Skip to main content
Nature Portfolio logoLink to Nature Portfolio
. 2024 Nov 21;44(4):193–207. doi: 10.1038/s41388-024-03227-6

Combination strategies with PARP inhibitors in BRCA-mutated triple-negative breast cancer: overcoming resistance mechanisms

Aditi Jain 1,, Alan Barge 2,#, Christopher N Parris 3,✉,#
PMCID: PMC11746151  PMID: 39572842

Abstract

Triple-negative breast cancer (TNBC) is a particularly aggressive breast cancer subtype, characterised by a higher incidence in younger women, rapid metastasis, and a generally poor prognosis. Patients with TNBC and BRCA mutations face additional therapeutic challenges due to the cancer’s intrinsic resistance to conventional therapies. Poly (ADP-ribose) polymerase inhibitors (PARPis) have emerged as a promising targeted treatment for BRCA-mutated TNBC, exploiting vulnerabilities in the homologous recombination repair (HRR) pathway. However, despite initial success, the efficacy of PARPis is often compromised by the development of resistance mechanisms, including HRR restoration, stabilisation of replication forks, reduced PARP1 trapping, and drug efflux. This review explores latest breakthroughs in overcoming PARPi resistance through combination therapies. These strategies include the integration of PARPis with chemotherapy, immunotherapy, antibody-drug conjugates, and PI3K/AKT pathway inhibitors. These combinations aim to enhance the therapeutic efficacy of PARPis by targeting multiple cancer progression pathways. The review also discusses the evolving role of PARPis within the broader treatment paradigm for BRCA-mutated TNBC, emphasising the need for ongoing research and clinical trials to optimise combination strategies. By tackling the challenges associated with PARPi resistance and exploring novel combination therapies, this review sheds light on the future possibilities for improving outcomes for patients with BRCA-mutated TNBC.

Subject terms: Breast cancer, Targeted therapies

Introduction

Breast cancer (BC) remains one of the most prevalent forms of cancer globally, with an estimated 2.3 million new cases and ~685,000 deaths reported in 2020 alone [1]. Among the various subtypes of breast cancer, triple-negative breast cancer (TNBC) is notably aggressive and presents significant treatment challenges. Defined by the absence of oestrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptor 2 (HER2), TNBC represents about ~15% of all breast cancer cases [2]. This lack of receptor expression renders TNBC unresponsive to hormonal therapies effective in other subtypes, thereby complicating treatment and often necessitating reliance on conventional chemotherapy [3].

Research indicates that late detection of TNBC is frequently linked with dysregulated autophagy, tumour heterogeneity, genetic mutations, and alterations in drug targets, leading to suboptimal therapeutic outcomes [4, 5]. The stage of neoplasm significantly influences the treatment strategy, which may include surgery, radiation, conventional chemotherapy, targeted therapies, and immune therapeutics [6]. The current chemotherapeutic strategies often involve a cocktail of drugs that act through multiple mechanistic pathways to enhance tumour cell eradication [7]. This multi-agent approach aims to target different cellular pathways and cell cycle phases to reduce tumour cell survival effectively.

TNBC is characterised by the reduced expression of three key biomarker proteins: ER, HER2, and PR, affecting ~15–20% of breast cancer patients [8]. This subtype is associated with poorer clinical outcomes, higher rates of recurrence, and metastasis, particularly to the liver, lungs, and central nervous system. TNBC also disproportionately affects younger women and exhibits a higher aggressiveness in specific populations, such as African Americans, where it tends to present at an earlier age and in more advanced stages [9, 10]. In contrast, Asian and European populations show a lower incidence and relatively better survival rates compared to African American women [11].

The unfavourable prognosis of TNBC is primarily attributed to its genomic instability, often driven by mutations in the BRCA1 and BRCA2 genes, which play a critical role in repairing double-strand DNA breaks via the homologous recombination repair (HRR) pathway [12]. Mutations in these genes compromise the HRR mechanism, leading to increased genomic instability—a hallmark of cancer progression [13].

Traditionally, the treatment of TNBC involves surgery followed by adjuvant radiation and chemotherapy. However, the efficacy of chemotherapy is limited by its severe side effects, including cardiotoxicity, myelosuppression, and gastrointestinal issues, as well as the development of chemoresistance [14]. In light of these challenges, there is growing interest in developing targeted therapies that exploit the unique molecular defects of TNBC. One of the most promising advances in this field is the use of poly (ADP-ribose) polymerase inhibitors (PARPi), which target tumours with HRR deficiencies such as those caused by BRCA mutations [15].

