Polym. 2019, 205, 192–202. [Google Scholar] [CrossRef]
  109. Jadidi-Niaragh, F.; Atyabi, F.; Rastegari, A.; Kheshtchin, N.; Arab, S.; Hassannia, H.; Ajami, M.; Mirsanei, Z.; Habibi, S.; Masoumi, F.; et al. CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. J. Control. Release 2017, 246, 46–59. [Google Scholar] [CrossRef]
  110. Liu, Y.; Qiao, L.; Zhang, S.; Wan, G.; Chen, B.; Zhou, P.; Zhang, N.; Wang, Y. Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy. Acta Biomater. 2018, 66, 310–324. [Google Scholar] [CrossRef]
  111. Wu, T.; Qiao, Q.; Qin, X.; Zhang, D.; Zhang, Z. Immunostimulatory cytokine and doxorubicin co-loaded nanovesicles for cancer immunochemotherapy. Nanomedicine 2019, 18, 66–77. [Google Scholar] [CrossRef] [PubMed]
  112. Zheng, D.W.; Chen, J.L.; Zhu, J.Y.; Rong, L.; Li, B.; Lei, Q.; Fan, J.X.; Zou, M.Z.; Li, C.; Cheng, S.X.; et al. Highly Integrated Nano-Platform for Breaking the Barrier between Chemotherapy and Immunotherapy. Nano Lett. 2016, 16, 4341–4347. [Google Scholar] [CrossRef] [PubMed]
  113. Ni, J.; Song, J.; Wang, B.; Hua, H.; Zhu, H.; Guo, X.; Xiong, S.; Zhao, Y. Dendritic cell vaccine for the effective immunotherapy of breast cancer. Biomed. Pharmacother. 2020, 126, 110046. [Google Scholar] [CrossRef] [PubMed]
  114. Cai, Q.; Kublo, L.; Cumberland, R.; Gooding, W.; Baar, J. Optimized systemic dosing with CpG DNA enhances dendritic cell-mediated rejection of a poorly immunogenic mammary tumor in BALB/c mice. Clin. Transl. Sci. 2009, 2, 62–66. [Google Scholar] [CrossRef] [PubMed]
  115. Noh, J.Y.; Yoon, S.R.; Kim, T.D.; Choi, I.; Jung, H. Toll-Like Receptors in Natural Killer Cells and Their Application for Immunotherapy. J. Immunol. Res. 2020, 2020, 2045860. [Google Scholar] [CrossRef] [PubMed]
  116. Ballas, Z.K. Modulation of NK cell activity by CpG oligodeoxynucleotides. Immunol. Res. 2007, 39, 15–21. [Google Scholar] [CrossRef] [PubMed]
  117. Hodge, D.R.; Hurt, E.M.; Farrar, W.L. The role of IL-6 and STAT3 in inflammation and cancer. Eur. J. Cancer 2005, 41, 2502–2512. [Google Scholar] [CrossRef] [PubMed]
  118. Ngamcherdtrakul, W.; Reda, M.; Nelson, M.A.; Wang, R.; Zaidan, H.Y.; Bejan, D.S.; Hoang, N.H.; Lane, R.S.; Luoh, S.W.; Leachman, S.A.; et al. In Situ Tumor Vaccination with Nanoparticle Co-Delivering CpG and STAT3 siRNA to Effectively Induce Whole-Body Antitumor Immune Response. Adv. Mater. 2021, 33, 2100628. [Google Scholar] [CrossRef] [PubMed]
  119. Liu, T.; Han, C.; Wang, S.; Fang, P.; Ma, Z.; Xu, L.; Yin, R. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. J. Hematol. Oncol. 2019, 12, 86. [Google Scholar] [CrossRef]
  120. Pei, L.; Liu, Y.; Liu, L.; Gao, S.; Gao, X.; Feng, Y.; Sun, Z.; Zhang, Y.; Wang, C. Roles of cancer-associated fibroblasts (CAFs) in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol. Cancer 2023, 22, 29. [Google Scholar] [CrossRef]
  121. Zhang, J.; Yang, J.; Zuo, T.; Ma, S.; Xokrat, N.; Hu, Z.; Wang, Z.; Xu, R.; Wei, Y.; Shen, Q. Heparanase-driven sequential released nanoparticles for ferroptosis and tumor microenvironment modulations synergism in breast cancer therapy. Biomaterials 2021, 266, 120429. [Google Scholar] [CrossRef]
