Next Article in Journal
The Potential and Challenges of Proton FLASH in Head and Neck Cancer Reirradiation
Previous Article in Journal
Enhancing the Efficacy of Breast Cancer Immunotherapy Using a Smac-Armed Oncolytic Virus
Previous Article in Special Issue
Pre-Existing Immunity Predicts Response to First-Line Immunotherapy in Non-Small Cell Lung Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Present and Future of Immunotherapy for Triple-Negative Breast Cancer

1
Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
2
Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
3
Department of Radiation Oncology, UH Seidman Cancer Center, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
4
Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, USA
5
Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(19), 3250; https://doi.org/10.3390/cancers16193250
Submission received: 12 August 2024 / Revised: 18 September 2024 / Accepted: 23 September 2024 / Published: 24 September 2024
(This article belongs to the Special Issue Immunotherapy for Cancers)

Abstract

:

Simple Summary

Immunotherapy has changed the way we treat triple-negative breast cancer (TNBC), a type of breast cancer that does not have common markers like estrogen, progesterone, or HER2. TNBC has the worst outlook among breast cancers, but it may respond better to immunotherapy because it often shows higher levels of PD-L1 and has more immune cells attacking the tumor. Recently, the FDA approved pembrolizumab (Keytruda) combined with chemotherapy for advanced TNBC, offering new hope for patients. However, not all patients respond equally well, mainly because not everyone has high PD-L1 levels. To improve treatment success, combining immunotherapy with other treatments like chemotherapy, targeted therapies, or radiation seems promising. This review explains TNBC and immunotherapy, discusses current and future combination treatment strategies, and explores the challenges and potential new approaches that might soon be available for treating TNBC.

Abstract

Triple-negative breast cancer (TNBC) lacks the expression of estrogen receptors (ERs), human epidermal growth factor receptor 2 (HER2), and progesterone receptors (PRs). TNBC has the poorest prognosis among breast cancer subtypes and is more likely to respond to immunotherapy due to its higher expression of PD-L1 and a greater percentage of tumor-infiltrating lymphocytes. Immunotherapy has revolutionized TNBC treatment, especially with the FDA’s approval of pembrolizumab (Keytruda) combined with chemotherapy for advanced cases, opening new avenues for treating this deadly disease. Although immunotherapy can significantly improve patient outcomes in a subset of patients, achieving the desired response rate for all remains an unmet clinical goal. Strategies that enhance responses to immune checkpoint blockade, including combining immunotherapy with chemotherapy, molecularly targeted therapy, or radiotherapy, may improve response rates and clinical outcomes. In this review, we provide a short background on TNBC and immunotherapy and explore the different types of immunotherapy strategies that are currently being evaluated in TNBC. Additionally, we review why combination strategies may be beneficial, provide an overview of the combination strategies, and discuss the novel immunotherapeutic opportunities that may be approved in the near future for TNBC.

1. Introduction

Triple-negative breast cancer (TNBC) represents 15–20% of breast cancers (BCs) that are newly diagnosed and is a subtype with the fewest approved targeted therapies [1]. TNBC does not overexpress human epidermal growth factor receptor 2 (HER2) and lacks expression of either estrogen receptor (ER) or progesterone receptor (PR) [2]. Clinically, TNBC tumors are typically larger upon diagnosis and tend to develop nodal metastasis to the draining lymphatics [3]. This subtype is known for developing more lethal metastases which are more likely to originate in viscera, especially the brain and lungs, and less likely to spread to the bone [4]. TNBC patients are at a greater risk of early relapse compared to those with other BC subtypes, and only a subset of TNBC patients show a more favorable response to chemotherapy [4,5]. TNBC recurrence often peaks between the first- and third-year following diagnosis, then sharply declines in the years that follow. Relapses after eight or ten years are extremely rare [5]. Developments in treatment strategies remained limited for years, and cytotoxic chemotherapy continues to be the primary systemic treatment for TNBC and is often used in the neoadjuvant setting [1]. This enables a reduction in tumor burden and facilitates an in vivo evaluation of treatment response, making pathological complete response (pCR) a valuable prognostic marker for survival [6]. While there are several drugs targeting ER and HER2 in clinical subgroups, the paucity of progress in developing targeted therapies for TNBC is apparent [1]. The understanding of the intricate molecular and genetic basis for TNBC has steadily improved over the last decade or so, with several classification schemas for TNBC proposed [3,7,8,9]. Using emerging technologies, such as next-generation sequencing (NGS), Lehmann et al. validated both intratumoral and intertumoral heterogeneity and simplified the molecular classification of TNBC into six different subtypes: basal-like 1 and 2 (BL1 and BL2), immunomodulatory (IM), luminal androgen receptor (LAR), mesenchymal (M), and mesenchymal stem-like (MSL) [10]. In 2016, Lehmann revised the classification into four distinct subtypes: BL1, BL2, M, and LAR. IM and MSL were left out due to their low cellularity and dependability on tumor-infiltrating lymphocytes (TILs) and tumor-associated stromal cells [11].
Despite the widely held notion that BC is not immunogenic, numerous studies demonstrated that TNBC can activate the immune system. A subset of TNBC has BRCA 1/2 mutations [12], which are associated with higher tumor immunogenicity [13]. However, the immunogenicity of TNBC goes beyond BRCA1/2 mutations since the TNBC subtype has the greatest incidence in patients with a robust tumor immune infiltrate. Moreover, TNBC prognostic signatures include B cells which can secrete antibodies that bind to tumor antigens and amplify the adaptive immune responses [14]. Compared to other BC subtypes, TNBC displays lower clonal heterogeneity and higher immune gene expression [15]. The immune microenvironment of TNBC is a dynamic, intricate network of different immune cell populations, cytokines, and signaling pathways [16]. The immunological components of the TNBC tumor microenvironment (TME) are complex, as described in Figure 1.
Significant recent advancements have led to FDA approval of several drugs for TNBC (Table 1). Novel strategies such as immunotherapy, ionizing radiation therapy [17], platinum agents [18], and PARP inhibitors [19] have been explored to increase the pCR rates. TNBC is most likely to benefit from immunotherapy because several studies reported higher tumor mutational burden, increased expression of programmed cell death-ligand1 (PD-L1), and higher TILs in the TME compared to the other BC subtypes [20,21]. Based on several clinical trials, the immunotherapy and chemotherapy combination was approved in both early and advanced TNBC settings [22]. In this review, we discuss the different types of immunotherapy strategies that are currently employed in TNBC, articulate why combination strategies may be beneficial, provide an overview of the combination strategies, review the current clinical challenges encountered, and provide insight into potential future clinical developments.

2. Current Immunotherapy Approaches: Approved Treatments and Agents under Investigation for TNBC

The treatment landscape for TNBC has evolved significantly in recent years, with immunotherapy emerging as a promising avenue. This section provides an overview of the current clinical immunotherapy approaches being investigated for TNBC. These include cytokine therapies, monoclonal antibodies (mAbs), antibody–drug conjugates (ADCs), immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell therapies (ACTs), and oncolytic virus therapy (OV). Each of these strategies offers a unique mechanism to boost antitumor immunity, with several approaches showing potential in clinical trials. The following illustration (Figure 2) summarizes these diverse therapeutic strategies.

2.1. Cytokines

Cytokines are small proteins that are essential for immune response and cell signaling [23]. While cytokine-based therapies have been explored in several cancers including BC, their role in TNBC treatment is still actively explored. Cytokines regulate the host immune response to cancer cells and aid in prompting cancer cell death, making cytokine-based immunotherapy an intriguing possibility in cancer treatment [24]. The two cytokines approved by the FDA for the treatment of cancer, though not breast cancer, are IL-2 and IFN-α; nevertheless, their high toxicity profile has limited their use [23]. Several other cytokines including GM-CSF [25], IL-12, IL-15, and TNF are being tested in numerous clinical trials for their safety and effectiveness as cancer treatments [26]. Moreover, as single-agent immunotherapies, over 40 known cytokines have been approved for a restricted range of indications, including the treatment of cancer [27]. A recombinant adenovirus expressing IL-12 (AdIL-12) administered intratumorally has been demonstrated to cause substantial tumor regression in animal models of BC [27]. Patients with metastatic TNBC (mTNBC) demonstrated improved antigen presentation and a treatment-related spike in CD8+ TIL density following intratumoral administration of IL-12 (phase-1 pilot study) [28]. An engineered cytokine called empegaldesleukin or NKTR-214 preferentially activates the IL-2 receptor with a focus on metastatic solid tumors, including TNBC (phase-1 trial; NCT02869295) [29]. Cytokine activity facilitates both tumor-promoting and tumor-suppressive effects. Proinflammatory cytokines such as TNF-α and IL-6, for instance, regulate immunological interactions to promote anticancer effects. Cytokines present in the TME promote angiogenesis, epithelial-to-mesenchymal transition, invasion, and tumor growth—all processes linked to cancer development [30]. Several factors contribute to the poor efficacy of cytokine immune therapy including short half-life, increased toxicity, and low efficacy. High intratumoral cytokine dosages might cause systemic adverse effects including renal insufficiency, hypotension, neuropsychiatric symptoms, and respiratory failure [31]. Similarly, patients also have difficulty tolerating systemic treatment with recombinant IFN-α [32]. IRX-2, a novel therapy comprising numerous cytokines, demonstrated activation of the TME by elevating TIL numbers, PD-L1 expression, and lymphocyte activation in early TNBC patients in a phase-I study. A phase-II follow-up trial is underway (NCT04373031) [33]. Even though cytokines were thought to have several benefits when used as a monotherapy, most clinical trials that employed systemic cytokine monotherapy failed. This could be due to inadequate cytokine concentrations in the tumor upon parenteral administration, as well as major toxicities related to the activation of humoral or cellular checkpoints [34].

2.2. Monoclonal Antibodies

The goal of using humanized mAbs is to decrease immunotolerance and boost the immune response against tumors by blocking immunosuppressive checkpoints that the tumor uses to evade immune system control [35]. Trastuzumab was the first FDA-approved monoclonal antibody for the treatment of HER2-positive breast cancer [36]. TNBC tumors do not express HER2; thus, these patients cannot be treated with trastuzumab or other HER2-specific agents [37]. In 2009, the FDA authorized bevacizumab (Avastin), a humanized mAb that specifically targets VEGF-A. VEGF regulates tumor-induced immunosuppression in addition to blood vessel development [38]. Therefore, bevacizumab’s immunomodulatory characteristics open possibilities for novel combination treatment approaches [38]. In 2010, the approval of bevacizumab for breast cancer was recommended for withdrawal by the Office of New Drugs (OND). Significant benefits were not confirmed by the required follow-up trials, and increased serious adverse events with a lack of survival benefit were revealed, leading to the conclusion that the risks outweigh the benefits of this indication [39]. Aspartic protease Cath-D is an extracellular target unique to TNBC. As a promising immunotherapy, an immunomodulatory antibody-based approach against Cath-D is presently in its developmental phase to treat TNBC patients [40].

2.3. Antibody–Drug Conjugates

ADCs can identify antigens that are tumor-specific or overexpressed in tumors and thus can kill cancer cells via antigen-dependent cell-mediated cytotoxicity (ADCC) [41]. ADCs utilize the specificity of mAbs on cellular-antigen identification to administer potent cytotoxic drugs in a tailored approach [42]. TNF receptor superfamily members such as CD40 can help DCs to stimulate antitumor T cells and retrain macrophages to kill tumor stroma. The efficacy of anti-CD40 antibodies has been evaluated in several clinical trials [43].
ADC trastuzumab deruxtecan (DS-8201) comprises a cytotoxic topoisomerase I inhibitor and an anti-HER2 antibody connected by a cleavable tetrapeptide linker [44]. Its capacity to produce cytotoxic action against antigen-negative cells (bystander effect) may have led the FDA to approve it in BC patients who have been pretreated with trastuzumab emtansine (TDM1) [45]. The DESTINY-Breast04 and DESTINY-Breast06 trials have yielded critical data on the efficacy of trastuzumab deruxtecan in treating low-HER2 breast cancer. DESTINY-Breast04 demonstrated that patients with low-HER2 breast cancer (IHC 1+ or IHC 2+ without FISH amplification) had significantly superior progression-free survival (PFS) when treated with trastuzumab deruxtecan compared to those who received the physician’s choice of cytotoxic chemotherapy. This trial established trastuzumab deruxtecan as a viable treatment option for low-HER2 breast cancer, highlighting its potential to change clinical practice [46].
Similarly, the DESTINY-Breast06 trial expanded the understanding of trastuzumab deruxtecan’s efficacy by showing benefits for patients with “ultra-low”-IHC breast cancer (IHC 0 but with subtle HER2 expression). This finding is particularly notable as it extends the applicability of trastuzumab deruxtecan to a broader patient population, including those previously not considered for HER2-targeted therapies (NCT04494425). These results are especially significant for TNBC patients, who traditionally have limited treatment options. Moreover, it is important to underscore the CNS activity of trastuzumab deruxtecan, as its ability to penetrate the central nervous system could provide substantial benefits for patients with brain metastases, a common and challenging complication in breast cancer. The TUXEDO-1 trial (NCT04752059) demonstrated that trastuzumab deruxtecan achieved a high intracranial response rate in patients with active brain metastases from HER2-positive breast cancer, establishing it as a viable treatment option for this condition [47]. Another ADC, Disitamab vedotin, consists of a HER2 mAb conjugated to cytotoxic drug monomethyl auristatin E with a cleavable linker and is also being tested in clinical trials for several solid tumors including low-/HER2+ BCs [48].
Sacituzumab govitecan is the first ADC to receive FDA approval to treat TNBC which consists of a human trophoblast cell-surface antigen-2 (TROP2) antibody that is connected with a topoisomerase I inhibitor (SN-38) via a unique hydrolyzable linker [49]. Recently, a phase-III trial (ASCENT) demonstrated noticeably prolonged OS and PFS while using sacituzumab govitecan instead of single-agent chemotherapy [50]. Apart from HER2 and TROP2-based ADC, the activities of folate receptor alpha (FRα) and zinc transporter LIV-1-based ADC have been clinically evaluated for TNBC [40]. Ladiratuzumab vedotin is an ADC that targets Syndecan-1 on cancer cells. It binds to these cells, is internalized, and releases a cytotoxic agent that disrupts microtubules, leading to cell cycle arrest and apoptosis [51]. In early-phase clinical trials, ladiratuzumab vedotin is being explored as a monotherapy for patients with BC; some of these studies have already shown encouraging results [51]. In a phase-I, multi-part, dose-escalation SGNLVA-001 trial, mTNBC exhibited a superior overall response rate (ORR) and disease control rate (DCR) with Ladiratuzumab vedotin [52]. Recently, Tsai and colleagues reported an ORR of 28% with Ladiratuzumab vedotin at 1.25 mg/kg, indicating the favorable activity of the ADC [53].
Recently, a newly developed ADC (ESG401), which consists of a humanized anti-TROP2 IgG1 monoclonal antibody connected to the Topoisomerase I Inhibitor SN-38 through a stable cleavable linker, demonstrated promising effectiveness and tolerability in a phase-Ia trial [54]. Moreover, the effectiveness of ESG401 in treating brain metastases in first-line mTNBC patients corresponds with our prior findings in late-line mTNBC and HR+/HER2− BC patients (NCT04892342) [55]. Datopotamab deruxtecan (Dato-DXd) is another ADC where the antibody datopotamab, targeting TROP2 on breast cancer cells, is linked to the cytotoxic drug DXd. Once datopotamab binds to TROP2 and is internalized, the linker breaks down, releasing DXd to kill the cancer cell [56]. Dato-DXd’s ability to recruit immune cells to cancer sites suggests that combining it with durvalumab, which blocks PD-L1 and enhances immune cell activity, may enhance its effectiveness. In the phase-I study of Dato-DXd (NCT03401385), promising antitumor activity and a manageable safety profile were observed in patients with heavily pretreated advanced HR+/HER2− breast cancer and TNBC [57]. The ongoing TROPION-Breast03 trial (NCT05629585) will compare Dato-DXd alone or with durvalumab against standard care in patients with non-mTNBC with residual cancer cells post-surgery [56]. Thus, ADCs continue to play an ever evolving and significant role in the management of mTNBC.