PARP inhibitors function by blocking the repair of single-strand breaks in DNA, leading to the accumulation of double-strand breaks that are lethal to cells with defective HRR mechanisms, such as BRCA-mutant cells. This mechanism of action, known as synthetic lethality, has been a significant breakthrough in TNBC treatment [16]. Pioneering studies identified the synthetic lethal interactions between PARPis and BRCA1 or BRCA2 mutations. Currently, five PARPis, including olaparib, niraparib, rucaparib, talazoparib and veliparib, have been developed, each with varying capacities to trap PARP on DNA and inhibit its catalytic activity [17]. These inhibitors have demonstrated efficacy in treating BRCA-mutated TNBC by exacerbating DNA damage and inducing cancer cell death in tumours with defective BRCA genes [17].

Despite the promise shown by PARPis, their application in TNBC is limited by the disease’s heterogeneity and the potential for resistance development. Challenges in managing associated toxicities and the restriction of PARPi use to patients with confirmed BRCA mutations further complicate treatment. Current research efforts are focused on overcoming these limitations and exploring combination therapies that may enhance the efficacy of PARPis. This review will be exploring the basis for clinical use of PARPis, mechanisms of resistance and the current landscape of PARPi combination strategies in BRCA mutated TNBC treatment. Additionally, it will present a progressive analysis of the novel approaches and emerging therapies, emphasising the unique contributions of this work. By understanding these aspects, we can better navigate the future of TNBC therapy and explore potential combination strategies to enhance the effectiveness of PARPi and improve patient outcomes.

Rationale for clinical application of PARP inhibitors

PARP1 is an enzyme essential for repairing single-stranded DNA breaks (SSBs), which is a crucial part of the bigger DNA damage response (DDR) machinery [1820]. Detection of an SSB by PARP1, leads to their binding, consequently auto—PARylation of PARP1s catalytic domains which is the covalent binding of PARP enzymes to poly (ADP-ribose) [PAR] chains. The latter aids in DNA damage repair as auto-PARylation leads to the formation of PARP complexes which further induce chromatic remodelling by recruiting SSB proteins [2, 21]. However, in case of disruption or inhibition of auto-PARylation, SSBs accumulate, triggering replication-dependent DNA DSBs and “PARP trapping”, where PARP1 is unable to detach from sites of DNA damage [22, 23]. PARPis induce PARP trapping which directs the collapse of the replication fork collapse and an increase in DSBs, resulting in clinically significant cytotoxicity [24, 25]. Further, cells repair DNA integrity at double-DSBs through two main pathways: homologous recombination (HR) and the error-prone nonhomologous end joining (NHEJ) pathway [26, 27]. HR, a high-fidelity repair process that relies on BRCA1 and BRCA2 proteins, is primarily active during the S and G2 phases of the cell cycle. In contrast, NHEJ predominantly operates in the G1 phase [2]. Both healthy and cancer cells utilise HR and NHEJ, but BRCA1/2-mutant cancer cells have impaired HR capabilities, which forces them to rely on NHEJ. This reliance can lead to genomic instability due to inaccurate repairs [28].

PARPis take advantage of this vulnerability through a mechanism known as synthetic lethality. By inhibiting PARP1, these drugs trap PARP1 at SSB sites, leading to the accumulation of DSBs and replication stress that BRCA-mutant cells cannot repair [26]. This targeted cytotoxicity is especially effective in BRCA-mutant tumours, where defective DSB repair mechanisms necessitate reliance on PARP-induced SSB repair pathways [27]. Unlike wild-type cells, BRCA-mutant cells cannot utilise HR for repair, making them more sensitive to PARPi and supporting the clinical rationale for using PARPi in these specific tumour subsets [2, 28].

PARP inhibitors resistance mechanisms

PARPis, such as olaparib and talazoparib, have shown significant improvements in progression-free survival (PFS) among patients with germline BRCA1/2-mutated metastatic breast cancer (BC), as evidenced by the OlympiAD and EMBRACA Phase III trials. However, these trials did not demonstrate a significant benefit in overall survival (OS), highlighting a critical challenge in the clinical application of PARPis [12]. Similar to other targeted therapies, resistance to PARPis can emerge, posing a significant barrier to their widespread use in cancer treatment. Notably, more than 40% of BRCA-mutated cancer patients have shown limited benefit from PARPi therapies [29]. Understanding the mechanisms behind PARPi resistance (Fig. 1) is therefore crucial to enhancing the efficacy and durability of these therapies in clinical practice [30].

Fig. 1. Mechanism of resistance of PARPis.