  122. Zhang, J.; Zuo, T.; Yang, J.; Hu, Z.; Wang, Z.; Xu, R.; Ma, S.; Wei, Y.; Shen, Q. Hierarchically Releasing Bio-Responsive Nanoparticles for Complete Tumor Microenvironment Modulation via TGF-β Pathway Inhibition and TAF Reduction. ACS Appl. Mater. Interfaces 2021, 13, 2256–2268. [Google Scholar] [CrossRef] [PubMed]
  123. Chauhan, V.P.; Chen, I.X.; Tong, R.; Ng, M.R.; Martin, J.D.; Naxerova, K.; Wu, M.W.; Huang, P.; Boucher, Y.; Kohane, D.S.; et al. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2019, 116, 10674–10680. [Google Scholar] [CrossRef]
  124. Ji, T.; Zhao, Y.; Ding, Y.; Wang, J.; Zhao, R.; Lang, J.; Qin, H.; Liu, X.; Shi, J.; Tao, N.; et al. Transformable Peptide Nanocarriers for Expeditious Drug Release and Effective Cancer Therapy via Cancer-Associated Fibroblast Activation. Angew. Chem. Int. Ed. Engl. 2016, 55, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, Y.; Yu, F.; Tan, Y.; Hong, Y.; Meng, T.; Liu, Y.; Dai, S.; Qiu, G.; Yuan, H.; Hu, F. Reversing activity of cancer associated fibroblast for staged glycolipid micelles against internal breast tumor cells. Theranostics 2019, 9, 6764–6779. [Google Scholar] [CrossRef] [PubMed]
  126. Cun, X.; Chen, J.; Li, M.; He, X.; Tang, X.; Guo, R.; Deng, M.; Li, M.; Zhang, Z.; He, Q. Tumor-Associated Fibroblast-Targeted Regulation and Deep Tumor Delivery of Chemotherapeutic Drugs with a Multifunctional Size-Switchable Nanoparticle. ACS Appl. Mater. Interfaces 2019, 11, 39545–39559. [Google Scholar] [CrossRef]
  127. Li, M.; Shi, K.; Tang, X.; Wei, J.; Cun, X.; Long, Y.; Zhang, Z.; He, Q. Synergistic tumor microenvironment targeting and blood-brain barrier penetration via a pH-responsive dual-ligand strategy for enhanced breast cancer and brain metastasis therapy. Nanomedicine 2018, 14, 1833–1843. [Google Scholar] [CrossRef]
  128. Qin, Y.; Liu, T.; Guo, M.; Liu, Y.; Liu, C.; Chen, Y.; Qu, D. Mild-heat-inducible sequentially released liposomal complex remodels the tumor microenvironment and reinforces anti-breast-cancer therapy. Biomater. Sci. 2020, 8, 3916–3925. [Google Scholar] [CrossRef]
  129. Zhen, Z.; Tang, W.; Wang, M.; Zhou, S.; Wang, H.; Wu, Z.; Hao, Z.; Li, Z.; Liu, L.; Xie, J. Protein Nanocage Mediated Fibroblast-Activation Protein Targeted Photoimmunotherapy To Enhance Cytotoxic T Cell Infiltration and Tumor Control. Nano Lett. 2017, 17, 862–869. [Google Scholar] [CrossRef]
  130. Cheng, Y.; Zou, J.; He, M.; Hou, X.; Wang, H.; Xu, J.; Yuan, Z.; Lan, M.; Yang, Y.; Chen, X.; et al. Spatiotemporally controlled Pseudomonas exotoxin transgene system combined with multifunctional nanoparticles for breast cancer antimetastatic therapy. J. Control. Release 2024, 367, 167–183. [Google Scholar] [CrossRef]
  131. Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef] [PubMed]
  132. Gungabeesoon, J.; Gort-Freitas, N.A.; Kiss, M.; Bolli, E.; Messemaker, M.; Siwicki, M.; Hicham, M.; Bill, R.; Koch, P.; Cianciaruso, C.; et al. A neutrophil response linked to tumor control in immunotherapy. Cell 2023, 186, 1448–1464. [Google Scholar] [CrossRef] [PubMed]