2.4. Immune Checkpoint Inhibitors

ICIs kill tumor cells by disabling the immune system’s “braking” function on the immune cells that attack cancer. ICIs target the three best-characterized targets, PD-L1 (also called as B7 homolog 1) and PD-1 (also known as CD279), while blocking cytotoxic T-lymphocyte-associated protein 4 (CTLA-4/CD152) [58]. The mechanism of action of ICIs targeting PD-L1, PD-1, and CTLA-4 is shown in Figure 3. ICIs have the potential to be beneficial for both immunoinflammatory and immunological-suppressive types, potentially changing the TME from an “immune cold” to an “immune hot” phenotype [59]. The immunosuppressive PD-1 protein is mostly expressed on the cell surface (i.e., plasma membrane) of B, T, myeloid, and NK cells of the immune system [60]. The FDA-approved PD-1/PD-L1 inhibitors include atezolizumab, avelumab, cemiplimab, durvalumab, nivolumab, and pembrolizumab [61]. PD-L1 expression is directly correlated to histological grade and lymphocyte infiltration and is observed in 20–30% of TNBC cases [61].
Pembrolizumab was found to be safe with a good ORR in TNBC patients (KEYNOTE-012; [62]) and pembrolizumab monotherapy provided long-lasting antitumor efficacy in patients with both early and advanced PD-L1-positive TNBC with a combined positive score ≥ 1 [63,64]. PD-1 inhibitor JS001 demonstrated good safety and efficacy in mTNBC patients (NCT02838823) who failed prior multi-line treatments [65,66]. Pembrolizumab received approval based on the KEYNOTE-355 trial (NCT02819518), a multicenter, double-blind, randomized, placebo-controlled study involving patients with locally recurrent unresectable or metastatic TNBC who had not previously received chemotherapy for metastatic disease [66]. Similarly, atezolizumab monotherapy offered enduring clinical advantages for patients with mTNBC (NCT01375842; [67]), while avelumab provided an ORR of 44.4% (PD-L1 ≥ 10%) and 2.6% (PD-L1 < 10%) in TNBC patients (JAVELIN trial; [68]). The FDA initially approved atezolizumab in combination with nab-paclitaxel for first-line treatment of TNBC based on the IMpassion130 trial. However, this approval was later withdrawn following the negative results of the IMpassion131 trial [69].
T cells receive positive and negative feedback from the CD28 and CTLA-4 receptors, respectively [70]. CTLA-4 maintains T-cell homeostasis since it specifically regulates CD4+ T-cell responses [71]. Importantly, tissues from lymph node metastases show considerably higher CTLA-4 levels than tissue samples from the original breast tumor as seen in axial lymph node (ALN) metastasis [72]. There is presently no approved CTLA-4 inhibitor that can be used exclusively for TNBC, but ipilimumab is FDA-approved to treat several other cancers.

2.5. Vaccines

Known antigens associated with breast tumors are the main target of therapeutic vaccines, which work by actively immunizing against the tumor. A patient-specific tumor mutanome is used in cutting-edge settings to produce vaccines [73]. Vaccines can modify the TME through chemokines which can directly impact tumor growth as well as cytotoxic CD8+ T-cell (CTL) and NK responses. Several independent approaches have been used to develop therapeutic vaccines that use DC, DNA, RNA, peptides, carbohydrates, or all of the above [74]. CD4+ helper T lymphocytes can be stimulated by AE37, which is an Ii-Key hybrid of the MHC class II peptide. The randomized phase-II trial comparing GM-CSF alone with the AE37 + GM-CSF vaccine to prevent BC recurrence revealed no statistically significant differences in the five-year DFS between the treatment arms [75,76]. Similarly, a phase-1 trial with FRα peptide vaccine was well tolerated and produced responses that lasted over a year in more than 90% of patients with BC [77]. Poxvirus used in the PANVAC vaccine encodes mucin-1 (MUC-1) and carcinoembryonic antigen (CEA) along with T-cell-stimulating proteins LFA-350, ICAM-1, and B7.1 [78]. Favorable clinical responses to this vaccine have been observed in a limited number of patients [79]. Similarly, mixed subtypes of BC subjects treated with an autologous DC vaccine pulsed with various p53 peptides resulted in stable disease in ~30% of patients, and a small subgroup of these had increased CD8+ T-cell responses [80]. Autologous DC vaccine increased PFS to over 3 years in a subgroup of ER/PR double-negative [81] and PR-negative stage IV BC subjects [82]. Early clinical investigations of the DC vaccine including hTERT [83] peptides and FRα [77] have demonstrated T-cell activation, further supporting the role of the DC vaccine in BC management. These include a novel alpha-lactalbumin vaccine in patients with stage II-III TNBC (phase I) and in individuals at risk for TNBC who are planning to undergo preventative bilateral mastectomy (phase I) [84], STEMVAC, a DNA plasmid-based vaccine (phase II, NCT05455658) on patients with curatively treated stage I–III TNBC. STEMVAC targets proteins expressed on breast cancer stem cells, working to enhance the immune system’s ability to detect and eliminate the cancer cells responsible for the disease [85].

2.6. Adoptive Cell Therapy

T cells are crucial for cell-mediated immunity. Two forms of ACT that can alter natural T cells ex vivo and reintroduce them into the body to make them more potent tumor-destroying agents are chimeric antigen receptor (CAR) T-cell and T-cell receptor (TCR)-engineered T-cell therapies [86]. CAR T cells are designed to exclusively identify surface antigens by fusing antibody fragments on the T cell’s antigen-binding region. In contrast, MHC-expressed intracellular antigens are recognized by TCRs through the utilization of an alpha–beta chain heterodimer. Consequently, since TCRs may target a larger variety of antigens than CAR-T, they might be more advantageous in solid tumors [86]. Enhancing CAR-T cell infiltration in tumor tissues is a main obstacle for CAR-T therapy in BC. This obstacle may be addressed by combining the delivery of CAR-T cells with strong stimulation of antigen-presenting cells (APCs) to produce chemotactic cytokines [87]. Receptor tyrosine kinase c-Met is overexpressed in approximately 50% of BCs. The intratumoral injections of c-Met-CAR-T cells were well tolerated and induced an inflammatory response in metastatic BC patients with c-Met-expression in a phase-0 trial (NCT01837602) [88]. Mesothelin is overexpressed in TNBC, which is linked to a poorer prognosis [89]. This led to the development of mesothelin-specific CAR-T cells. Initial findings from a phase-I/II trial (NCT02414269) in patients with advanced solid tumors demonstrated antitumor activity of mesothelin-targeted CAR-T cells without any significant toxicities [90]. Similarly, a combination of mesothelin-targeted CAR-T cells with pembrolizumab was found to be safe and well tolerated in patients with malignant pleural disease (NCT02792114) and demonstrated antitumor efficacy [91]. Over 90% of TNBC cases express high MUC1 protein, which is linked to a poor prognosis [92]. An anti-MUC1 CAR-T cell-based phase-I clinical trial is currently underway (NCT04020575) for patients with advanced MUC1-positive BC [93]. Another well-known marker for BC’s adverse prognosis is CEA [94]. A phase-I trial that aimed to analyze the safety and tolerability of anti-CEA T-cell therapy (NCT00673829) in patients with metastatic BC has been suspended without any published results [95]. Luen et al. showcased how TILs could be a crucial factor in adapting clinical trial designs. Currently, TILs do not have clinical utility in any cancer type, and thus should not be utilized as a biomarker to tailor clinical therapies in daily practice. To fully investigate their clinical relevance, the next logical progression would be to consider using TILs as an adaptive factor in clinical trials [96]. Additional clinical testing of adoptive cell therapies in TNBC is warranted to improve the clinical outcome in subjects with TNBC, especially late-stage and mTNBC.

2.7. Oncolytic Virus Therapy

Natural or genetically engineered viruses that can proliferate selectively in cancer cells without harming healthy cells are known as OVs [97,98]. OVs may lyse tumor cells by infecting them directly, multiplying within them, or stimulating the immune system [99]. OVs are designed to target several stages in the cancer–immunity cycle. Oncolytic viruses cause immunogenic cell death, which triggers the innate and adaptive immune systems by releasing danger signals and neo-antigens [100]. At present, the only type of OV authorized for cancer therapy is talimogene laherparepvec (T-VEC), a herpes simplex virus-1 (HSV) modified to express GM-CSF [101]. An additional phase-II trial (NCT02658812) assessed the effectiveness of intratumoral T-VEC as monotherapy for locoregionally inoperable BC recurrence, irrespective of whether there was a distant recurrence. The results demonstrated that uncontrolled disease development made intratumoral T-VEC monotherapy less effective, and concurrent systemic treatment administration may be necessary [102]. The most researched OV for BC management is adenovirus. The ICOVIR-7 trial included patients with advanced and refractory solid cancer including BC. Although the OV was declared safe and a majority of subjects (16 out of 18) developed neutralizing antibodies, all BC subjects (n = 3) failed to reach efficacy endpoints [103]. Conversely, Ad5/3-D24-GM-CSF, an OV that codes for GM-CSF, effectively immunized patients with advanced BCs including TNBC [104]. Currently, an OV, MEM-288, that carries recombinant chimeric CD40 (MEM40) and human interferon beta (IFNβ) is being investigated (NCT05076760) in various solid cancers including TNBC [105]. In vivo and ex vivo testing of several different OVs, including Coxsackie, Maraba, measles, Newcastle disease, Polio, and Vaccinia, has cleared the path for human safety studies [106]. A phase-I clinical trial (NCT01846091) using a measles virus that encodes human thyroidal sodium iodide symporter (MV-NIS) is presently being evaluated in a range of cancers including mTNBC [107].

3. Rationale of Combining Immunotherapy with Other Therapies

TNBC is an aggressive disease and often develops resistance to standard-of-care (SOC) treatment. Thus, immunotherapy in combination with SOC is expected to improve the outcome for several reasons: (1) Different therapies target cancer cells through distinct mechanisms. Combining immunotherapy with chemotherapy, targeted therapy, or radiation therapy can attack cancer cells through multiple pathways simultaneously, resulting in a robust response [108]. (2) Some therapies can increase the immune system’s capacity to identify and target cancer cells. For example, chemotherapy can induce immunogenic cell death, releasing tumor antigens that activate the immune response, thereby synergizing with immunotherapy [109]. (3) TNBC often develops resistance to single-agent therapies [110]. Combination therapies can target several pathways implicated in tumorigenesis and immune evasion, reducing the likelihood of developing resistance [111]. (4) Not all patients respond to immunotherapy alone. Combining immunotherapy with other therapies may broaden the spectrum of patients who benefit from treatment, improving overall outcomes [112]. (5) Combining lower doses of different therapies may reduce individual treatment-related toxicities while maintaining efficacy, improving patients’ quality of life [111]. (6) Targeting TNBC with a combination of therapies can potentially decrease the risk of metastasis or recurrence by eradicating residual cancer cells that may metastasize to other tissues/organs [113]. Overall, combination strategies with immunotherapy represent a comprehensive approach to treating TNBC, addressing its heterogeneity and resistance mechanisms while maximizing therapeutic efficacy and minimizing toxicity. Several key TNBC clinical trials that include(d) immunotherapy in combination with other treatment modalities are listed in Table 2 (non-exhaustive list).