Fig. 1

(1) Gene reversion mutations can restore HRR genes like BRCA1/2 and RAD51C/D, causing drug resistance by restoring HRR function. Inactivating NHEJ proteins (e.g., 53BP1) enhances HRR efficiency during PARPi treatment, promoting DNA repair and stability. Factors like Shieldin complex, RIF1 and REV7 support. (2) BRCA mutations lowering EZH2 and PTIP reduce nuclease recruitment, leading to stable replication forks and consequently drug resistance. Stable replication forks mitigate DNA damage during replication stress induced by PARPis. Reduced chromatin remodelers (HLTF, ZRANB3, and SMARCAL1), which facilitate nuclease-dependent degradation of replication forks, contribute to this resistance lowering replication stress. SLFN11 normally regulates replication fork arrest, and its absence decreases PARPi cytotoxicity, further leading to drug resistance. (3) Changes or reduction in PARP can reduce PARP‐1 trapping on DNA. This lowered PARP trapping facilitates replication fork stability and further contributes to DNA repair signaling, which promotes PARPi resistance. (4) Long‐term PARPi treatment enhances P‐glycoprotein (P-gp) expression, resulting in lower intracellular PARPi concentration and PARP resistance. Liu et al. [12, 21, 204]. Flowchart created by Diagramly.

Restoration of homologous recombination repair (HRR)

The most common mechanism of resistance to PARPi therapy is the restoration of HRR function (Fig. 1.1). Gene reversion mutations can reactivate HRR genes such as BRCA1 and BRCA2, which were originally impaired [29, 31]. This reactivation allows cancer cells to regain the ability to effectively repair double-strand DNA breaks, thereby evading the synthetic lethal effects of PARP inhibition. Studies have documented the occurrence of reversion mutations in BRCA1/2 among breast cancer patients, leading to the development of resistance to PARPi therapy [32, 33]. While the restoration of HRR is a well-documented mechanism, it is important to note that it does not account for all instances of resistance. Some patients with persistent BRCA mutations also develop resistance through alternative pathways.

Stable replication forks

Another mechanism by which BRCA1/2-mutant cells develop resistance to PARPis involves the stabilisation of replication forks [12] (Fig. 1.2). Under normal conditions, BRCA1/2 proteins protect stalled replication forks during DNA damage repair [21, 34]. In the absence of functional BRCA1/2, other factors such as EZH2 and PTIP facilitate the recruitment of nucleases like MRE11 and MUS81 to the stalled replication forks, leading to fork collapse and subsequent cell death [35]. However, when the activities of EZH2 and PTIP are reduced, nuclease recruitment is inhibited, resulting in the stabilisation of replication forks. This process protects the genomic integrity of the cancer cells, thereby generating resistance to PARPis [34, 35]. While the stabilisation of replication forks is a significant contributor to resistance, it is not the sole mechanism, as some BRCA1/2-mutant cells can still succumb to PARPi treatment despite fork stabilisation.

Reduced PARP1 trapping

Some PARPis (talazoparib, saruparib) exert their pharmacologic effects in part, by ‘trapping’ -PARP1 enzyme onto single-stranded DNA, thereby compromising the DNA repair process [36, 37] (Fig. 1.3). Alterations or depletion of PARP1, or the loss of poly (ADP-ribose) glycohydrolase (PARG), can reduce the trapping of PARP1 on the DNA strand complex [36]. In particular, the loss of PARG results in the accumulation of poly (ADP-ribose) (PAR), which in turn restores PARP1-dependent DNA damage signalling and diminishes PARP1 trapping. This reduction in PARP1 trapping decreases the extent of DNA damage induced by PARPis, rendering cancer cells less sensitive to the treatment [37]. ‘PARP-trappers’ such as talazoparib and saruparib exhibit a longer residency time in the NADH-binding domain of PARP-1. They do not change the binding of PARP-1 to single-stranded DNA. Evidence for this mechanism is seen in PARP1 knockout cells, which exhibit a high level of resistance to olaparib in BRCA mutated patient [24] however it was ovarian cancer [36] and not TNBC thus further research is needed. Nonetheless, the reduced trapping of PARP1 alone does not fully explain the resistance observed in all cases, suggesting the involvement of other contributing factors.

Drug efflux

The overexpression of ATP-binding cassette drug transport proteins is another mechanism associated with PARPi resistance [38] (Fig. 1.4). Long-term treatment with PARPis has been shown to upregulate the expression of P-glycoprotein (P-gp, also known as MDR1, encoded by ABCB1), which acts as a drug efflux pump [39]. This upregulation leads to a reduction in the intracellular concentration of PARPis, thereby diminishing their therapeutic efficacy and leading to resistance. Although the precise mechanisms underlying the increased expression of the P-gp efflux pump in relation to PARPi resistance are not fully understood, the restoration of PARPi sensitivity in tumours upon co-treatment with tariquidar, a P-gp inhibitor, supports the role of drug efflux in resistance [40]. However, it remains unclear whether P-gp overexpression alone is sufficient to confer complete resistance or if it acts in concert with other resistance mechanisms.