  133. Li, J.; Ge, Z.; Toh, K.; Liu, X.; Dirisala, A.; Ke, W.; Wen, P.; Zhou, H.; Wang, Z.; Xiao, S.; et al. Enzymatically Transformable Polymersome-Based Nanotherapeutics to Eliminate Minimal Relapsable Cancer. Adv. Mater. 2021, 33, e2105254. [Google Scholar] [CrossRef] [PubMed]
  134. de Bono, J.S.; Rowinsky, E.K. The ErbB receptor family: A therapeutic target for cancer. Trends Mol. Med. 2002, 8, S19–S26. [Google Scholar] [CrossRef] [PubMed]
  135. Gandullo-Sánchez, L.; Ocaña, A.; Pandiella, A. HER3 in cancer: From the bench to the bedside. J. Exp. Clin. Cancer Res. 2022, 41, 310. [Google Scholar] [CrossRef] [PubMed]
  136. Tao, J.J.; Castel, P.; Radosevic-Robin, N.; Elkabets, M.; Auricchio, N.; Aceto, N.; Weitsman, G.; Barber, P.; Vojnovic, B.; Ellis, H.; et al. Antagonism of EGFR and HER3 enhances the response to inhibitors of the PI3K-Akt pathway in triple-negative breast cancer. Sci. Signal. 2014, 7, ra29. [Google Scholar] [CrossRef]
  137. Miricescu, D.; Totan, A.; Stanescu-Spinu, I.I.; Badoiu, S.C.; Stefani, C.; Greabu, M. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects. Int. J. Mol. Sci. 2020, 22, 173. [Google Scholar] [CrossRef] [PubMed]
  138. Reeves, A.; Vinogradov, S.V.; Morrissey, P.; Chernin, M.; Ahmed, M.M. Curcumin-encapsulating Nanogels as an Effective Anti-cancer Formulation for Intracellular Uptake. Mol. Cell Pharmacol. 2015, 7, 25–40. [Google Scholar] [PubMed]
  139. Luo, L.; Xu, F.; Peng, H.; Luo, Y.; Tian, X.; Battaglia, G.; Zhang, H.; Gong, Q.; Gu, Z.; Luo, K. Stimuli-responsive polymeric pro-drug-based nanomedicine delivering nifuroxazide and doxorubicin against primary breast cancer and pulmonary metastasis. J. Control. Release 2020, 318, 124–135. [Google Scholar] [CrossRef]
  140. Dias, A.M.M.; Courteau, A.; Bellaye, P.S.; Kohli, E.; Oudot, A.; Doulain, P.E.; Petitot, C.; Walker, P.M.; Decréau, R.; Collin, B. Superparamagnetic Iron Oxide Nanoparticles for Immunotherapy of Cancers through Macrophages and Magnetic Hyperthermia. Pharmaceutics 2022, 14, 2388. [Google Scholar] [CrossRef]
  141. Farcas, C.G.; Dehelean, C.; Pinzaru, I.A.; Mioc, M.; Socoliuc, V.; Moaca, E.A.; Avram, S.; Ghiulai, R.; Coricovac, D.; Pavel, I.; et al. Thermosensitive Betulinic Acid-Loaded Magnetoliposomes: A Promising Antitumor Potential for Highly Aggressive Human Breast Adenocarcinoma Cells Under Hyperthermic Conditions. Int. J. Nanomed. 2020, 15, 8175–8200. [Google Scholar] [CrossRef] [PubMed]
  142. Yhee, J.Y.; Jeon, S.; Yoon, H.Y.; Shim, M.K.; Ko, H.; Min, J.; Na, J.H.; Chang, H.; Han, H.; Kim, J.H.; et al. Effects of tumor microenvironments on targeted delivery of glycol chitosan nanoparticles. J. Control. Release 2017, 267, 223–231. [Google Scholar] [CrossRef] [PubMed]
  143. Li, J.; Burgess, D.J. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm. Sin. B 2020, 10, 2110–2124. [Google Scholar] [CrossRef] [PubMed]
  144. Dreaden, E.C.; Austin, L.A.; Mackey, M.A.; El-Sayed, M.A. Size matters: Gold nanoparticles in targeted cancer drug delivery. Ther. Deliv. 2012, 3, 457–478. [Google Scholar] [CrossRef] [PubMed]