3.1. Combination of Immunotherapy with PARP Inhibitors

The combination of Poly ADP-ribose polymerase (PARP) inhibitors (PARPis) with immunotherapy is an active area of investigation for TNBC therapeutics. PARP plays an important role in the DNA repair process. Olaparib and talazoparib are key PARPis approved to treat BRCA1/2 mutant cancers, while niraparib is specifically approved for use in gynecologic cancers [114]. PARP inhibitors were approved by the FDA for patients with metastatic and early TNBC who have germline mutations in BRCA1/2 [115,116]. However, recent studies have shown that PARPis may also have immunomodulatory effects that could enhance the efficacy of immunotherapy in TNBC [117]. TNBCs with BRCA mutations or other DNA repair deficiencies are significantly more sensitive to PARPis [116]. Several clinical trials are currently investigating the combination of PARPis with immunotherapy in TNBC.
Patients with germline BRCA1/2 mutation-associated HER2-negative BC undergoing treatment in the first- through third-line of therapy were randomized to receive either olaparib or chemotherapy in the phase-3 OLYMPIAD trial [115]. Olaparib did not considerably increase OS in the study population as compared to chemotherapy, but the median PFS was 2.8 months longer. However, among patients receiving first-line treatment, olaparib increased OS by 7.9 months compared to chemotherapy alone [116]. This led to FDA approval for Olaparib therapy in women with TNBC with germline BRCA mutations. In the OlympiA trial (NCT02032823), adjuvant olaparib significantly enhanced invasive and distant disease-free survival in patients with high-risk, HER2-negative early breast cancer and germline BRCA1 or BRCA2 mutations, compared to a placebo. However, olaparib had a minimal effect on the overall quality of life reported by patients [118]. Likewise, patients receiving first- through fourth-line therapy for germline BRCA1/2 mutation-associated HER2-negative BC in the phase-3 EMBRACA trial were randomly assigned to receive talazoparib or chemotherapy [115]. In comparison to chemotherapy, talazoparib had a greater ORR and mPFS, but talazoparib did not considerably increase OS when compared to chemotherapy [115]. Based on these results, PARPi monotherapy was suggested as the SOC for metastatic HER2-negative BC patients with BRCA1/2 mutations [119]. Despite having strong response rates, PARPi-induced responses are generally less durable. Although checkpoint inhibitor monotherapy has a lower response rate than PARPis, it produces longer-lasting effects in mTNBC [120].
Comparing the results of the phase-II KEYNOTE-162 trial [121], which combined PD-1 checkpoint inhibitor pembrolizumab with PARPi niraparib, to trials with PARPi monotherapy, it became evident that patients with BRCA1 or BRCA2 pathogenic variants (PVs) displayed long-lasting responses with combination treatment [122]. In the phase-I/II MEDIOLA basket trial combining olaparib and durvalumab, the patients with metastatic HER-2-negative BC with germline BRCA1/2 PVs were explicitly included in one of the four cohorts [123]. Overall, the combined regimen was well tolerated but it is unclear whether the combined approach would be more effective in this group of patients with germline BRCA1/2 PVs than the PARP inhibitor alone, especially in terms of extending the duration of response [123]. Avelumab in combination with talazoparib has been assessed in two JAVELIN basket trials for patients with previously treated solid malignancies, including BC. The JAVELIN trials showed an acceptable level of safety [124]. While initial results from early-phase clinical trials have shown promising activity, larger randomized controlled trials are needed to establish the optimal combination regimen, patient selection criteria, and long-term outcomes. Combining PARP inhibitors with immunotherapy may also pose challenges, such as increased toxicity or the development of resistance. Therefore, careful monitoring and management of adverse events are essential moving forward.

3.2. Combination of Immunotherapy with Chemotherapy

Based on several preclinical and clinical studies, it is clear that several chemotherapies kill tumor cells by promoting the recruitment and maturation of APCs, enhancing the antigen presentation, and encouraging T-cell activation [125]. Additionally, by releasing MHC molecules and cell surface antigens, chemotherapy drugs improve the immunogenicity of tumors [126]. Chemotherapy-induced transient immunosuppression results in an enormous release of cyto- and chemokines which boosts immune cell infiltration and activation. Therefore, the combination of PD-(L)1 inhibitors and chemotherapy is a viable strategy to improve immunotherapy efficacy and promote synergistic antitumor activity [21]. Numerous clinical trials combining immunotherapy and chemotherapy are being carried out based on this concept with significant clinical advantage attained.
Most clinical trials currently use immunotherapy and chemotherapy together, in part because the response to ICIs is slower, whereas chemotherapeutic agents kill tumor cells and alter the TME during the therapy period [21]. Previously untreated advanced TNBC patients were treated with pembrolizumab plus chemotherapy or placebo plus chemotherapy as the first-line treatment in KEYNOTE-355. The chemotherapy regimens were chosen by the treating physicians and included gemcitabine/carboplatin, paclitaxel, and nab-paclitaxel [127]. Patients treated with the pembrolizumab and chemotherapy combination had a significant and clinically meaningful increase in PFS compared to the placebo–chemotherapy group with a combined positive score (CPS) ≥ 10. CPS is determined by the ratio of PD-L1-positive cells (tumor cells, lymphocytes, and macrophages) to the total number of tumor cells using the 22C3 assay [128]. The release of follow-up data showed that when pembrolizumab was added to chemotherapy, OS increased by about 7 months in the CPS ≥ 10 group and side effects were tolerable [128]. In the phase-IB/II KEYNOTE-150 trial, patients with mTNBC who had undergone at least two lines of previous therapy were treated with pembrolizumab plus eribulin mesylate. Of the 167 patients recruited, 40% were categorized as stratum 1 since they had not previously received systemic therapy. In this group of patients, the survival was greatest for PD-L1-positive individuals [129]. The IMpassion130 phase-III trial [66] further validated the efficacy of this immuno-chemotherapy combination for mTNBC patients who did not receive systemic therapy, following a phase-Ib trial (NCT01633970) that showed the safety of atezolizumab plus nab-paclitaxel in patients with locally recurrent or mTNBC [130]. Based on preliminary findings, adding atezolizumab was associated with a benefit for PFS in both PD-L1-positive and intention-to-treat (ITT) populations. Atezolizumab was found to significantly enhance OS in the PD-L1-positive group from 18.0 months to 25.0 months in the second set of intermediate findings; however, in the ITT population, there was no significant difference [131]. As a result, the FDA approved atezolizumab in March 2019 for use as a first-line treatment for patients with late-stage TNBC in combination with nab-paclitaxel [132]. The combination of AKT inhibitor ipatasertib with atezolizumab + paclitaxel/nab-paclitaxel demonstrated excellent efficacy in treating advanced and mTNBC [133].
Nevertheless, these different results were noted in the phase-III IMpassion131 trial which examined atezolizumab plus paclitaxel as first-line therapy for patients with advanced or mTNBC and compared the results to placebo plus paclitaxel. This study did not find any discernible differences in PFS between the two groups based on PD-L1 expression [134]. Based on recent single-cell sequencing (scSeq) data, paclitaxel may decrease critical antitumor immune cells in the TME while it may increase immunosuppressive macrophages, which could impact the efficacy of atezolizumab [135]. However, additional investigations are warranted to validate these findings. In addition to concurrent chemotherapy, another novel approach for the immuno-chemotherapy combination is to induce low-dose chemotherapy before immunotherapy. Patients with mTNBC received no induction or 2 weeks of low-dose cyclophosphamide, cisplatin, doxorubicin, and hypo-fractionated irradiation, followed by nivolumab, in the phase-II TONIC trial. The results suggested that doxorubicin or cisplatin induction, even for a short period, can shift the TIME toward an inflammatory state and enhance the response to nivolumab in TNBC [136]. Pembrolizumab and chemotherapy combination has consistently increased pCR in multiple clinical trials (I-SPY2 [137], KEYNOTE-173 [66], and KEYNOTE-522 [66]) in patients with early-stage TNBC. In July 2021, the FDA approved pembrolizumab as a neoadjuvant treatment for early-stage, high-risk TNBC alongside chemotherapy based on the improved pCR rates noted in these trials [138].
In the phase-II GeparNuevo trial, durvalumab or placebo was administered every 4 weeks in addition to chemotherapy and their efficacy was assessed. Addition of durvalumab significantly increased the pCR rates in the window cohort but not in the overall study population [139]. In the NeoTRIP trial, a separate trial of the PD-L1 inhibitor atezolizumab, the efficacy of eight cycles of carboplatin and nab-paclitaxel with or without atezolizumab was examined in high-risk TNBC. As an adjuvant treatment, four cycles of anthracycline regimen chemotherapy were given. The pCR rate in the ITT group was not significantly different with the addition of atezolizumab in a neoadjuvant setting [140]. Conversely, in the IMpassion031 study, atezolizumab given as a single drug with a standard chemotherapy regimen including doxorubicin and paclitaxel significantly increased pCR from 41% in the chemotherapy-alone arm to 58% in the ateza-plus-chemotherapy arm [141]. The IMpassion130 phase-3 trial, reported in 2018, showed that first-line treatment with atezolizumab plus nab-paclitaxel significantly improved PFS compared to placebo plus nab-paclitaxel in mTNBC. While the overall survival boundary was not crossed in this interim analysis and was not formally tested for statistical significance, numerical increases in median OS were observed in both the intention-to-treat and PD-L1-positive subgroups [17]. However, the FDA approval of atezolizumab was withdrawn due to the negative results from the IMpassion131 trial [134]. It should also be noted that the IMpassion130 and IMpassion131 trials used the SP142 assay for PD-L1 expression, which differs from other assays in detecting PD-L1 levels.

3.3. Combination of Immunotherapy with Radiotherapy

Radiotherapy (RT) is still a major treatment modality in TNBC even after recent advancements in endocrine therapy, chemotherapy, and targeted therapy for BC [112]. Numerous randomized trials have demonstrated that adjuvant radiation therapy decreases locoregional recurrence and improves survival in women with both early-stage and advanced-stage breast cancer. The impact of radiotherapy on immune signaling is still being elucidated [142,143]. Immune cells are attracted to the TME by radiation in several ways. It triggers the release of warning signals from dying tumor cells. DCs consume antigens from cancerous cells and deliver them to lymph nodes. The T cells are then exposed to them, which activates CD8+ and CD4+ T cells. Consequently, chemokines drive effector T-cell recruitment to tumors [144]. In BC patients, RT plus immunotherapy may produce systemic antitumor effects, especially when RT is administered at higher doses using more advanced techniques. Potential explanations for these systemic effects include the growth and dissemination of effector immune cells to distant sites because of the local immune priming by RT [145].
The adaptive phase-2 TONIC study, however, demonstrated a limited increase (~10%) in ORR with low-dose radiation combination as compared to nivolumab alone [136]. The best ORRs were observed with nivolumab and chemotherapy combinations (doxorubicin 35%, cisplatin ORR 23%) in this trial as mentioned above. A multicenter phase-2 trial assessed the safety and efficacy of pembrolizumab plus RT in patients with mTNBC (NCT02730130) [146]. This study discovered that the unselected PD-L1 population’s ORR in the ITT cohort was 17.6%, which was greater than the ORR of mTNBC patients who had previously received ICI monotherapy. Fifty patients who had received fewer than two lines of systemic therapy were included in a phase-II AZTEC trial to receive atezolizumab plus RT (NCT03464942) [147]. Patients were randomized to receive either 24 Gy stereotactic ablative radiotherapy (SABR) in three fractions or 20 Gy SABR in one fraction. Five days following the last RT segment, atezolizumab was initiated. There was no discernible variation in median progression-free survival (mPFS) between the two cohorts. TIL levels of 5% and the PD-L1 expression had little impact on the efficacy [147]. It would be interesting to see the results of numerous trials that are currently investigating the clinical benefit of RT in combination with ICI in women with TNBC.

3.4. Dual Antibody Combinations and Dual Immunotherapies

ADCs can interact with anti-PD-(L)1 agents to improve tumor control [148]. Sacituzumab govitecan targets TROP2 and delivers topoisomerase I inhibitor SN38 to the tumor. The clinical efficiency of this agent with pembrolizumab as a first-line treatment for mTNBC is currently being assessed [21]. Similarly, the safety and efficacy of ladiratuzumab vedotin an ADC that targets LIV-1 [149] is being evaluated in combination with pembrolizumab in a phase-Ib/II trial [150].
VEGF is a crucial factor in vascular endothelial cells that promotes angiogenesis, cell invasion, migration, proliferation, and survival, and enhances vascular permeability [151]. The combination of low-dose VEGFR inhibitor apatinib with anti-PD-1 agent camrelizumab and PARP1/2i fuzoloparib demonstrated a manageable safety profile and preliminary antitumor activity in patients with advanced TNBC [152]. The FUTURE-C-Plus trial demonstrated that CD8+ and/or PD-L1-positive patients benefit more from the combination of famitinib (VEGFRi), camrelizumab (ICI), and chemotherapy (nab-paclitaxel) [153]. The trial validated the safety, efficacy, and feasibility of triple therapy in TNBC and identified CD8+ positivity as a marker of favorable response with the triple therapy combination in the clinical setting [153].
Dual ICI therapies have been designed to overcome PD-(L)1 inhibitor resistance and reverse the tumor immunosuppressive microenvironment [154]. Durvalumab plus tremelimumab showed preliminary effectiveness and a manageable safety profile (NCT02536794) only in mTNBC patients (ORR 43%) while there was no response in ER+ BC [155]. Responders had higher expression of CD8, granzyme A, and perforin-1 post-therapy as compared to non-responders [155]. In the I/II SYNERGY (NCT03616886) trial, locally advanced or mTNBC patients were treated with a combination of oleclumab (anti-CD73 antibody), durvalumab, and chemotherapy (carboplatin and paclitaxel). The phase-II part of this trial was randomized 1:1 with/without oleclumab. However, this trial did not meet its primary endpoint (insignificant clinical benefit at 24 weeks) [156].
Similarly, in patients with mTNBC, a phase-I trial (NCT03256344) assessed the safety of intrahepatic injection of T-VEC in combination with intravenous atezolizumab [157]. The five TNBC patients in their DLT cohorts did not have any dose-limiting toxicities (DLT); however, the majority of TNBC patients (90%) in this trial presented with grade 3 adverse events (AEs) with limited evidence of antitumor activity [157]. A first-in-human trial that included multimodality treatments including chemoradiation, NK cell therapy, typhoid conjugate vaccine (TCV), and a PD-L1 inhibitor as third-line therapy for mTNBC (NCT03387085) found the combination treatment to be safe and well tolerated, and it achieved a 56% ORR in early efficacy results [158]. These encouraging results suggest that multimodal treatment will be the way forward for the management of recurrent and metastatic TNBC and may lead to the development of additional multimodality clinical trials.