Overall, the development of resistance to PARPis in BRCA-mutated breast cancer is a complex phenomenon involving various mechanisms, such as the restoration of HRR, stabilisation of replication forks, reduced PARP1 trapping, and drug efflux [30]. Each of these mechanisms presents unique challenges and opportunities for improving the effectiveness of PARPi therapies. By elucidating the molecular underpinnings of PARPi resistance, researchers and clinicians can develop more robust strategies to overcome resistance and enhance clinical outcomes for patients with BRCA-mutated cancers. To address this resistance, combination strategies will be discussed further as a promising approach.

Advances in combination strategies with PARP inhibitors

PARP inhibitors and chemotherapy

Chemotherapy has long been the cornerstone of treatment for triple-negative breast cancer. Recently, the integration of PARPis into treatment regimens has shown promise, particularly in patients with BRCA1/2 mutations, due to their impaired DNA repair mechanisms via HR [15]. PARP inhibitors, when combined with chemotherapy, aim to exploit this vulnerability by further preventing the repair of DNA damage, thus driving tumour cells towards apoptosis. Clinical studies (Table 1) suggest that this combination can enhance treatment efficacy, leading to better clinical outcomes compared to chemotherapy alone. Specifically, PARP inhibitors such as veliparib, olaparib, and Niraparib have been studied in combination with traditional chemotherapeutic agents, including platinum-based compounds, with varying degrees of success.

Table 1.

Summary of clinical trials investigating PARP inhibitors in combination with chemotherapy (Adapted from Kunwor et al., [46]).

Trial Experimental Group Control Group Median PFS (months) Median OS (months)
BROCADE [41, 43] Veliparib with Carboplatin/Paclitaxel Placebo plus carboplatin/paclitaxel 14.1 vs 12.3 28.3 vs 25.9
Veliparib with temozolomide Placebo plus carboplatin/paclitaxel 7.4 vs 12.3 19.1 vs 25.9
BROCADE-3 [45] Veliparib with Carboplatin/Paclitaxel Placebo plus carboplatin/paclitaxel 14.5 vs 12.6 33.5 vs 28.2
SWOG S1416 [47] Veliparib with Cisplatin Placebo with Cisplatin 6.2 vs 6.4 14.2 vs 14.6
BRAVO Niraparib Physician’s choice CT Study was prematurely closed after an interim analysis showed too many patients were not completing the necessary assessments in the control arm, and it was no longer suitable as a registration trial.

In the BROCADE series of trials, veliparib was combined with platinum-based chemotherapy. The phase II BROCADE trial [4144] revealed a numerical but not significant difference in PFS and overall survival (OS) between the veliparib and control arms. However, there was a significant improvement in the overall response rate (ORR) (77.8% vs 61.3%). The BROCADE 3 trial, a phase III study, demonstrated a more consistent PFS benefit in the veliparib group compared to standard chemotherapy (14.5 months vs 12.6 months; HR 0.71; 95% CI 0.57–0.88; P = 0.0016) [45]. This study also allowed for the continuation of veliparib monotherapy post-chemotherapy, leading to a persistent PFS benefit, particularly notable in the delayed separation of Kaplan-Meier curves and a prolonged tail in the veliparib group [46].

Conversely, the SWOG S1416 trial, which evaluated veliparib in combination with cisplatin in TNBC patients, including those with BRCA mutations, did not show a significant PFS or OS benefit in the BRCA-mutated subgroup [47]. This trial’s limited accrual [15], likely influenced by the concurrent FDA approvals of olaparib and talazoparib for BRCA-mutated TNB, reduced its statistical power. However, the trial did indicate potential benefits in BRCA-like mutation groups, suggesting that the effectiveness of PARP inhibitors may extend beyond just BRCA-mutated cancers.

Despite the encouraging results, the combination of PARP inhibitors with chemotherapy is not without challenges. The addition of PARP inhibitors often exacerbates the toxicities associated with chemotherapy. For instance, in phase I trials where olaparib was combined with carboplatin and paclitaxel, there was a marked increase in the frequency, severity, and duration of adverse events, leading to dose reductions of olaparib [48, 49]. Significant attempts to modulate the myelosuppression produced by the combination of Olaparib with carboplatin were made, include optimal prophylaxis with filgrastim (r-metHuG-CSF). Despite this, it proved impossible to safely combine Olaparib, or any of the first-generation PARP-inhibitors with myelosuppressive chemotherapy. Veliparib, considered a weaker PARP trapping agent, has shown a slightly better safety profile with less myelosuppression [16], as seen in the BROCADE 3 trial, where common grade 3 or worse adverse events included neutropenia (81% vs 84%), anaemia (42% vs 40%), and thrombocytopenia (40% vs 28%) [45]. This probably reflects its lower potency relative to Olaparib, niraparib and talazoparib.