  145. Khlebtsov, N.; Dykman, L. Biodistribution and toxicity of engineered gold nanoparticles: A review of in vitro and in vivo studies. Chem. Soc. Rev. 2011, 40, 1647–1671. [Google Scholar] [CrossRef]
  146. Wen, P.; Ke, W.; Dirisala, A.; Toh, K.; Tanaka, M.; Li, J. Stealth and pseudo-stealth nanocarriers. Adv. Drug Deliv. Rev. 2023, 198, 114895. [Google Scholar] [CrossRef] [PubMed]
  147. Thakor, P.; Bhavana, V.; Sharma, R.; Srivastava, S.; Singh, S.B.; Mehra, N.K. Polymer-drug conjugates: Recent advances and future perspectives. Drug Discov. 2020, 25, 1718–1726. [Google Scholar] [CrossRef] [PubMed]
  148. Swetha, K.L.; Maravajjala, K.S.; Li, S.D.; Singh, M.S.; Roy, A. Breaking the niche: Multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv. Transl. Res. 2023, 13, 105–134. [Google Scholar] [CrossRef] [PubMed]
  149. Li, D.; Zhang, R.; Liu, G.; Kang, Y.; Wu, J. Redox-Responsive Self-Assembled Nanoparticles for Cancer Therapy. Adv. Healthc. Mater. 2020, 9, e2000605. [Google Scholar] [CrossRef]
  150. Rahimkhoei, V.; Alzaidy, A.H.; Abed, M.J.; Rashki, S.; Salavati-Niasari, M. Advances in inorganic nanoparticles-based drug delivery in targeted breast cancer theranostics. Adv. Colloid Interface Sci. 2024, 329, 103204. [Google Scholar] [CrossRef]
  151. Pulaski, B.A.; Ostrand-Rosenberg, S. Mouse 4T1 breast tumor model. Curr. Protoc. Immunol. 2001. Chapter 20: Unit 20.2. [Google Scholar] [CrossRef]
  152. Yang, L.; Yong, L.; Zhu, X.; Feng, Y.; Fu, Y.; Kong, D.; Lu, W.; Zhou, T.Y. Disease progression model of 4T1 metastatic breast cancer. J. Pharmacokinet. Pharmacodyn. 2020, 47, 105–116. [Google Scholar] [CrossRef] [PubMed]
  153. Vakhshiteh, F.; Bagheri, Z.; Soleimani, M.; Ahvaraki, A.; Pournemat, P.; Alavi, S.E.; Madjd, Z. Heterotypic tumor spheroids: A platform for nanomedicine evaluation. J. Nanobiotechnol. 2023, 21, 249. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Transforming cold tumors into hot tumors by modulating the cancer–immunity cycle. The cancer–immunity cycle starts with the release of cancer cell antigens (I), followed by their presentation to T cells by dendritic cells (II). This leads to T cells activation and their migration to the tumor site, where they infiltrate, recognize and kill the cancer cells (III–VII). The destruction of cancer cells releases new antigens, perpetuating the cycle and sustaining the immune response against the tumor (VII–I). Each step in this cycle offers potential targets for enhancing immunotherapy treatments.
Figure 1. Transforming cold tumors into hot tumors by modulating the cancer–immunity cycle. The cancer–immunity cycle starts with the release of cancer cell antigens (I), followed by their presentation to T cells by dendritic cells (II). This leads to T cells activation and their migration to the tumor site, where they infiltrate, recognize and kill the cancer cells (III–VII). The destruction of cancer cells releases new antigens, perpetuating the cycle and sustaining the immune response against the tumor (VII–I). Each step in this cycle offers potential targets for enhancing immunotherapy treatments.