4. Conclusions and Future Perspectives

In conclusion, while immunotherapy has made significant strides in the treatment of TNBC, there is still much progress to be made in optimizing these approaches for broader clinical benefit. The successes seen with ICIs have paved the way for more innovative strategies, yet challenges such as limited response rates and the development of resistance must be addressed. Moving forward, the integration of immunotherapy into “window of opportunity” trials could provide crucial insights into the tumor immune microenvironment and help identify biomarkers that predict responses to treatment. Furthermore, combination therapies hold considerable promise in enhancing the efficacy of immunotherapies in TNBC. Combining immune checkpoint inhibitors with chemotherapy, targeted therapies, radiation, or other immunomodulatory agents such as cytokines and oncolytic viruses may enhance the immune response and overcome resistance mechanisms. The use of adoptive cell therapies, such as CAR-T cells and TILs, may also be explored in combination with immune checkpoint inhibitors to boost the antitumor immune response. Additionally, the future of immunotherapy in TNBC will likely involve more personalized approaches, tailoring treatments based on individual tumor profiles and immune characteristics. Identifying new biomarkers, improving patient selection, and understanding the mechanisms of resistance will be critical in advancing the field. Continued exploration of novel immunotherapeutic strategies, such as cancer vaccines and bispecific antibodies, may further expand the treatment arsenal for TNBC. While immunotherapy has shown promise in TNBC, its full potential will likely be realized through the strategic design of combination therapies and personalized treatment approaches informed by ongoing research.
The advancements in immunotherapy have brought renewed optimism for women battling local, recurrent, and metastatic TNBC. Despite the success of integrating immunogenic chemotherapy with ICIs, particularly in early-stage TNBC, trials evaluating adjuvant immune checkpoint inhibitors in operable TNBC like IMpassion030 (NCT03498716) have yielded unsatisfactory outcomes. These innovative treatments provide additional therapeutic options for patients who have undergone extensive prior therapies and developed resistance. Over the past few years, a growing number of small-molecule inhibitors including tyrosine kinase inhibitors (EGFR and VEGFR), serine/threonine kinase inhibitors (ATM, ATR, AKT, CDK1, CDK4/6, CHK1, DNA-PKcs, mTOR, PI3K, and WEE1), dual specific kinase inhibitors (TTK1, MEK), and proteasome, PARP, and epigenetic (HDAC) inhibitors have been tested in TNBC as monotherapy or in combination with other targeted agents or ICI [159,160]. While several of these therapeutic combinations are still in the experimental stage or undergoing clinical trials, they represent promising avenues for the treatment of TNBC (Table 3). Despite the clinical success of targeted small-molecule treatments for TNBC, drug resistance is still an ongoing challenge. Other possibilities include combination treatments, novel mutation inhibitors, and multi-targeted drugs. Novel therapeutic targets, such as BUB1, LIG4, Hh, and XPO1, are being investigated in preclinical or clinical studies for targeting TNBC [160,161,162,163,164]. It is anticipated that in the near future, these small-molecule inhibitors and immunotherapy will be able to work together to increase the antitumor activity of these drugs. Combination therapy may prove effective, but uncertainties still exist relating to method, sequence, dosage, and duration with a careful eye toward balancing toxicity and affordability.
Integrating artificial intelligence (AI) in treatment planning may revolutionize the prediction of therapeutic outcomes, enabling personalized precision medicine. High-throughput sequencing and AI can identify novel molecular markers and gene signatures, aiding clinicians in selecting the most effective patient-specific therapies. This may then assist the clinical care team in optimally selecting a “tailored patient-specific” therapy that is most likely to work, thus opening new horizons in precision medicine [165]. This will require scientists, organizations, and medical professionals to work together to develop databases, remove technological obstacles, and support the creation of AI-assisted systems that can precisely identify the target populations/patients, predict the efficacy and prognosis, and strongly support the use of AI-assisted treatment. We anticipate that with the development of novel drug discovery/prediction datasets, large-scale genomic/genetic datasets, and immune signatures combined with large multicenter clinical trials will soon make significant strides in treating TNBC efficiently [166,167,168,169,170].

Author Contributions

S.N. and S.S.: conceptualization; S.S.: writing—original draft; S.T., C.S. and S.N.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NCI R21 (1R21CA252010-01A1), HFHS Research Administration Start Up, the HFHS Proposal Development Award, the Game on Cancer Award and HFHS-Radiation Oncology Start Up to Shyam Nyati.

Acknowledgments

We thank HFCI for providing a Translational Oncology Postdoctoral Fellowship to Sushmitha Sriramulu.

Conflicts of Interest

S.S., S.T., S.N.: no COIs; C.S.: Exact Sciences (paid consultant—no direct conflict).