Further complicating the treatment landscape is the inconsistency in clinical trial outcomes. For instance, the phase II trial by O’Shaughnessy et al. [50], which added iniparib to carboplatin and gemcitabine, initially showed enhanced clinical benefits, including prolonged PFS and OS. However, these results were not replicated in the phase III trial, which failed to meet its primary endpoints, likely due to patient variability, including differing metastatic burdens.

Other studies exploring different chemotherapy agents in combination with PARP inhibitors, such as cyclophosphamide or doxorubicin, have also produced mixed results. In some cases, while the antitumor effectiveness was promising, significant adverse effects, including secondary cancers like oral squamous cell carcinoma, emerged, raising concerns about the long-term safety of these combinations [51].

The mechanism of myelosuppression exhibited by 1st-generation PARP inhibitors, either as monotherapy, or especially when used in combination with chemotherapy, has been the subject of significant research. First-generation compounds are equipotent against both PARP-1 and PARP-2. Pre-clinical studies demonstrate a significant role of PARP-2 in erythroid and myeloid progenitor maturation [52]. As a consequence, a number of second-generation PARP-1 selective inhibitors are in development, such as AZD5305 (saruparib). Saruparib has a 600-fold selectivity for PARP-1 relative to PARP-2. Early clinical data suggest that this approach only partially reduces the degree myelosuppression, suggesting that the underlying mechanism may also be partially dependent on PARP-1 [53].

The controversy over the optimal chemotherapy-PARP inhibitor combination and sequencing continues [54]. Notably, the GeparOcto trial found no significant difference in pathological complete response (pCR) rates between two intensive chemotherapy regimens, regardless of the addition of carboplatin [55]. Similarly, the BrighTNess trial showed that while the addition of veliparib to paclitaxel and carboplatin improved pCR rates compared to paclitaxel alone, it did not outperform the paclitaxel-carboplatin combination without veliparib [56]. These findings suggest that while PARP inhibitors may enhance chemotherapy’s effectiveness in some contexts, their universal application in neoadjuvant settings remains unproven.

Moreover, the phenomenon of BRCA1/2 reversion mutations and other mechanisms of resistance to PARP inhibitors complicates treatment strategies, underscoring the need for further research [57]. Large, randomised trials are essential to determine the most effective combinations and sequences, balancing efficacy with manageable toxicity profiles [58]. The ongoing debate about whether the benefits of adding PARP inhibitors to chemotherapy outweigh the risks of increased toxicity highlights the need for personalised treatment approaches, particularly in the diverse and complex landscape of TNBC.

Thus, while combining PARP inhibitors with chemotherapy represents a promising strategy for improving outcomes in BRCA-mutated TNBC, particularly in advanced cases, the approach is fraught with challenges. These include managing severe toxicities, overcoming inconsistent clinical trial results, and addressing emerging resistance mechanisms. Continued research into optimising treatment regimens and understanding patient-specific responses will be crucial in advancing this therapeutic strategy.

PARP inhibitors and immunotherapy

The tumour microenvironment (TME) of BC, particularly in TNBC, is characterised by decreased expression of tumour-infiltrating lymphocytes (TILs) and increased presence of immunosuppressive cells, such as regulatory T cells (Tregs) and tumour-associated macrophages (TAMs) [5961]. This immunosuppressive environment contributes to the negative effects on chemotherapy and targeted therapies. Studies have shown that the immune response initiated by PARPis is necessary for the elimination of breast tumours, particularly those with BRCA mutations [62]. PARPis increase cellular DNA damage in BRCA-mutant tumours and activate the type I interferon (IFN) pathway via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signalling pathway, which promotes dendritic cell (DC) maturation, release of interferons, and initiation of antitumor T-cell immune responses [63].