Ijms 25 07208 g001
Figure 2. The biological functions and secreted cytokines of tumor-associated macrophages (TAMs). (1) Angiogenesis: TAMs promote the formation of new blood vessels, which supply tumors with oxygen and nutrients. (2) Immunosuppression: TAMs suppress immune responses against tumors by producing immunosuppressive cytokines. (3) Cell Survival and Proliferation: TAMs release growth factors that enhance tumor cell survival and proliferation. (4) Tumor Metabolism: TAMs adapt the tumor metabolism for rapid growth by enhancing the nutrient availability and adapting to low oxygen. (5) Treatment Resistance: TAMs promote resistance by altering the tumor environment and reducing the effectiveness of therapies. (6) Metastasis: TAMs assist in cancer metastasis by degrading extracellular matrices, facilitating the tumor cell invasion of other tissues.
Figure 2. The biological functions and secreted cytokines of tumor-associated macrophages (TAMs). (1) Angiogenesis: TAMs promote the formation of new blood vessels, which supply tumors with oxygen and nutrients. (2) Immunosuppression: TAMs suppress immune responses against tumors by producing immunosuppressive cytokines. (3) Cell Survival and Proliferation: TAMs release growth factors that enhance tumor cell survival and proliferation. (4) Tumor Metabolism: TAMs adapt the tumor metabolism for rapid growth by enhancing the nutrient availability and adapting to low oxygen. (5) Treatment Resistance: TAMs promote resistance by altering the tumor environment and reducing the effectiveness of therapies. (6) Metastasis: TAMs assist in cancer metastasis by degrading extracellular matrices, facilitating the tumor cell invasion of other tissues.
Ijms 25 07208 g002
Figure 3. The immune regulation of DC in tumor microenvironment. cDCs migrate to lymph nodes to prime T cells: cDC1s activate both CD4+ and CD8+ T cells, while cDC2s primarily activate CD4+ T cells. cDC1s produce CXCL9 and CXCL10, attracting activated T cells via CXCR3. DCs also regulate T cells activation through various co-stimulatory molecules. Interaction with NK cells enhances this dynamic, with NK cells promoting cDC1 recruitment and activation via chemokines like CCL5 and XCL1 and cDC1s, in turn, boosting NK cell function with IL-12. After antigen uptake, cDCs can become regulatory DCs expressing PD-L1, modulating immune responses towards either suppression or tolerance.
Figure 3. The immune regulation of DC in tumor microenvironment. cDCs migrate to lymph nodes to prime T cells: cDC1s activate both CD4+ and CD8+ T cells, while cDC2s primarily activate CD4+ T cells. cDC1s produce CXCL9 and CXCL10, attracting activated T cells via CXCR3. DCs also regulate T cells activation through various co-stimulatory molecules. Interaction with NK cells enhances this dynamic, with NK cells promoting cDC1 recruitment and activation via chemokines like CCL5 and XCL1 and cDC1s, in turn, boosting NK cell function with IL-12. After antigen uptake, cDCs can become regulatory DCs expressing PD-L1, modulating immune responses towards either suppression or tolerance.
Ijms 25 07208 g003
Figure 4. Diagrammatic illustration of selected pro-tumorigenic roles of cancer-associated fibroblasts (CAFs).
Figure 4. Diagrammatic illustration of selected pro-tumorigenic roles of cancer-associated fibroblasts (CAFs).
Ijms 25 07208 g004
Table 1. Cancer antigens and therapeutic bispecific antibodies of the treatment of breast cancer.
Table 1. Cancer antigens and therapeutic bispecific antibodies of the treatment of breast cancer.
Cancer AntigenAntibody TargetsReferences
HER2CD3 × HER2
mPEG × HER2
CD47 × HER2
[51,52,53]
HER3CD3 × HER3
HER2 × HER3
EFGR × HER3
[51,54,55]
p95HER2CD3 × p95HER2[56]
Trop-2CD3 × Trop-2[57]
CEACAM-5CD3 × CEACAM-5[57]
EphA10CD3 × EphA10[58]
EpCAMCD3 × EpCAM[59,60]
p-CadherinCD3 × p-Cadherin[61]
EGFRCD3 × EGFR
mPEG × EGFR
[62,63]
NotchEFGR × notch[64]
MesothelinCD16 × mesothelin[65]
Muc1CD16 × Muc1[66]
CTLA-4PD-1 × CTLA-4[67]
LAG-3PD-1 × LAG-3[68]
TGFβPD-L1 × TGFβ[69,70]
Table 2. TAMs-mediated nanoparticles against breast cancer.