References

  1. Zagami, P.; Carey, L.A. Triple negative breast cancer: Pitfalls and progress. NPJ Breast Cancer 2022, 8, 95. [Google Scholar] [CrossRef]
  2. Bauer, K.R.; Brown, M.; Cress, R.D.; Parise, C.A.; Caggiano, V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: A population-based study from the California cancer Registry. Cancer 2007, 109, 1721–1728. [Google Scholar] [CrossRef] [PubMed]
  3. Loizides, S.; Constantinidou, A. Triple negative breast cancer: Immunogenicity, tumor microenvironment, and immunotherapy. Front. Genet. 2022, 13, 1095839. [Google Scholar] [CrossRef] [PubMed]
  4. Liedtke, C.; Mazouni, C.; Hess, K.R.; Andre, F.; Tordai, A.; Mejia, J.A.; Symmans, W.F.; Gonzalez-Angulo, A.M.; Hennessy, B.; Green, M.; et al. Response to Neoadjuvant Therapy and Long-Term Survival in Patients with Triple-Negative Breast Cancer. J. Clin. Oncol. 2023, 41, 1809–1815. [Google Scholar] [CrossRef] [PubMed]
  5. Blows, F.M.; Driver, K.E.; Schmidt, M.K.; Broeks, A.; van Leeuwen, F.E.; Wesseling, J.; Cheang, M.C.; Gelmon, K.; Nielsen, T.O.; Blomqvist, C.; et al. Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: A collaborative analysis of data for 10,159 cases from 12 studies. PLoS Med. 2010, 7, e1000279. [Google Scholar] [CrossRef] [PubMed]
  6. Cortazar, P.; Zhang, L.; Untch, M.; Mehta, K.; Costantino, J.P.; Wolmark, N.; Bonnefoi, H.; Cameron, D.; Gianni, L.; Valagussa, P.; et al. Pathological complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014, 384, 164–172. [Google Scholar] [CrossRef]
  7. Burstein, M.D.; Tsimelzon, A.; Poage, G.M.; Covington, K.R.; Contreras, A.; Fuqua, S.A.; Savage, M.I.; Osborne, C.K.; Hilsenbeck, S.G.; Chang, J.C.; et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin. Cancer Res. 2015, 21, 1688–1698. [Google Scholar] [CrossRef]
  8. Yin, L.; Duan, J.J.; Bian, X.W.; Yu, S.C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef]
  9. Speers, C.; Tsimelzon, A.; Sexton, K.; Herrick, A.M.; Gutierrez, C.; Culhane, A.; Quackenbush, J.; Hilsenbeck, S.; Chang, J.; Brown, P. Identification of novel kinase targets for the treatment of estrogen receptor-negative breast cancer. Clin. Cancer Res. 2009, 15, 6327–6340. [Google Scholar] [CrossRef]
  10. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef]
  11. Lehmann, B.D.; Jovanovic, B.; Chen, X.; Estrada, M.V.; Johnson, K.N.; Shyr, Y.; Moses, H.L.; Sanders, M.E.; Pietenpol, J.A. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368. [Google Scholar] [CrossRef]
  12. Pavese, F.; Capoluongo, E.D.; Muratore, M.; Minucci, A.; Santonocito, C.; Fuso, P.; Concolino, P.; Di Stasio, E.; Carbognin, L.; Tiberi, G.; et al. BRCA Mutation Status in Triple-Negative Breast Cancer Patients Treated with Neoadjuvant Chemotherapy: A Pivotal Role for Treatment Decision-Making. Cancers 2022, 14, 4571. [Google Scholar] [CrossRef] [PubMed]
  13. Nolan, E.; Savas, P.; Policheni, A.N.; Darcy, P.K.; Vaillant, F.; Mintoff, C.P.; Dushyanthen, S.; Mansour, M.; Pang, J.-M.B.; Fox, S.B. Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer. Sci. Transl. Med. 2017, 9, eaal4922. [Google Scholar] [CrossRef] [PubMed]
  14. Disis, M.L.; Stanton, S.E. Triple-negative breast cancer: Immune modulation as the new treatment paradigm. Am. Soc. Clin. Oncol. Educ. Book 2015, 35, e25–e30. [Google Scholar] [CrossRef] [PubMed]
  15. Safonov, A.; Jiang, T.; Bianchini, G.; Győrffy, B.; Karn, T.; Hatzis, C.; Pusztai, L. Immune Gene Expression Is Associated with Genomic Aberrations in Breast Cancer. Cancer Res. 2017, 77, 3317–3324. [Google Scholar] [CrossRef] [PubMed]
  16. Fan, Y.; He, S. The Characteristics of Tumor Microenvironment in Triple Negative Breast Cancer. Cancer Manag. Res. 2022, 14, 1–17. [Google Scholar] [CrossRef]
  17. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Dieras, V.; Hegg, R.; Im, S.A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
  18. Lin, Y.Y.; Gao, H.F.; Yang, X.; Zhu, T.; Zheng, X.X.; Ji, F.; Zhang, L.L.; Yang, C.Q.; Yang, M.; Li, J.Q.; et al. Neoadjuvant therapy in triple-negative breast cancer: A systematic review and network meta-analysis. Breast 2022, 66, 126–135. [Google Scholar] [CrossRef]
  19. Loibl, S.; O’Shaughnessy, J.; Untch, M.; Sikov, W.M.; Rugo, H.S.; McKee, M.D.; Huober, J.; Golshan, M.; von Minckwitz, G.; Maag, D.; et al. Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): A randomised, phase 3 trial. Lancet Oncol. 2018, 19, 497–509. [Google Scholar] [CrossRef]
  20. Valencia, G.A.; Rioja, P.; Morante, Z.; Ruiz, R.; Fuentes, H.; Castaneda, C.A.; Vidaurre, T.; Neciosup, S.; Gomez, H.L. Immunotherapy in triple-negative breast cancer: A literature review and new advances. World J. Clin. Oncol. 2022, 13, 219–236. [Google Scholar] [CrossRef]
  21. Li, L.; Zhang, F.; Liu, Z.; Fan, Z. Immunotherapy for Triple-Negative Breast Cancer: Combination Strategies to Improve Outcome. Cancers 2023, 15, 321. [Google Scholar] [CrossRef] [PubMed]
  22. Shah, M.; Osgood, C.L.; Amatya, A.K.; Fiero, M.H.; Pierce, W.F.; Nair, A.; Herz, J.; Robertson, K.J.; Mixter, B.D.; Tang, S.; et al. FDA Approval Summary: Pembrolizumab for Neoadjuvant and Adjuvant Treatment of Patients with High-Risk Early-Stage Triple-Negative Breast Cancer. Clin. Cancer Res. 2022, 28, 5249–5253. [Google Scholar] [CrossRef] [PubMed]
  23. Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Pérez-Gracia, J.L.; Rodríguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castañón, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [PubMed]
  24. Lee, S.; Margolin, K. Cytokines in cancer immunotherapy. Cancers 2011, 3, 3856–3893. [Google Scholar] [CrossRef]
  25. Petrina, M.; Martin, J.; Basta, S. Granulocyte macrophage colony-stimulating factor has come of age: From a vaccine adjuvant to antiviral immunotherapy. Cytokine Growth Factor. Rev. 2021, 59, 101–110. [Google Scholar] [CrossRef]
  26. Deckers, J.; Anbergen, T.; Hokke, A.M.; de Dreu, A.; Schrijver, D.P.; de Bruin, K.; Toner, Y.C.; Beldman, T.J.; Spangler, J.B.; de Greef, T.F.A.; et al. Engineering cytokine therapeutics. Nat. Rev. Bioeng. 2023, 1, 286–303. [Google Scholar] [CrossRef]
  27. Nguyen, K.G.; Vrabel, M.R.; Mantooth, S.M.; Hopkins, J.J.; Wagner, E.S.; Gabaldon, T.A.; Zaharoff, D.A. Localized Interleukin-12 for Cancer Immunotherapy. Front. Immunol. 2020, 11, 575597. [Google Scholar] [CrossRef]
  28. Abdou, Y.; Goudarzi, A.; Yu, J.X.; Upadhaya, S.; Vincent, B.; Carey, L.A. Immunotherapy in triple negative breast cancer: Beyond checkpoint inhibitors. npj Breast Cancer 2022, 8, 121. [Google Scholar] [CrossRef]
  29. Walker, J.M.; Rolig, A.S.; Charych, D.H.; Hoch, U.; Kasiewicz, M.J.; Rose, D.C.; McNamara, M.J.; Hilgart-Martiszus, I.F.; Redmond, W.L. NKTR-214 immunotherapy synergizes with radiotherapy to stimulate systemic CD8(+) T cell responses capable of curing multi-focal cancer. J. Immunother. Cancer 2020, 8, e000464. [Google Scholar] [CrossRef]
  30. Cavazzoni, A.; Digiacomo, G. Role of Cytokines and Other Soluble Factors in Tumor Development: Rationale for New Therapeutic Strategies. Cells 2023, 12, 2532. [Google Scholar] [CrossRef]
  31. Conlon, K.C.; Miljkovic, M.D.; Waldmann, T.A. Cytokines in the Treatment of Cancer. J. Interferon Cytokine Res. 2019, 39, 6–21. [Google Scholar] [CrossRef] [PubMed]
  32. Dutcher, J.P.; Schwartzentruber, D.J.; Kaufman, H.L.; Agarwala, S.S.; Tarhini, A.A.; Lowder, J.N.; Atkins, M.B. High dose interleukin-2 (Aldesleukin)—Expert consensus on best management practices-2014. J. ImmunoTherapy Cancer 2014, 2, 26. [Google Scholar] [CrossRef] [PubMed]
  33. Page, D.B.; Pucilowska, J.; Sanchez, K.G.; Conrad, V.K.; Conlin, A.K.; Acheson, A.K.; Perlewitz, K.S.; Imatani, J.H.; Aliabadi-Wahle, S.; Moxon, N.; et al. A Phase Ib Study of Preoperative, Locoregional IRX-2 Cytokine Immunotherapy to Prime Immune Responses in Patients with Early-Stage Breast Cancer. Clin. Cancer Res. 2020, 26, 1595–1605. [Google Scholar] [CrossRef] [PubMed]
  34. Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef] [PubMed]
  35. Wesolowski, J.; Tankiewicz-Kwedlo, A.; Pawlak, D. Modern Immunotherapy in the Treatment of Triple-Negative Breast Cancer. Cancers 2022, 14, 3860. [Google Scholar] [CrossRef]
  36. Gemmete, J.J.; Mukherji, S.K. Trastuzumab (herceptin). AJNR Am. J. Neuroradiol. 2011, 32, 1373–1374. [Google Scholar] [CrossRef]
  37. Baez Navarro, X.; van den Ender, N.S.; Nguyen, A.; Sinke, R.; Westenend, P.; van Brakel, J.B.; Stobbe, C.; Westerga, J.; van Deurzen, C.H.M. HER2-low and tumor infiltrating lymphocytes in triple negative breast cancer: Are they mutually connected? Eur. J. Cancer 2024, 200, 113844. [Google Scholar] [CrossRef]
  38. Garcia, J.; Hurwitz, H.I.; Sandler, A.B.; Miles, D.; Coleman, R.L.; Deurloo, R.; Chinot, O.L. Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat. Rev. 2020, 86, 102017. [Google Scholar] [CrossRef]
  39. Sasich, L.D.; Sukkari, S.R. The US FDAs withdrawal of the breast cancer indication for Avastin (bevacizumab). Saudi Pharm. J. 2012, 20, 381–385. [Google Scholar] [CrossRef]
  40. Luo, C.; Wang, P.; He, S.; Zhu, J.; Shi, Y.; Wang, J. Progress and Prospect of Immunotherapy for Triple-Negative Breast Cancer. Front. Oncol. 2022, 12, 919072. [Google Scholar] [CrossRef]
  41. Chang, H.L.; Schwettmann, B.; McArthur, H.L.; Chan, I.S. Antibody-drug conjugates in breast cancer: Overcoming resistance and boosting immune response. J. Clin. Investig. 2023, 133, e172156. [Google Scholar] [CrossRef] [PubMed]
  42. Panowski, S.; Bhakta, S.; Raab, H.; Polakis, P.; Junutula, J.R. Site-specific antibody drug conjugates for cancer therapy. MAbs 2014, 6, 34–45. [Google Scholar] [CrossRef] [PubMed]
  43. Vonderheide, R.H. CD40 Agonist Antibodies in Cancer Immunotherapy. Annu. Rev. Med. 2020, 71, 47–58. [Google Scholar] [CrossRef] [PubMed]
  44. Azar, I.; Alkassis, S.; Fukui, J.; Alsawah, F.; Fedak, K.; Al Hallak, M.N.; Sukari, A.; Nagasaka, M. Spotlight on Trastuzumab Deruxtecan (DS-8201,T-DXd) for HER2 Mutation Positive Non-Small Cell Lung Cancer. Lung Cancer 2021, 12, 103–114. [Google Scholar] [CrossRef] [PubMed]
  45. Mark, C.; Lee, J.S.; Cui, X.; Yuan, Y. Antibody-Drug Conjugates in Breast Cancer: Current Status and Future Directions. Int. J. Mol. Sci. 2023, 24, 3726. [Google Scholar] [CrossRef]
  46. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N. Engl. J. Med. 2022, 387, 9–20. [Google Scholar] [CrossRef]
  47. Bartsch, R.; Berghoff, A.S.; Furtner, J.; Marhold, M.; Bergen, E.S.; Roider-Schur, S.; Starzer, A.M.; Forstner, H.; Rottenmanner, B.; Dieckmann, K.; et al. Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: A single-arm, phase 2 trial. Nat. Med. 2022, 28, 1840–1847. [Google Scholar] [CrossRef]
  48. Shi, F.; Liu, Y.; Zhou, X.; Shen, P.; Xue, R.; Zhang, M. Disitamab vedotin: A novel antibody-drug conjugates for cancer therapy. Drug Deliv. 2022, 29, 1335–1344. [Google Scholar] [CrossRef]
  49. Fenn, K.M.; Kalinsky, K. Sacituzumab govitecan: Antibody-drug conjugate in triple-negative breast cancer and other solid tumors. Drugs Today 2019, 55, 575–585. [Google Scholar] [CrossRef]
  50. Bardia, A.; Rugo, H.S.; Tolaney, S.M.; Loirat, D.; Punie, K.; Oliveira, M.; Brufsky, A.; Kalinsky, K.; Cortés, J.; Shaughnessy, J.O.; et al. Final Results From the Randomized Phase III ASCENT Clinical Trial in Metastatic Triple-Negative Breast Cancer and Association of Outcomes by Human Epidermal Growth Factor Receptor 2 and Trophoblast Cell Surface Antigen 2 Expression. J. Clin. Oncol. 2024, 42, 1738–1744. [Google Scholar] [CrossRef]
  51. Rizzo, A.; Cusmai, A.; Acquafredda, S.; Rinaldi, L.; Palmiotti, G. Ladiratuzumab vedotin for metastatic triple negative cancer: Preliminary results, key challenges, and clinical potential. Expert. Opin. Investig. Drugs 2022, 31, 495–498. [Google Scholar] [CrossRef] [PubMed]
  52. Modi, S.; Pusztai, L.; Forero, A.; Mita, M.; Miller, K.; Weise, A.; Krop, I.; Burris, H., III; Kalinsky, K.; Tsai, M.; et al. Abstract PD3-14: Phase 1 study of the antibody-drug conjugate SGN-LIV1A in patients with heavily pretreated triple-negative metastatic breast cancer. Cancer Res. 2018, 78, PD3-14. [Google Scholar] [CrossRef]
  53. Tsai, M.; Han, H.S.; Montero, A.J.; Tkaczuk, K.H.; Assad, H.; Pusztai, L.; Hurvitz, S.A.; Wilks, S.T.; Specht, J.M.; Nanda, R.; et al. 259P Weekly ladiratuzumab vedotin monotherapy for metastatic triple-negative breast cancer. Ann. Oncol. 2021, 32, S474–S475. [Google Scholar] [CrossRef]
  54. Ma, F.; Qiu, F.; Tong, Z.; Wang, J.; Tan, Y.; Bai, R.; Zhou, Q.; Xing, X. Preliminary results from a first-in-human study of ESG401, a trophoblast cell-surface antigen 2 (TROP2) antibody drug conjugate (ADC), in patients with locally advanced/metastatic solid tumors. J. Clin. Oncol. 2023, 41, 1100. [Google Scholar] [CrossRef]
  55. Ma, F.; Qiu, F.; Tong, Z.; Shi, Y.; Yu, G.; Wu, X.; Wang, H.; Wang, J.; Yang, H.; Liu, A.; et al. ESG401, a trophoblast cell-surface antigen 2 (TROP2) antibody drug conjugate (ADC), for the treatment of first-line metastatic triple negative breast cancer (mTNBC). J. Clin. Oncol. 2024, 42, e13132. [Google Scholar] [CrossRef]
  56. Bardia, A.; Pusztai, L.; Albain, K.; Ciruelos, E.M.; Im, S.A.; Hershman, D.; Kalinsky, K.; Isaacs, C.; Loirat, D.; Testa, L.; et al. TROPION-Breast03: A randomized phase III global trial of datopotamab deruxtecan ± durvalumab in patients with triple-negative breast cancer and residual invasive disease at surgical resection after neoadjuvant therapy. Ther. Adv. Med. Oncol. 2024, 16, 17588359241248336. [Google Scholar] [CrossRef]