Preclinical investigations have highlighted a compelling synergy between PARP inhibitors (PARPis) and immunotherapeutic approaches, which can be attributed to two main mechanisms. First, PARPis induce extensive DNA damage, leading to the accumulation of cytosolic DNA, which activates immune pathways. Second, these inhibitors modulate T-cell responses and enhance the expression of programmed death ligand 1 (PD-L1) [62, 6466]. These changes within the TME create favourable conditions for integrating immune checkpoint blockade with PARP inhibition, particularly in tumours characterised by HR deficiency [6770]. Furthermore, the DNA damage caused by the collapse of the replication fork and increased DSBs as a result of PARP1 inhibition can be detected by the immune system particularly through the cyclic GMP-AMP synthase (cGAS) pathway [71]. Activation of this pathway subsequently enhances T-cell infiltration and promotes type I interferon expression, stimulating dendritic cell activity and reinforcing the immune response [25, 72]. Studies indicate that both talazoparib and olaparib can activate the cGAS-STING pathway, promoting T-cell infiltration in BRCA1-deficient models [30, 69, 73]. Additionally, research has shown that PARPis can increase PD-L1 expression by inactivating GSK3β, further supporting the rationale for combining these agents with immune checkpoint blockers (ICBs) [74] (Table 2). Preclinical studies have also shown promising results when anti-PD-1/L1 therapies are combined with PARPis such as olaparib and talazoparib, indicating their potential in effective combination strategies [65]. This is further supported by the phase II MEDIOLA trial which assessed the combination therapy of olaparib with durvalumab (an anti-PD-L1 antibody) in BRCA-mutant metastatic HER2-negative BC patients. The study reported a disease control rate (DCR) of 80% at 12 weeks and 50% at 28 weeks [75]. Common adverse events included anaemia (12%) and neutropenia (9%), with no significant drug interactions noted [76]. Further phase III trials are needed to validate the efficacy and safety, especially considering the higher toxicity of talazoparib compared to olaparib [77]. Moreover, results from the TOPACIO trial (phase II study) evaluating the efficacy of combination therapy, niraparib with pembrolizumab in metastatic TNBC showed overall response rates (ORR) of 45% and disease control rates (DCR) of 73% (experimental group), compared to 25% and 68% in the overall cohort. BRCA1/2 wild-type patients also benefited, with an ORR of 24%, suggesting therapeutic effects beyond specific mutations [67, 78]. Staniszewska et al. [63] further demonstrated that the combination of olaparib with ICBs leads to enhanced antitumor efficacy and immune modulation in BRCA-deficient tumours.

Table 2.

Summary of clinical trials investigating PARP Inhibitors in combination with immunotherapy.

Trial Treatment Combination Median PFS (months) ORRa (%)
TOPACIO Niraparib + Pembrolizumab 2.3 21%
MEDIOLA Olaparib + durvalumab 8.2 63%
DORA Maintenance olaparib + durvalumab v/s Olaparib alone 4 v/s 6.1 NA

aData related to the OS unavailable.

Overall, recent clinical findings highlight the importance of PARPi and ICB (anti-PD-L1 and anti-CTLA-4) combination therapies in treating BRCA-mutated TNBC due to their ability to affect T-cell activation and promote apoptosis in cancer cells, consequently showing significant clinical benefits [7981].

Despite the promise shown by PARPis and ICB combinations, there are challenges to be addressed, including resistance mechanisms and optimising combination strategies [30, 82]. The heterogeneity of BC subtypes also complicates the effectiveness of these therapies. Immune-related adverse events are a significant concern, with common toxicities including anaemia, neutropenia, and pancreatitis. The clinical toxicity of talazoparib is notably greater than that of olaparib, raising concerns about its combination with ICBs [67, 78]. Further clinical trials are necessary to validate the efficacy and safety of PARPis in combination with ICIs across different BC subtypes and patient populations.

Thus, the synergy between PARPis and ICIs offers a promising avenue for improving treatment outcomes for patients with BRCA-mutated BC. By exploring combination strategies and addressing resistance mechanisms, researchers aim to enhance the clinical efficacy of these therapies and improve patient outcomes in BC.

PARP inhibitors and antibody–drug conjugates

Antibody-drug conjugates (ADCs) and PARP Inhibitors represent a novel treatment approach, enabling the targeted delivery of potent cytotoxic agents directly to cancer cells [83]. ADCs are composed of three main components: a monoclonal antibody (mAb) that binds specifically to a tumour antigen, a cytotoxic agent that induces cell death, and a linker that connects the two. This structure enhances tumour specificity, improving the therapeutic index and reducing off-target toxicity [8486]. Over recent years, ADCs have entered clinical studies across multiple cancer types.

One promising ADC is sacituzumab govitecan (Trodelvy), which combines SN-38, an active metabolite of the topoisomerase I inhibitor irinotecan, with a humanised monoclonal antibody (hRS7 IgG1κ) targeting the Trop-2 antigen. Trop-2 is overexpressed in many epithelial tumours, including breast cancer subtypes, with particularly high levels in TNBC [87, 88]. Clinical trials, such as the IMMU-132-01 phase I/II and the ASCENT phase III trials, have demonstrated durable responses in metastatic TNBC patients, making sacituzumab govitecan a strong candidate for combination therapies, including with PARP inhibitors [89]. Ongoing studies, such as the SEASTAR trial (NCT03992131), have further explored this combination, evaluating rucaparib (a PARP inhibitor) in conjunction with sacituzumab govitecan [53]. Despite promising early results, the SEASTAR trial was discontinued due to changes in development priorities. However, data from the trial indicated that further investigation into PARP inhibitor and ADC combinations is warranted, particularly to optimise dosing and reduce myelosuppression [89].