Table 2. TAMs-mediated nanoparticles against breast cancer.
Payloads or Associated AgentsNanocarrierFunctionReferences
Doxorubicincleavable PEG chains covering the folate–modified liposometarget tumor cells and M2-TAMs; induce ICD at tumor sites; activate effector T cells (combined with CpG immune adjuvant); reduce M2-TAMs; promote maturation of DCs;[91]
Doxorubicinpoly(lactic-co-glycolic) acid NPs functionalized by acid-sensitive sheddable PEGylation and mannose modificationreduce TAM population and density in tumor tissues[92]
Doxorubicin, mitomycin CiRGD peptide (internalizing Arg-Gly-Asp peptide mimetic) functionalized terpolymer and poly(methacrylic acid)- polysorbate 80-grafted starch-lipid NPcross intact blood–brain barrier;
enhance cellular uptake, cytotoxicity and drug delivery; selective targetability to human TNBC cells and murine macrophages
[93]
Doxorubicin and zymosanNP complex composed of pegylated polyethylenimine and zymosan and Doxenhance cellular uptake; induce apoptosis; induce secretion of proinflammatory cytokine; modify the biodistribution of DOX; reversed TAMs polarization from M2 to M1 phenotype; anti- angiogenic effect[94]
Macrophage migration inhibitory factor-siRNAglucan -based NPsreduce tumor cell proliferation and enhance apoptosis; reduce the number of MIF at tumor; antitumor and anti-metastasis; increase CD4+ T cells infiltration[95]
siCCR2siCCR2-encapsulated cationic polymeric NPblock monocytes recruitment, reduce TAMs abundance in tumor tissues; reverse tumor immune suppression; enhance the antitumor effect of chemotherapy[96]
VEGF siRNA (siVEGF) and PIGF siRNA (siPIGF)PEG = MT/PC/siVEGF/siPIGF NPs a novel dual-stage pH-sensitive carrier composed of cationic polyethylene glycol (PEG) and mannose modified trimethyl chitosan conjugate (PEG = MT), and an anionic poly- (allylamine hydrochloride)-citraconic anhydride (PAH-Cit, PC)inhibit the proliferation of BCs; reverse TAMs polarization from M2 to M1 phenotype; inhibit BC lung metastasis; anti-angiogenic effect[97]
HydrazinocurcuminRR-11a-coupled liposomal NPssuppress STAT3 activity; reverse TAMs polarization from M2 to M1 phenotype[98]
MetforminHollow mesoporous manganese dioxide NPs coated with macrophage membranesreverse TAMs polarization from M2 to M1 phenotype; target TAMs; suppress tumor growth[99]
Zoledronic acid (ZOL)Asn-Gly-Arg and PEG2000 modified liposomesinhibit tumor growth; reduce TAMs; inhibit tumor angiogenesis[100]
Table 3. TAM-mediated nanoparticles against breast cancer.
Table 3. TAM-mediated nanoparticles against breast cancer.