  57. Bardia, A.; Krop, I.E.; Kogawa, T.; Juric, D.; Tolcher, A.W.; Hamilton, E.P.; Mukohara, T.; Lisberg, A.; Shimizu, T.; Spira, A.I.; et al. Datopotamab Deruxtecan in Advanced or Metastatic HR+/HER2– and Triple-Negative Breast Cancer: Results From the Phase I TROPION-PanTumor01 Study. J. Clin. Oncol. 2024, 42, 2281–2294. [Google Scholar] [CrossRef]
  58. Byun, D.J.; Wolchok, J.D.; Rosenberg, L.M.; Girotra, M. Cancer immunotherapy—Immune checkpoint blockade and associated endocrinopathies. Nat. Rev. Endocrinol. 2017, 13, 195–207. [Google Scholar] [CrossRef]
  59. Liu, Y.; Hu, Y.; Xue, J.; Li, J.; Yi, J.; Bu, J.; Zhang, Z.; Qiu, P.; Gu, X. Correction: Advances in immunotherapy for triple-negative breast cancer. Mol. Cancer 2023, 22, 154. [Google Scholar] [CrossRef]
  60. Ahmed, F.S.; Gaule, P.; McGuire, J.; Patel, K.; Blenman, K.; Pusztai, L.; Rimm, D.L. PD-L1 Protein Expression on Both Tumor Cells and Macrophages are Associated with Response to Neoadjuvant Durvalumab with Chemotherapy in Triple-negative Breast Cancer. Clin. Cancer Res. 2020, 26, 5456–5461. [Google Scholar] [CrossRef]
  61. Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [Google Scholar] [CrossRef] [PubMed]
  62. Nanda, R.; Chow, L.Q.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients with Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef] [PubMed]
  63. Adams, S.; Loi, S.; Toppmeyer, D.; Cescon, D.W.; De Laurentiis, M.; Nanda, R.; Winer, E.P.; Mukai, H.; Tamura, K.; Armstrong, A.; et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: Cohort B of the phase II KEYNOTE-086 study. Ann. Oncol. 2019, 30, 405–411. [Google Scholar] [CrossRef] [PubMed]
  64. Ganguly, S.; Gogia, A. Pembrolizumab monotherapy in advanced triple-negative breast cancer. Lancet Oncol. 2021, 22, e224. [Google Scholar] [CrossRef] [PubMed]
  65. Bian, L.; Zhang, H.; Wang, T.; Zhang, S.; Song, H.; Xu, M.; Yao, S.; Jiang, Z. JS001, an anti-PD-1 mAb for advanced triple negative breast cancer patients after multi-line systemic therapy in a phase I trial. Ann. Transl. Med. 2019, 7, 435. [Google Scholar] [CrossRef]
  66. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kümmel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef]
  67. Emens, L.A.; Cruz, C.; Eder, J.P.; Braiteh, F.; Chung, C.; Tolaney, S.M.; Kuter, I.; Nanda, R.; Cassier, P.A.; Delord, J.P.; et al. Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients with Metastatic Triple-Negative Breast Cancer: A Phase 1 Study. JAMA Oncol. 2019, 5, 74–82. [Google Scholar] [CrossRef]
  68. Dirix, L.Y.; Takacs, I.; Jerusalem, G.; Nikolinakos, P.; Arkenau, H.T.; Forero-Torres, A.; Boccia, R.; Lippman, M.E.; Somer, R.; Smakal, M.; et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b JAVELIN Solid Tumor study. Breast Cancer Res. Treat. 2018, 167, 671–686. [Google Scholar] [CrossRef]
  69. Jacobs, F.; Agostinetto, E.; Miggiano, C.; De Sanctis, R.; Zambelli, A.; Santoro, A. Hope and Hype around Immunotherapy in Triple-Negative Breast Cancer. Cancers 2023, 15, 2933. [Google Scholar] [CrossRef]
  70. Hosseini, A.; Gharibi, T.; Marofi, F.; Babaloo, Z.; Baradaran, B. CTLA-4: From mechanism to autoimmune therapy. Int. Immunopharmacol. 2020, 80, 106221. [Google Scholar] [CrossRef]
  71. Peng, Z.; Su, P.; Yang, Y.; Yao, X.; Zhang, Y.; Jin, F.; Yang, B. Identification of CTLA-4 associated with tumor microenvironment and competing interactions in triple negative breast cancer by co-expression network analysis. J. Cancer 2020, 11, 6365–6375. [Google Scholar] [CrossRef] [PubMed]
  72. Kaewkangsadan, V.; Verma, C.; Eremin, J.M.; Cowley, G.; Ilyas, M.; Eremin, O. Tumour-draining axillary lymph nodes in patients with large and locally advanced breast cancers undergoing neoadjuvant chemotherapy (NAC): The crucial contribution of immune cells (effector, regulatory) and cytokines (Th1, Th2) to immune-mediated tumour cell death induced by NAC. BMC Cancer 2018, 18, 123. [Google Scholar] [CrossRef]
  73. Zhang, X.; Sharma, P.K.; Peter Goedegebuure, S.; Gillanders, W.E. Personalized cancer vaccines: Targeting the cancer mutanome. Vaccine 2017, 35, 1094–1100. [Google Scholar] [CrossRef] [PubMed]
  74. Burke, E.E.; Kodumudi, K.; Ramamoorthi, G.; Czerniecki, B.J. Vaccine Therapies for Breast Cancer. Surg. Oncol. Clin. N. Am. 2019, 28, 353–367. [Google Scholar] [CrossRef] [PubMed]
  75. Mittendorf, E.A.; Ardavanis, A.; Litton, J.K.; Shumway, N.M.; Hale, D.F.; Murray, J.L.; Perez, S.A.; Ponniah, S.; Baxevanis, C.N.; Papamichail, M.; et al. Primary analysis of a prospective, randomized, single-blinded phase II trial evaluating the HER2 peptide GP2 vaccine in breast cancer patients to prevent recurrence. Oncotarget 2016, 7, 66192–66201. [Google Scholar] [CrossRef]
  76. Mittendorf, E.A.; Clifton, G.T.; Holmes, J.P.; Schneble, E.; van Echo, D.; Ponniah, S.; Peoples, G.E. Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients. Ann. Oncol. 2014, 25, 1735–1742. [Google Scholar] [CrossRef]
  77. Kalli, K.R.; Block, M.S.; Kasi, P.M.; Erskine, C.L.; Hobday, T.J.; Dietz, A.; Padley, D.; Gustafson, M.P.; Shreeder, B.; Puglisi-Knutson, D.; et al. Folate Receptor Alpha Peptide Vaccine Generates Immunity in Breast and Ovarian Cancer Patients. Clin. Cancer Res. 2018, 24, 3014–3025. [Google Scholar] [CrossRef]
  78. Heery, C.R.; Ibrahim, N.K.; Arlen, P.M.; Mohebtash, M.; Murray, J.L.; Koenig, K.; Madan, R.A.; McMahon, S.; Marté, J.L.; Steinberg, S.M.; et al. Docetaxel Alone or in Combination with a Therapeutic Cancer Vaccine (PANVAC) in Patients with Metastatic Breast Cancer: A Randomized Clinical Trial. JAMA Oncol. 2015, 1, 1087–1095. [Google Scholar] [CrossRef]
  79. Mohebtash, M.; Tsang, K.Y.; Madan, R.A.; Huen, N.Y.; Poole, D.J.; Jochems, C.; Jones, J.; Ferrara, T.; Heery, C.R.; Arlen, P.M.; et al. A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clin. Cancer Res. 2011, 17, 7164–7173. [Google Scholar] [CrossRef]
  80. Svane, I.M.; Pedersen, A.E.; Johansen, J.S.; Johnsen, H.E.; Nielsen, D.; Kamby, C.; Ottesen, S.; Balslev, E.; Gaarsdal, E.; Nikolajsen, K.; et al. Vaccination with p53 peptide-pulsed dendritic cells is associated with disease stabilization in patients with p53 expressing advanced breast cancer; monitoring of serum YKL-40 and IL-6 as response biomarkers. Cancer Immunol. Immunother. 2007, 56, 1485–1499. [Google Scholar] [CrossRef]
  81. Qi, C.J.; Ning, Y.L.; Han, Y.S.; Min, H.Y.; Ye, H.; Zhu, Y.L.; Qian, K.Q. Autologous dendritic cell vaccine for estrogen receptor (ER)/progestin receptor (PR) double-negative breast cancer. Cancer Immunol. Immunother. 2012, 61, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  82. Avigan, D.; Vasir, B.; Gong, J.; Borges, V.; Wu, Z.; Uhl, L.; Atkins, M.; Mier, J.; McDermott, D.; Smith, T.; et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses. Clin. Cancer Res. 2004, 10, 4699–4708. [Google Scholar] [CrossRef] [PubMed]
  83. Vonderheide, R.H.; Glennie, M.J. Agonistic CD40 antibodies and cancer therapy. Clin. Cancer Res. 2013, 19, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  84. Budd, G.T.; Johnson, J.M.; Rhoades, E.E.; Moore, H.C.F.; Kruse, M.L.; Roesch, E.E.; Abraham, J.; Elliott, B.; Lach, D.; Tuohy, V.K. Phase I trial of an alpha-lactalbumin vaccine in patients with moderate- to high-risk operable triple-negative breast cancer (TNBC). J. Clin. Oncol. 2022, 40, TPS1125. [Google Scholar] [CrossRef]
  85. Disis, M.; Liu, Y.; Stanton, S.; Gwin, W.; Coveler, A.; Liao, J.; Childs, J.; Cecil, D. 546 A phase I dose escalation study of STEMVAC, a multi-antigen, multi-epitope Th1 selective plasmid-based vaccine, targeting stem cell associated proteins in patients with advanced breast cancer. J. ImmunoTherapy Cancer 2022, 10, A571. [Google Scholar] [CrossRef]
  86. Zhao, L.; Cao, Y.J. Engineered T Cell Therapy for Cancer in the Clinic. Front. Immunol. 2019, 10, 2250. [Google Scholar] [CrossRef]
  87. Xu, N.; Palmer, D.C.; Robeson, A.C.; Shou, P.; Bommiasamy, H.; Laurie, S.J.; Willis, C.; Dotti, G.; Vincent, B.G.; Restifo, N.P.; et al. STING agonist promotes CAR T cell trafficking and persistence in breast cancer. J. Exp. Med. 2021, 218, e20200844. [Google Scholar] [CrossRef]
  88. 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]
  89. Liu, Y.R.; Jiang, Y.Z.; Xu, X.E.; Yu, K.D.; Jin, X.; Hu, X.; Zuo, W.J.; Hao, S.; Wu, J.; Liu, G.Y.; et al. Comprehensive transcriptome analysis identifies novel molecular subtypes and subtype-specific RNAs of triple-negative breast cancer. Breast Cancer Res. 2016, 18, 33. [Google Scholar] [CrossRef]
  90. Ghosn, M.; Cheema, W.; Zhu, A.; Livschitz, J.; Maybody, M.; Boas, F.E.; Santos, E.; Kim, D.; Beattie, J.A.; Offin, M.; et al. Image-guided interventional radiological delivery of chimeric antigen receptor (CAR) T cells for pleural malignancies in a phase I/II clinical trial. Lung Cancer 2022, 165, 1–9. [Google Scholar] [CrossRef]
  91. 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]
  92. Kufe, D.W. MUC1-C oncoprotein as a target in breast cancer: Activation of signaling pathways and therapeutic approaches. Oncogene 2013, 32, 1073–1081. [Google Scholar] [CrossRef]
  93. Bamdad, C.C.; Yuan, Y.; Specht, J.M.; Stewart, A.K.; Smagghe, B.J.; Lin, S.C.-M.; Carter, M.G.; Synold, T.W.; Frankel, P.H.; Parekh, V.; et al. Phase I/II first-in-human CAR T–targeting MUC1 transmembrane cleavage product (MUC1*) in patients with metastatic breast cancer. J. Clin. Oncol. 2022, 40, TPS1130. [Google Scholar] [CrossRef]
  94. Gaglia, P.; Caldarola, B.; Bussone, R.; Potente, F.; Lauro, D.; Jayme, A.; Caldarola, L. Prognostic value of CEA and ferritin assay in breast cancer: A multivariate analysis. Eur. J. Cancer Clin. Oncol. 1988, 24, 1151–1155. [Google Scholar] [CrossRef]
  95. Phase Ia/Ib Trial of 2nd Generation Anti-CEA Designer T Cells in Metastatic Breast Cancer. 2008. Available online: https://clinicaltrials.gov/study/NCT00673829 (accessed on 17 June 2024).
  96. Luen, S.J.; Savas, P.; Fox, S.B.; Salgado, R.; Loi, S. Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology 2017, 49, 141–155. [Google Scholar] [CrossRef]
  97. Fukuhara, H.; Ino, Y.; Todo, T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 2016, 107, 1373–1379. [Google Scholar] [CrossRef]
  98. Thoidingjam, S.; Sriramulu, S.; Freytag, S.; Brown, S.L.; Kim, J.H.; Chetty, I.J.; Siddiqui, F.; Movsas, B.; Nyati, S. Oncolytic virus-based suicide gene therapy for cancer treatment: A perspective of the clinical trials conducted at Henry Ford Health. Transl. Med. Commun. 2023, 8, 11. [Google Scholar] [CrossRef]
  99. Zheng, M.; Huang, J.; Tong, A.; Yang, H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. Mol. Ther. Oncolytics 2019, 15, 234–247. [Google Scholar] [CrossRef]
  100. Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef]
  101. Kaufman, H.L.; Shalhout, S.Z.; Iodice, G. Talimogene Laherparepvec: Moving From First-In-Class to Best-In-Class. Front. Mol. Biosci. 2022, 9, 834841. [Google Scholar] [CrossRef]
  102. Kai, M.; Marx, A.N.; Liu, D.D.; Shen, Y.; Gao, H.; Reuben, J.M.; Whitman, G.; Krishnamurthy, S.; Ross, M.I.; Litton, J.K.; et al. A phase II study of talimogene laherparepvec for patients with inoperable locoregional recurrence of breast cancer. Sci. Rep. 2021, 11, 22242. [Google Scholar] [CrossRef]
  103. Nokisalmi, P.; Pesonen, S.; Escutenaire, S.; Särkioja, M.; Raki, M.; Cerullo, V.; Laasonen, L.; Alemany, R.; Rojas, J.; Cascallo, M.; et al. Oncolytic adenovirus ICOVIR-7 in patients with advanced and refractory solid tumors. Clin. Cancer Res. 2010, 16, 3035–3043. [Google Scholar] [CrossRef]
  104. Hemminki, O.; Parviainen, S.; Juhila, J.; Turkki, R.; Linder, N.; Lundin, J.; Kankainen, M.; Ristimäki, A.; Koski, A.; Liikanen, I.; et al. Immunological data from cancer patients treated with Ad5/3-E2F-Δ24-GMCSF suggests utility for tumor immunotherapy. Oncotarget 2015, 6, 4467–4481. [Google Scholar] [CrossRef]
  105. Monaco, M.L.; Idris, O.A.; Essani, K. Triple-Negative Breast Cancer: Basic Biology and Immuno-Oncolytic Viruses. Cancers 2023, 15, 2393. [Google Scholar] [CrossRef]
  106. Lauer, U.M.; Beil, J. Oncolytic viruses: Challenges and considerations in an evolving clinical landscape. Future Oncol. 2022, 18, 2713–2732. [Google Scholar] [CrossRef]
  107. Liu, M.; Peng, K.-W.; Federspiel, M.; Russell, S.; Brunton, B.; Zhou, Y.; Packiriswamy, N.; Hubbard, J.; Loprinzi, C.; Peethambaram, P.; et al. Abstract P6-21-03: Phase I trial of intratumoral (IT) administration of a NIS-expressing derivative manufactured from a genetically engineered strain of measles virus (MV). Cancer Res. 2019, 79, P6-21-03. [Google Scholar] [CrossRef]
  108. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef]
  109. Fabian, K.P.; Wolfson, B.; Hodge, J.W. From Immunogenic Cell Death to Immunogenic Modulation: Select Chemotherapy Regimens Induce a Spectrum of Immune-Enhancing Activities in the Tumor Microenvironment. Front. Oncol. 2021, 11, 728018. [Google Scholar] [CrossRef]
  110. Nedeljković, M.; Damjanović, A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge. Cells 2019, 8, 957. [Google Scholar] [CrossRef]
  111. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef]
  112. Jungles, K.M.; Holcomb, E.A.; Pearson, A.N.; Jungles, K.R.; Bishop, C.R.; Pierce, L.J.; Green, M.D.; Speers, C.W. Updates in combined approaches of radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Front. Oncol. 2022, 12, 1022542. [Google Scholar] [CrossRef]