Another ADC of interest is datopotamab deruxtecan (Dato-DXd), which consists of a humanised anti-Trop-2 IgG1 monoclonal antibody linked to a potent topoisomerase I inhibitor via a stable tetrapeptide-based linker [87]. Dato-DXd offers advantages over sacituzumab govitecan due to its more stable structure, longer half-life, and lower haematologic toxicity. These properties make it a compelling candidate for combination with PARP inhibitors, as both agents induce DNA damage and interfere with DNA repair mechanisms [87]. The ongoing phase 1/2 PETRA trial, AZD5305 (a PARP1 inhibitor) has demonstrated safety and activity in a preliminary analysis of 40 patients, including those with BRCA mutated TNBC, with an ORR of 25% and a disease control rate of 53%. This agent is being investigated for its efficacy either as monotherapy or in combination with Dato-DXd in patients with advanced solid tumours [53]. While these early results are promising, more clinical trials are necessary to confirm the efficacy and safety of AZD5305 in BRCA-mutated TNBC patients.

Thus, early clinical data support the potential of combining ADCs with PARP inhibitors in treating cancers such as TNBC, further research is needed. Future studies should focus on optimising dosing strategies, minimising toxicity, and clarifying the contributions of each agent to the observed antitumor effects. These efforts are essential for advancing the use of these novel therapeutic combinations in clinical practice.

PARP inhibitors and PI3K/AKT pathway inhibitors

PARPis with inhibitors of the PI3K/AKT pathway, which plays a key role in regulating cell growth, survival, and metabolism, as well as acting as a sensor for DNA double-strand breaks (DSBs) within the DDR machinery [90]. Preclinical studies have demonstrated that silencing PIK3CA in BRCA wild-type, PTEN-mutant TNBC leads to an accumulation of phosphorylated histone-2AX (γ-H2AX), a marker of DNA damage and replication stress. The elevated γ-H2AX levels suggest that PIK3CA loss may enhance the effectiveness of PARPis, creating a potential synergistic relationship between these inhibitors [91, 92].

Further research supports this concept, showing that PI3K inhibition impairs BRCA1/2 expression and sensitises BRCA1/2-proficient TNBC to PARPis [93]. Additionally, studies have indicated that decreased mTOR activity correlates with a favourable response in TNBC, while high tyrosine kinase receptor activity and mTOR activation are associated with resistance [94]. These findings underscore the importance of refining biomarkers of treatment response to identify specific patient populations that may benefit most from such combination therapies.

However it has been primarily evaluated in BRCA mutated ovarian cancer and despite the promising results seen in ovarian cancer, the application of PARPi combinations in TNBC remains underexplored and requires further investigation [30]. Given the distinct molecular heterogeneity of TNBC, it is imperative to determine whether these strategies will yield similar therapeutic benefits in this challenging breast cancer subtype. More research is necessary to validate these approaches and optimise treatment protocols for TNBC patients facing PARPi resistance.

Integrating PARP inhibitors in the treatment paradigm for BRCA mutated TNBC patients

As the therapeutic landscape of breast cancer broadens, novel questions arise about the optimal use of PARP inhibitors. First, genetic testing is not universally offered to patients with breast cancer despite its potential therapeutic implications [95]. Second olaparib’s integration with available (neo) adjuvant therapies, such as pembrolizumab for TNBC is debated. KEYNOTE-522 trial, in which pembrolizumab was added to neoadjuvant chemotherapy and administered adjuvantly, event-free survival was prolonged, precluding adjuvant olaparib [96]. However, in the OlympiAD trial the patients did not receive pembrolizumab only Olaparib and yet significantly increased PFS and doubled ORR. Thus, whether patients with residual disease after neoadjuvant pembrolizumab-chemotherapy should receive olaparib, pembrolizumab, or both is unknown [97]. As patients with germline BRCA1 or BRCA2 variants are excluded from most of the ongoing trials in this setting, it is unlikely that randomised data will soon clarify which is the best approach for the patients [98]. Further, when patients with BRCA1/2-associated metastatic breast cancer should receive PARP inhibitors is also debated. For patients with TNBC and BRCA1/2, to our knowledge, no existing evidence suggests how PARPis should be prioritised to newer options of chemoimmunotherapy or ADCs (eg, sacituzumab govitecan and trastuzumab deruxtecan). Informative subgroup analyses by BRCA1/2 alterations from phase 3 trials have not been published. As in OlympiAD, olaparib had the highest benefit when administered as first-line treatment but did not improve survival compared with chemotherapy. It may be reasonable to optimise first-line immunotherapy-based regimens and favour second line olaparib while acknowledging the lack of data to support any sequential strategy in this setting. Furthermore, whether and to what extent the activities PARPis and platinum chemotherapy overlap is debated [98]. Their mechanism of action is similar, as are their sensitivity and resistance spectra. However, head-to-head comparison between platinum chemotherapy and PARP inhibitors are lacking, and little is known about the activity of PARPis after platinum chemotherapy, and vice versa. Results from BROCADE suggested that PARPis may improve outcomes when added to carboplatin [99] but opposite evidence emerged from BrighTNess [100]. Data from Gepa Rola, though not comparative in its design, suggested that olaparib and carboplatin may be equivalent in the early-stage setting [101]. Future studies combining platinum salts with less toxic PARP1 selective inhibitors may help to clarify whether these agents have synergic or redundant activity. If shown to be equivalent, PARP inhibitors may be privileged to platinum chemotherapy given the better safety profile; however, the different cost-effectiveness profiles should also be considered. Finally, the role of maintenance PARP inhibitors after chemotherapy should be clarified. The aforementioned data from the BROCADE3 [99] and DORA trial [102] support this strategy, and further efforts are ongoing (KEYLYNK-009 trial, NCT04191135).