Payloads or Associated AgentsNanocarrierFunctionReferences
Cytosine-phosphate-guanine (CpG)3-aminopropyltriethoxysilane-modified Fe3O4 NPssuppress the metastasis of BC to the lungs; increases infiltrating lymphocytes in tumors; stimulate humoral immune response[106]
Macrophage Inflammatory Protein 3 Beta (MIP-3β)NP complex composed of 1,2-Dioleoyl-3-trimethylammonium-propane, folic acid modified poly (ethylene glycol)-b-poly(ε-caprolactone) and methoxy poly (ethylene glycol)-poly(lactide)activate CD8+ T-lymphocytes; induce DCs maturation; inhibit M2 polarization; suppress angiogenesis; suppress tumor growth and metastasis[107]
Ganoderma lucidumpolysaccharideGanoderma lucidumpolysaccharide contained gold nanocompositesinduce DCs maturation; reverse the decline of CD4+ T cells and CD8+ T cells population; stimulate T cells proliferation; suppress tumor growth and metastasis [108]
CD73 specific siRNACD73-specific siRNA-loaded chitosanlactate NPsinhibit the expression of CD 73; reduce tumor growth rate; decrease Treg, MDSCs and TAMs, enhance CTL function; enhance the secretion of Th1 frequency and inflammatory cytokine network; antitumor and anti-metastasis;[109]
Resiquimod CR848 and doxorubicin-hyaluronic acid conjugate (HA-DOX)HA-DOX coated PHIS/R848 NPspromote DCs maturation and activation; selective effects on breast cancer cells; regulate antitumor immune response by promoting infiltration of T cells and CTLs;[110]
DOX,IL-2,IFN-γDC cell-derived nanovesiclessuppress tumor growth and metastasis; adsorb IFN-γ;
enhance DCs mature; increase the infiltration of CTLs and activation of NK cells; increase the recruitment of CD45+immune cells and Ly6G+neutrophils
[111]
Doxorubicinhighly integrated mesoporous silica NPsinduce DCs maturation; improve drug accumulate in the tumor; induce anticancer immune response[112]
DoxorubicinLow-dose doxorubicin hydrochloride cancer cell membrane coated calcium carbonate NPsinduce ICD; CRT exposure; promote DCs maturation[113]
Table 4. CAF-mediated nanoparticles against breast cancer.
Table 4. CAF-mediated nanoparticles against breast cancer.
Payloads or Associated AgentsNanocarrierFunctionReferences
DoxorubicinCleavable amphiphilic peptide (containing a TGPA peptide sequence)-NPsincrease the effective drug concentration at the FAP-α-rich tumor sites; facilitate drug penetration through the stromal barrier; possess tumor targeting specificity[124]
Telmisartan, DoxorubicinGlycolipid polymer-based micelles composed of chitosan and stearic acidDecrease activity of CAFs; inhibit CAFs secreted cytokines;[125]
Gemcitabine,18β-glycyrrhetinic acidNPs composed of dendrigraft poly-l-lysine, PEG-PCL, substrate peptide of MMP-2regulate the chemoexposed TAFs; deliver drug to the deep region of tumor tissue[126]
PaclitaxelLiposome co-modified with acid-cleavable folic acid and dNP2 peptideenhance the uptake and deep penetration in FR-positive tumor cells and FR-negative CAFs; exhibit synergistic TME targeting and blood brain barrier transmigration; accumulate in brain metastatic sites;[127]
Sodium tanshinone IIA sulfonate and celastrolA gold nanorod-anchored thermo- liposomal complexnormalize the tumor blood vessel; decrease the density of TAFs and collagen; regulate the secretion of cytokines[128]
ZnF16PcZnF16Pc-nanoparticle protein cage conjugated with a FAP- targeted single chain variable fragmentselectively eliminate CAFs; ECM destruction; suppress CXCL12 secretion; enhance CD8+ T cells infiltration[129]
Pseudomonas exotoxin A/anisamidePoly L lysine-based cationic carrier was coupled with the glutathione-sensitive disulfide bound vitamin E succinate and was covered with a sigma1 receptor and integrin αvβ3 receptor multitargetingsuppress angiogenesis; suppress tumor growth and metastasis; target elimination of CAFs;[130]
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

Yang, L.; Hu, Q.; Huang, T. Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert “Cold” Tumors to “Hot” Tumors. Int. J. Mol. Sci. 2024, 25, 7208. https://doi.org/10.3390/ijms25137208

AMA Style

Yang L, Hu Q, Huang T. Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert “Cold” Tumors to “Hot” Tumors. International Journal of Molecular Sciences. 2024; 25(13):7208. https://doi.org/10.3390/ijms25137208

Chicago/Turabian Style

Yang, Liucui, Qingyi Hu, and Tao Huang. 2024. "Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert “Cold” Tumors to “Hot” Tumors" International Journal of Molecular Sciences 25, no. 13: 7208. https://doi.org/10.3390/ijms25137208

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