  113. Obidiro, O.; Battogtokh, G.; Akala, E.O. Triple Negative Breast Cancer Treatment Options and Limitations: Future Outlook. Pharmaceutics 2023, 15, 1796. [Google Scholar] [CrossRef]
  114. Ragupathi, A.; Singh, M.; Perez, A.M.; Zhang, D. Targeting the BRCA1/2 deficient cancer with PARP inhibitors: Clinical outcomes and mechanistic insights. Front. Cell Dev. Biol. 2023, 11, 1133472. [Google Scholar] [CrossRef]
  115. Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Gonçalves, A.; Lee, K.-H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef]
  116. Robson, M.; Im, S.-A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 2017, 377, 523–533. [Google Scholar] [CrossRef]
  117. Barchiesi, G.; Roberto, M.; Verrico, M.; Vici, P.; Tomao, S.; Tomao, F. Emerging Role of PARP Inhibitors in Metastatic Triple Negative Breast Cancer. Current Scenario and Future Perspectives. Front. Oncol. 2021, 11, 769280. [Google Scholar] [CrossRef]
  118. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; Azambuja, E.d.; Fielding, A.; Balmaña, J.; et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef]
  119. Gupta, T.; Purington, N.; Liu, M.; Han, S.; Sledge, G.; Schapira, L.; Kurian, A.W. Incident comorbidities after tamoxifen or aromatase inhibitor therapy in a racially and ethnically diverse cohort of women with breast cancer. Breast Cancer Res. Treat. 2022, 196, 175–183. [Google Scholar] [CrossRef]
  120. Miglietta, F.; Cinquini, M.; Dieci, M.V.; Cortesi, L.; Criscitiello, C.; Montemurro, F.; Del Mastro, L.; Zambelli, A.; Biganzoli, L.; Levaggi, A.; et al. PARP-inhibitors for BRCA1/2-related advanced HER2-negative breast cancer: A meta-analysis and GRADE recommendations by the Italian Association of Medical Oncology. Breast 2022, 66, 293–304. [Google Scholar] [CrossRef]
  121. Vinayak, S.; Tolaney, S.M.; Schwartzberg, L.S.; Mita, M.M.; McCann, G.A.-L.; Tan, A.R.; Hendrickson, A.E.W.; Forero-Torres, A.; Anders, C.K.; Wulf, G.M.; et al. TOPACIO/Keynote-162: Niraparib + pembrolizumab in patients (pts) with metastatic triple-negative breast cancer (TNBC), a phase 2 trial. J. Clin. Oncol. 2018, 36, 1011. [Google Scholar] [CrossRef]
  122. Vinayak, S.; Tolaney, S.M.; Schwartzberg, L.; Mita, M.; McCann, G.; Tan, A.R.; Wahner-Hendrickson, A.E.; Forero, A.; Anders, C.; Wulf, G.M.; et al. Open-label Clinical Trial of Niraparib Combined with Pembrolizumab for Treatment of Advanced or Metastatic Triple-Negative Breast Cancer. JAMA Oncol. 2019, 5, 1132–1140. [Google Scholar] [CrossRef] [PubMed]
  123. Domchek, S.M.; Postel-Vinay, S.; Im, S.A.; Park, Y.H.; Delord, J.P.; Italiano, A.; Alexandre, J.; You, B.; Bastian, S.; Krebs, M.G.; et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): An open-label, multicentre, phase 1/2, basket study. Lancet Oncol. 2020, 21, 1155–1164. [Google Scholar] [CrossRef] [PubMed]
  124. Yap, T.A.; Bardia, A.; Dvorkin, M.; Galsky, M.D.; Beck, J.T.; Wise, D.R.; Karyakin, O.; Rubovszky, G.; Kislov, N.; Rohrberg, K.; et al. Avelumab Plus Talazoparib in Patients with Advanced Solid Tumors: The JAVELIN PARP Medley Nonrandomized Controlled Trial. JAMA Oncol. 2023, 9, 40–50. [Google Scholar] [CrossRef] [PubMed]
  125. Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015, 28, 690–714. [Google Scholar] [CrossRef] [PubMed]
  126. Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef] [PubMed]
  127. Cortes, J.; Cescon, D.W.; Rugo, H.S.; Nowecki, Z.; Im, S.-A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Holgado, E.; et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 2020, 396, 1817–1828. [Google Scholar] [CrossRef]
  128. Cortes, J.; Rugo, H.S.; Cescon, D.W.; Im, S.-A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Perez-Garcia, J.; Iwata, H.; et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2022, 387, 217–226. [Google Scholar] [CrossRef]
  129. Tolaney, S.M.; Kalinsky, K.; Kaklamani, V.G.; D’Adamo, D.R.; Aktan, G.; Tsai, M.L.; O’Regan, R.M.; Kaufman, P.A.; Wilks, S.T.; Andreopoulou, E.; et al. Eribulin Plus Pembrolizumab in Patients with Metastatic Triple-Negative Breast Cancer (ENHANCE 1): A Phase Ib/II Study. Clin. Cancer Res. 2021, 27, 3061–3068. [Google Scholar] [CrossRef]
  130. Adams, S.; Gatti-Mays, M.E.; Kalinsky, K.; Korde, L.A.; Sharon, E.; Amiri-Kordestani, L.; Bear, H.; McArthur, H.L.; Frank, E.; Perlmutter, J.; et al. Current Landscape of Immunotherapy in Breast Cancer: A Review. JAMA Oncol. 2019, 5, 1205–1214. [Google Scholar] [CrossRef]
  131. Schmid, P.; Rugo, H.S.; Adams, S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Henschel, V.; Molinero, L.; Chui, S.Y.; et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): Updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 44–59. [Google Scholar] [CrossRef]
  132. Emens, L.A.; Adams, S.; Barrios, C.H.; Diéras, V.; Iwata, H.; Loi, S.; Rugo, H.S.; Schneeweiss, A.; Winer, E.P.; Patel, S.; et al. First-line atezolizumab plus nab-paclitaxel for unresectable, locally advanced, or metastatic triple-negative breast cancer: IMpassion130 final overall survival analysis. Ann. Oncol. 2021, 32, 983–993. [Google Scholar] [CrossRef] [PubMed]
  133. Schmid, P.; Turner, N.C.; Barrios, C.H.; Isakoff, S.J.; Kim, S.B.; Sablin, M.P.; Saji, S.; Savas, P.; Vidal, G.A.; Oliveira, M.; et al. First-Line Ipatasertib, Atezolizumab, and Taxane Triplet for Metastatic Triple-Negative Breast Cancer: Clinical and Biomarker Results. Clin. Cancer Res. 2024, 30, 767–778. [Google Scholar] [CrossRef] [PubMed]
  134. Miles, D.; Gligorov, J.; André, F.; Cameron, D.; Schneeweiss, A.; Barrios, C.; Xu, B.; Wardley, A.; Kaen, D.; Andrade, L.; et al. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann. Oncol. 2021, 32, 994–1004. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, Y.; Chen, H.; Mo, H.; Hu, X.; Gao, R.; Zhao, Y.; Liu, B.; Niu, L.; Sun, X.; Yu, X.; et al. Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 2021, 39, 1578–1593.e8. [Google Scholar] [CrossRef] [PubMed]
  136. Voorwerk, L.; Slagter, M.; Horlings, H.M.; Sikorska, K.; van de Vijver, K.K.; de Maaker, M.; Nederlof, I.; Kluin, R.J.C.; Warren, S.; Ong, S.; et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: The TONIC trial. Nat. Med. 2019, 25, 920–928. [Google Scholar] [CrossRef]
  137. Nanda, R.; Liu, M.C.; Yau, C.; Shatsky, R.; Pusztai, L.; Wallace, A.; Chien, A.J.; Forero-Torres, A.; Ellis, E.; Han, H.; et al. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women with Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncol. 2020, 6, 676–684. [Google Scholar] [CrossRef]
  138. Davis, J.M.; Rushton, T.; Nsiah, F.; Stone, R.L.; Beavis, A.L.; Gaillard, S.L.; Dobi, A.; Fader, A.N. Long-term disease-free survival with chemotherapy and pembrolizumab in a patient with unmeasurable, advanced stage dedifferentiated endometrial carcinoma. Gynecol. Oncol. Rep. 2024, 53, 101380. [Google Scholar] [CrossRef]
  139. Loibl, S.; Untch, M.; Burchardi, N.; Huober, J.; Sinn, B.V.; Blohmer, J.U.; Grischke, E.M.; Furlanetto, J.; Tesch, H.; Hanusch, C.; et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: Clinical results and biomarker analysis of GeparNuevo study. Ann. Oncol. 2019, 30, 1279–1288. [Google Scholar] [CrossRef]
  140. Gianni, L.; Huang, C.S.; Egle, D.; Bermejo, B.; Zamagni, C.; Thill, M.; Anton, A.; Zambelli, S.; Bianchini, G.; Russo, S.; et al. Pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple-negative, early high-risk and locally advanced breast cancer: NeoTRIP Michelangelo randomized study. Ann. Oncol. 2022, 33, 534–543. [Google Scholar] [CrossRef]
  141. Mittendorf, E.A.; Zhang, H.; Barrios, C.H.; Saji, S.; Jung, K.H.; Hegg, R.; Koehler, A.; Sohn, J.; Iwata, H.; Telli, M.L.; et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): A randomised, double-blind, phase 3 trial. Lancet 2020, 396, 1090–1100. [Google Scholar] [CrossRef]
  142. Taylor, C.; Dodwell, D.; McGale, P.; Hills, R.K.; Berry, R.; Bradley, R.; Braybrooke, J.; Clarke, M.; Gray, R.; Holt, F.; et al. Radiotherapy to regional nodes in early breast cancer: An individual patient data meta-analysis of 14 324 women in 16 trials. Lancet 2023, 402, 1991–2003. [Google Scholar] [CrossRef] [PubMed]
  143. McGale, P.; Taylor, C.; Correa, C.; Cutter, D.; Duane, F.; Ewertz, M.; Gray, R.; Mannu, G.; Peto, R.; Whelan, T.; et al. Effect of radiotherapy after mastectomy and axillary surgery on 10-year recurrence and 20-year breast cancer mortality: Meta-analysis of individual patient data for 8135 women in 22 randomised trials. Lancet 2014, 383, 2127–2135. [Google Scholar] [CrossRef] [PubMed]
  144. Charpentier, M.; Spada, S.; Van Nest, S.J.; Demaria, S. Radiation therapy-induced remodeling of the tumor immune microenvironment. Semin. Cancer Biol. 2022, 86, 737–747. [Google Scholar] [CrossRef] [PubMed]
  145. Ho, A.Y.; Wright, J.L.; Blitzblau, R.C.; Mutter, R.W.; Duda, D.G.; Norton, L.; Bardia, A.; Spring, L.; Isakoff, S.J.; Chen, J.H.; et al. Optimizing Radiation Therapy to Boost Systemic Immune Responses in Breast Cancer: A Critical Review for Breast Radiation Oncologists. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 227–241. [Google Scholar] [CrossRef]
  146. McArthur, H.L.; Barker, C.A.; Gucalp, A.; Lebron-Zapata, L.; Wen, Y.H.; Phung, A.; Rodine, M.; Arnold, B.; Zhang, Z.; Ho, A. A single-arm, phase II study assessing the efficacy of pembrolizumab (pembro) plus radiotherapy (RT) in metastatic triple negative breast cancer (mTNBC). J. Clin. Oncol. 2018, 36, 14. [Google Scholar] [CrossRef]
  147. David, S.; Savas, P.; Siva, S.; White, M.; Neeson, M.W.; White, S.; Marx, G.; Cheuk, R.; Grogan, M.; Farrell, M.; et al. Abstract PD10-02: A randomised phase II trial of single fraction or multi-fraction SABR (stereotactic ablative body radiotherapy) with atezolizumab in patients with advanced triple negative breast cancer (AZTEC trial). Cancer Res. 2022, 82, PD10-02. [Google Scholar] [CrossRef]
  148. Sau, S.; Petrovici, A.; Alsaab, H.O.; Bhise, K.; Iyer, A.K. PDL-1 Antibody Drug Conjugate for Selective Chemo-Guided Immune Modulation of Cancer. Cancers 2019, 11, 232. [Google Scholar] [CrossRef]
  149. de Nonneville, A.; Finetti, P.; Boudin, L.; Denicolaï, E.; Birnbaum, D.; Mamessier, E.; Bertucci, F. Prognostic and Predictive Value of LIV1 Expression in Early Breast Cancer and by Molecular Subtype. Pharmaceutics 2023, 15, 938. [Google Scholar] [CrossRef]
  150. Meisel, J.L.; Pluard, T.J.; Vinayak, S.; Stringer-Reasor, E.M.; Brown-Glaberman, U.; Dillon, P.M.; Basho, R.K.; Varadarajan, R.; O’Shaughnessy, J.; Han, H.S.; et al. Phase 1b/2 study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of triple-negative breast cancer (SGNLVA-002, trial in progress). J. Clin. Oncol. 2022, 40, TPS1127. [Google Scholar] [CrossRef]
  151. Niu, G.; Chen, X. Vascular endothelial growth factor as an anti-angiogenic target for cancer therapy. Curr. Drug Targets 2010, 11, 1000–1017. [Google Scholar] [CrossRef]
  152. Zhang, Q.; Shao, B.; Tong, Z.; Ouyang, Q.; Wang, Y.; Xu, G.; Li, S.; Li, H. A phase Ib study of camrelizumab in combination with apatinib and fuzuloparib in patients with recurrent or metastatic triple-negative breast cancer. BMC Med. 2022, 20, 321. [Google Scholar] [CrossRef] [PubMed]
  153. Wu, S.Y.; Xu, Y.; Chen, L.; Fan, L.; Ma, X.Y.; Zhao, S.; Song, X.Q.; Hu, X.; Yang, W.T.; Chai, W.J.; et al. Combined angiogenesis and PD-1 inhibition for immunomodulatory TNBC: Concept exploration and biomarker analysis in the FUTURE-C-Plus trial. Mol. Cancer 2022, 21, 84. [Google Scholar] [CrossRef] [PubMed]
  154. Fujiwara, Y.; Mittra, A.; Naqash, A.R.; Takebe, N. A review of mechanisms of resistance to immune checkpoint inhibitors and potential strategies for therapy. Cancer Drug Resist. 2020, 3, 252–275. [Google Scholar] [CrossRef] [PubMed]
  155. Santa-Maria, C.A.; Kato, T.; Park, J.H.; Kiyotani, K.; Rademaker, A.; Shah, A.N.; Gross, L.; Blanco, L.Z.; Jain, S.; Flaum, L.; et al. A pilot study of durvalumab and tremelimumab and immunogenomic dynamics in metastatic breast cancer. Oncotarget 2018, 9, 18985–18996. [Google Scholar] [CrossRef] [PubMed]
  156. Buisseret, L.; Loirat, D.; Aftimos, P.; Maurer, C.; Punie, K.; Debien, V.; Kristanto, P.; Eiger, D.; Goncalves, A.; Ghiringhelli, F.; et al. Paclitaxel plus carboplatin and durvalumab with or without oleclumab for women with previously untreated locally advanced or metastatic triple-negative breast cancer: The randomized SYNERGY phase I/II trial. Nat. Commun. 2023, 14, 7018. [Google Scholar] [CrossRef]
  157. Hecht, J.R.; Raman, S.S.; Chan, A.; Kalinsky, K.; Baurain, J.F.; Jimenez, M.M.; Garcia, M.M.; Berger, M.D.; Lauer, U.M.; Khattak, A.; et al. Phase Ib study of talimogene laherparepvec in combination with atezolizumab in patients with triple negative breast cancer and colorectal cancer with liver metastases. ESMO Open 2023, 8, 100884. [Google Scholar] [CrossRef]
  158. Kistler, M.; Nangia, C.; To, C.; Sender, L.; Lee, J.; Jones, F.; Jafari, O.; Seery, T.; Rabizadeh, S.; Niazi, K.; et al. Abstract P5-04-02: Safety and efficacy from first-in-human immunotherapy combining NK and T cell activation with off-the-shelf high-affinity CD16 NK cell line (haNK) in patients with 2nd-line or greater metastatic triple-negative breast cancer (TNBC). Cancer Res. 2020, 80, P5-04-02. [Google Scholar] [CrossRef]
  159. Sriramulu, S.; Thoidingjam, S.; Brown, S.L.; Siddiqui, F.; Movsas, B.; Nyati, S. Molecular targets that sensitize cancer to radiation killing: From the bench to the bedside. Biomed. Pharmacother. 2023, 158, 114126. [Google Scholar] [CrossRef]
  160. Ou, Y.; Wang, M.; Xu, Q.; Sun, B.; Jia, Y. Small molecule agents for triple negative breast cancer: Current status and future prospects. Transl. Oncol. 2024, 41, 101893. [Google Scholar] [CrossRef]
  161. Sriramulu, S.; Thoidingjam, S.; Chen, W.M.; Hassan, O.; Siddiqui, F.; Brown, S.L.; Movsas, B.; Green, M.D.; Davis, A.J.; Speers, C.; et al. BUB1 regulates non-homologous end joining pathway to mediate radioresistance in triple-negative breast cancer. J. Exp. Clin. Cancer Res. 2024, 43, 163. [Google Scholar] [CrossRef]
  162. Cheng, Y.; Holloway, M.P.; Nguyen, K.; McCauley, D.; Landesman, Y.; Kauffman, M.G.; Shacham, S.; Altura, R.A. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivin-dependent oncogenic switch in triple-negative breast cancer. Mol. Cancer Ther. 2014, 13, 675–686. [Google Scholar] [CrossRef] [PubMed]
  163. Cicirò, Y.; Ragusa, D.; Sala, A. Expression of the checkpoint kinase BUB1 is a predictor of response to cancer therapies. Sci. Rep. 2024, 14, 4461. [Google Scholar] [CrossRef] [PubMed]
  164. Sriramulu, S.; Thoidingjam, S.; Siddiqui, F.; Brown, S.L.; Movsas, B.; Walker, E.; Nyati, S. BUB1 Inhibition Sensitizes TNBC Cell Lines to Chemotherapy and Radiotherapy. Biomolecules 2024, 14, 625. [Google Scholar] [CrossRef] [PubMed]
  165. Xie, J.; Luo, X.; Deng, X.; Tang, Y.; Tian, W.; Cheng, H.; Zhang, J.; Zou, Y.; Guo, Z.; Xie, X. Advances in artificial intelligence to predict cancer immunotherapy efficacy. Front. Immunol. 2022, 13, 1076883. [Google Scholar] [CrossRef] [PubMed]
  166. Boniolo, F.; Dorigatti, E.; Ohnmacht, A.J.; Saur, D.; Schubert, B.; Menden, M.P. Artificial intelligence in early drug discovery enabling precision medicine. Expert. Opin. Drug Discov. 2021, 16, 991–1007. [Google Scholar] [CrossRef]
  167. Garrone, O.; La Porta, C.A.M. Artificial Intelligence for Precision Oncology of Triple-Negative Breast Cancer: Learning from Melanoma. Cancers 2024, 16, 692. [Google Scholar] [CrossRef]
  168. Guo, J.; Hu, J.; Zheng, Y.; Zhao, S.; Ma, J. Artificial intelligence: Opportunities and challenges in the clinical applications of triple-negative breast cancer. Br. J. Cancer 2023, 128, 2141–2149. [Google Scholar] [CrossRef]
  169. Li, S.; Zhang, N.; Zhang, H.; Zhou, R.; Li, Z.; Yang, X.; Wu, W.; Li, H.; Luo, P.; Wang, Z.; et al. Artificial intelligence learning landscape of triple-negative breast cancer uncovers new opportunities for enhancing outcomes and immunotherapy responses. J. Big Data 2023, 10, 132. [Google Scholar] [CrossRef]
  170. Qureshi, R.; Irfan, M.; Gondal, T.M.; Khan, S.; Wu, J.; Hadi, M.U.; Heymach, J.; Le, X.; Yan, H.; Alam, T. AI in drug discovery and its clinical relevance. Heliyon 2023, 9, e17575. [Google Scholar] [CrossRef]
Figure 1. Immunological components of TNBC tumor microenvironment. This illustration sets forth the immunological components of the TNBC tumor microenvironment, which includes tumor-infiltrating lymphocytes (TILs), regulatory T cells (Tregs), tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), dendritic cells (DCs), natural killer (NK) cells, myeloid-derived suppressor cells (MDSCs), cancer-associated adipocytes (CAAs), and immune checkpoints.
Figure 1. Immunological components of TNBC tumor microenvironment. This illustration sets forth the immunological components of the TNBC tumor microenvironment, which includes tumor-infiltrating lymphocytes (TILs), regulatory T cells (Tregs), tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), dendritic cells (DCs), natural killer (NK) cells, myeloid-derived suppressor cells (MDSCs), cancer-associated adipocytes (CAAs), and immune checkpoints.
Cancers 16 03250 g001
Figure 2. Current clinical immunotherapy approaches in TNBC. Clinical immunotherapy approaches for TNBC have been diversified in recent years and this illustration summarizes those strategies which include cytokines, mAbs/ADCs, immune checkpoint inhibitors, vaccines, adoptive cell therapies, and oncolytic virus therapy.
Figure 2. Current clinical immunotherapy approaches in TNBC. Clinical immunotherapy approaches for TNBC have been diversified in recent years and this illustration summarizes those strategies which include cytokines, mAbs/ADCs, immune checkpoint inhibitors, vaccines, adoptive cell therapies, and oncolytic virus therapy.
Cancers 16 03250 g002
Figure 3. Mechanism of action of ICIs targeting PD-L1, PD-1, and CTLA-4. T-cell inactivation and the prevention of tumor cell death are caused by their binding to the corresponding ligands on the surface of cancer cells. Immune checkpoint inhibition promotes anticancer activity and promotes T-cell activation.
Figure 3. Mechanism of action of ICIs targeting PD-L1, PD-1, and CTLA-4. T-cell inactivation and the prevention of tumor cell death are caused by their binding to the corresponding ligands on the surface of cancer cells. Immune checkpoint inhibition promotes anticancer activity and promotes T-cell activation.
Cancers 16 03250 g003
Table 1. Updated list of FDA-approved drugs to treat TNBC (accessed information from https://www.fda.gov/ on 26 July 2024).
Table 1. Updated list of FDA-approved drugs to treat TNBC (accessed information from https://www.fda.gov/ on 26 July 2024).
Drug ClassAgents
Cytotoxic Chemotherapy Carboplatin, Docetaxel, Doxorubicin, Epirubicin, Ixabepilone, Liposomal doxorubicin, Nab-paclitaxel, Paclitaxel, Vinorelbine, Cisplatin
Immunotherapy Atezolizumab, Pembrolizumab
Antibody–Drug ConjugatesSacituzumab govitecan, Trastuzumab deruxtecan (TNBC with low/ultra-low HER2 expression)
PARP InhibitorsOlaparib, Talazoparib
Table 2. Combination of immunotherapy with other treatment modalities evaluated in clinical trials for TNBC (accessed information from https://www.clinicaltrials.gov/ on 17 June 2024).
Table 2. Combination of immunotherapy with other treatment modalities evaluated in clinical trials for TNBC (accessed information from https://www.clinicaltrials.gov/ on 17 June 2024).
TargetInterventionsClinical Status and IdentifierStatus
PARP and PD-1Drug: Niraparib
Biological: Pembrolizumab
Phase I/II
NCT02657889
Completed
PARP and PD-L1Drug: Avelumab Phase 1b
Drug: Talazoparib Phase 1b
Drug: Avelumab Phase 2
Drug: Talazoparib Phase 2
Phase Ib/II
NCT03330405
Completed
PD-1Biological: Pembrolizumab
Drug: Nab-paclitaxel
Drug: Paclitaxel
Drug: Gemcitabine
Drug: Carboplatin
Drug: Normal Saline Solution
Phase III
NCT02819518
Completed
PD-1Drug: Eribulin Mesylate
Drug: Pembrolizumab
Phase Ib/II
NCT02513472
Completed
PD-L1Drug: Atezolizumab (MPDL3280A)
Drug: Nab-Paclitaxel
Drug: Placebo
Phase III
NCT02425891
Completed
PD-L1Drug: Atezolizumab
Drug: Nab-paclitaxel
Phase Ib
NCT01633970
Completed
PD-L1Drug: Atezolizumab (MPDL3280A)
Drug: Atezolizumab Placebo
Drug: Paclitaxel
Phase III
NCT03125902
Completed
PD-1 Drug: Nivolumab
Radiation: Radiation therapy
Drug: Low-dose doxorubicin
Drug: Cyclophosphamide
Drug: Cisplatin
Phase II
NCT02499367
Ongoing
PD-1Biological: Pembrolizumab
Drug: Nab-paclitaxel
Drug: Anthracycline (doxorubicin)
Drug: Cyclophosphamide
Drug: Carboplatin
Drug: Paclitaxel
Phase I
NCT02622074
Completed
PD-1Biological: Pembrolizumab
Drug: Carboplatin
Drug: Paclitaxel
Drug: Doxorubicin
Drug: Epirubicin
Drug: Cyclophosphamide
Drug: Placebo
Biological: GM-CSF
Phase III
NCT03036488
Ongoing
PD-L1Drug: MEDI4736 (Durvalumab)
Drug: Placebo
Drug: Nab-Paclitaxel
Drug: Epirubicin
Drug: Cyclophosphamide
Phase II
NCT02685059
Completed
PD-L1Drug: Carboplatin
Drug: Abraxane
Drug: MPDL3280A (Atezolizumab)
Procedure: Surgery
Drug: Anthra
Phase III
NCT02620280
Ongoing
PD-L1Drug: Atezolizumab (MPDL3280A)
Drug: Placebo
Drug: Nab-paclitaxel
Drug: Doxorubicin
Drug: Cyclophosphamide
Drug: Filgrastim
Drug: Pegfilgrastim
Phase III
NCT03197935
Completed
PD-1 Drug: Pembrolizumab
Radiation: Radiotherapy
Phase II
NCT02730130
Completed
PD-L1 Radiation: SABR
Drug: Atezolizumab
Phase II
NCT03464942
Completed
PD-1 and LIV-1Drug: Ladiratuzumab vedotin
Drug: Pembrolizumab
Phase Ib/II
NCT03310957
Ongoing
PD-L1 and AKTDrug: Atezolizumab
Drug: Ipatasertib
Drug: Paclitaxel
Drug: Placebo for Atezolizumab
Drug: Placebo for Ipatasertib
Phase III
NCT04177108
Completed
PD-1, PARP, and VEGFR-2Drug: SHR-1210 + Apatinib + FluzoparibPhase I
NCT03945604
Completed
PD-1, VEGFR-2, c-KIT, and PDGFRbDrug: Camrelizumab + nab-paclitaxel + famitinibPhase II
NCT04129996
Completed
PD-L1 and CD73Drug: Paclitaxel
Drug: Carboplatin
Drug: MEDI4736
Drug: MEDI9447
Phase I/II
NCT03616886
Ongoing
PD-L1 and modified oncolytic herpes virusBiological: Talimogene Laherparepvec
Biological: Atezolizumab
Phase Ib
NCT03256344
Ongoing
PD-L1Avelumab, SBRT, haNK, and 15 other interventions/treatmentsPhase I/II
NCT03387085
Completed
Table 3. List of non-immune therapies under clinical trials for TNBC treatment (accessed information from https://www.clinicaltrials.gov/ on 17 June 2024).
Table 3. List of non-immune therapies under clinical trials for TNBC treatment (accessed information from https://www.clinicaltrials.gov/ on 17 June 2024).
TargetInterventionsClinical Status and IdentifierStatus
EGFRDrug: Metformin
Drug: Erlotinib
Phase I
NCT01650506
Completed
PI3KDrug: BKM120Phase II
NCT01790932
Completed
PI3KDrug: BKM120 and Olaparib
Drug: BYL719 and Olaparib
Phase I
NCT01623349
Completed
PI3KDrug: BYl719Phase II
NCT02506556
Completed
AKTDrug: Ipatasertib
Drug: Paclitaxel
Drug: Placebo
Phase II
NCT02301988
Completed
AKTDrug: Ipatasertib
Drug: Paclitaxel
Drug: Placebo
Phase II
NCT02162719
Completed
AKTDrug: Paclitaxel
Drug: AZD5363
Drug: Placebo
Phase II
NCT02423603
Unknown
AKTDrug: Capivasertib
Drug: Paclitaxel
Drug: Placebo
Phase III
NCT03997123
Ongoing
AKTDrug: Capivasertib
Other: Laboratory Biomarker Analysis
Drug: Olaparib
Other: Pharmacological Study
Drug: Vistusertib
Phase Ib
NCT02208375
Ongoing
AKTDrug: GSK1120212
Drug: GSK2141795
Phase I
NCT01138085
Completed
mTORDrug: Doxil
Drug: Bevacizumab
Drug: Temsirolimus
Phase I
NCT00761644
Completed
mTORDrug: EverolimusPhase II
NCT01931163
Completed
mTORDrug: Everolimus
Drug: Eribulin mesylate
Other: Pharmacological study
Other: Laboratory biomarker analysis
Phase I
NCT02120469
Completed
mTORDrug: Everolimus
Drug: Eribulin
Phase I
NCT02616848
Completed
CDK4/6Drug: Trilaciclib
Drug: Gemcitabine
Drug: Carboplatin
Phase 2
NCT02978716
Completed
CDK4/6Drug: Trilaciclib
Drug: Gemcitabine
Drug: Carboplatin
Phase 2
NCT02978716
Completed
ATRDrug: M6620
Drug: Gemcitabine
Drug: Cisplatin
Drug: Etoposide
Drug: Carboplatin
Drug: Irinotecan
Phase I
NCT02157792
Completed
ATRDrug: Olaparib
Drug: Ceralasertib
Drug: Adavosertib
Phase 2
NCT03330847
Ongoing
ATRProcedure: Biopsy
Drug: Capivasertib
Drug: Ceralasertib
Biological: Durvalumab
Drug: Olaparib
Other: Quality-of-Life Assessment
Drug: Selumetinib
Phase II
NCT03801369
Ongoing
CHK1Drug: LY2606368Phase II
NCT02203513
Completed
WEE1Drug: Cisplatin
Drug: AZD1775
Phase II
NCT03012477
Completed
MEKDrug: GSK1120212
Drug: GSK2141795
Phase I
NCT01138085
Completed
MEKDrug: Akt Inhibitor GSK2141795
Other: Laboratory Biomarker Analysis
Drug: Trametinib
Phase II
NCT01964924
Completed
MEKDrug: Ipatasertib
Drug: Cobimetinib
Phase I
NCT01562275
Completed
MET, VEGFR2, RET, AXL, FTL3, etc.Drug: CabozantinibPhase II
NCT01738438
Completed
VEGF, PDGFR, HGF, etc.Drug: Paclitaxel
Drug: Carboplatin
Drug: Sunitinib
Phase I/II
NCT00887575
Completed
VEGF, PDGFR, HGF, etc.Drug: SU011248
Drug: Chemotherapy
Phase II
NCT00246571
Completed
Aurora-A, VEGFR, FGFRDrug: ENMD-2076Phase II
NCT01639248
Completed
EGFR, HER2Drug: Veliparib + LapatinibPhase: N/A
NCT02158507
Ongoing
PI3K, mTORDrug: Prexasertib
Drug: Cisplatin
Drug: Cetuximab
Drug: G-CSF
Drug: Pemetrexed
Drug: Fluorouracil
Drug: LY3023414
Drug: Leucovorin
Phase I
NCT02124148
Completed
PARPDrug: PamiparibPhase I/II
NCT03333915
Completed
PARPDrug: TalazoparibPhase II
NCT03499353
Completed
PARPDrug: OlaparibPhase II
NCT02681562
Completed
PARPDrug: Olaparib
Radiation: Radiation therapy
Phase I
NCT03109080
Completed
PARPDrug: Iniparib
Drug: Gemcitabine
Drug: Carboplatin
Phase II
NCT01045304
Completed
PARPDrug: Cyclophosphamide
Drug: Placebo
Drug: Doxorubicin
Drug: Paclitaxel
Drug: Carboplatin
Drug: Veliparib
Drug: Placebo
Phase III
NCT02032277
Completed
HDACDrug: Chidamide + CisplatinPhase II
NCT04192903
Completed
HDACDrug: EntinostatPhase I
NCT03361800
Terminated
HDACDrug: Romidepsin
Drug: Cisplatin
Drug: Nivolumab
Phase I/II
NCT02393794
Ongoing
SMODrug: LDE225
Drug: Docetaxel
Phase I
NCT02027376
Completed
XPO1Drug: SelinexorPhase II
NCT02402764
Completed
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

Sriramulu, S.; Thoidingjam, S.; Speers, C.; Nyati, S. Present and Future of Immunotherapy for Triple-Negative Breast Cancer. Cancers 2024, 16, 3250. https://doi.org/10.3390/cancers16193250

AMA Style

Sriramulu S, Thoidingjam S, Speers C, Nyati S. Present and Future of Immunotherapy for Triple-Negative Breast Cancer. Cancers. 2024; 16(19):3250. https://doi.org/10.3390/cancers16193250

Chicago/Turabian Style

Sriramulu, Sushmitha, Shivani Thoidingjam, Corey Speers, and Shyam Nyati. 2024. "Present and Future of Immunotherapy for Triple-Negative Breast Cancer" Cancers 16, no. 19: 3250. https://doi.org/10.3390/cancers16193250

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