Future directions in research and clinical practice

As the treatment landscape for BRCA-mutated TNBC evolves, several promising directions (Fig. 2) in research and clinical practice are being explored to address the challenges of this aggressive subtype.

Fig. 2. Potential future research directions in the treatment of BRCA-mutated TNBC.

Fig. 2

Emerging treatment strategies for BRCA mutated TNBC focus on leveraging innovative approaches such as targeted inhibitors, personalized vaccines, and cell-based therapies to enhance patient outcomes. This roadmap illustrates promising areas of future research, aimed at driving advances and overcoming current therapeutic challenges for BRCA-mutated TNBC.

New-generation PARPis (Fig. 2.1) are being developed to address the limitations of current PARPis, such as resistance and off-target toxic effects, which have hindered their broader use in combination therapies. Among these novel agents, AZD5305 has shown promising pharmacokinetics, pharmacodynamics, and safety profiles in preclinical models. This agent exhibits higher selectivity for PARP1 over PARP2, leading to more stable PARP1 trapping and a strong antiproliferative effect. As mentioned above, AZD5305 demonstrated safety and activity in a preliminary analysis of the PETRA trial. While these early results are promising, more clinical trials are necessary to confirm the efficacy and safety of AZD5305 in BRCA mutated TNBC patients. Moreover, a newer, third generation series of compounds are in development [103], which are termed ‘DNA-locking’ PARPis. Unlike the first and second-generation compounds, such as Olaparib, talazoparib and saruparib, which are competitive NAD-mimetic compounds which can only work via synthetic-lethality, these compounds are allosteric modulators of the DNA-binding domain of PARP-1. As such, they are active via synthetic lethality but critically also by inhibiting DNA-replication in HR-competent tumours. Preclinical data show that they are active in both BRCA reversion-mutant and BRCA wild-type tumours.

In addition to PARP1-selective inhibitors, other novel therapeutic agents are also under development. These include dual inhibitors targeting PARP1 and intracellular molecules (such as histone deacetylases, DNA topoisomerases, and PI3K pathway effectors), PARP1 degraders using proteolysis-targeting chimera (PROTAC) technology, and PARP1-targeting prodrugs. These advancements aim to enhance the therapeutic potential of PARPis and expand their applicability across a broader patient population.

Furthermore, early clinical trials are investigating rational combination strategies (Fig. 2.2) to overcome PARPi resistance. These strategies include combining PARPis with ATR/Chk1/Wee1 inhibitors, antiangiogenic agents, and epigenetic modifiers (Table 3). These have been evaluated in ovarian cancer; however, they have the potential to be implemented in BRCA mutated TNBC. Further preclinical and clinical trials are needed to support this. Although initial analyses have shown promising responses and clinical benefits, the durability of these responses remains uncertain, and combination therapies often introduce additional toxicity. To optimise these approaches, further research is needed to identify meaningful molecular markers that can predict which patient groups will benefit most from specific combinations. Additionally, utilising circulating tumour DNA (ctDNA) analyses to monitor for BRCA and other HRR reversion mutations could help tailor treatment choices and optimise treatment duration. Investigating dosing schedules is also critical to minimising toxicity while maintaining therapeutic benefits beyond those expected from monotherapy.

Table 3.

Emerging combination therapies involving PARP inhibitors for BRCA-mutated TNBC (Adapted from [12, 30]).

Combination Therapy Pathway of Action Clinical Evidence
Combining PARPi + ATR, CHK1, and Wee1 Inhibitors These combinations target the downstream effector molecules and signalling pathways of HR, showing clinical benefits irrespective of HR status. Cells with DNA damage rely on cell cycle checkpoints to activate DNA DDR mechanisms. Tumours with p53 mutations often depend on G2 checkpoints to induce DDR. Inhibiting ATR, CHK1, and Wee1 has been shown to restore HR and replication fork stability, resensitizing BRCA1/2-deficient tumours to PARP inhibitors (PARPis). Lee, Matulonis [127, 128].

1. Synergistic DDR and cell cycle regulation have resulted in tumour suppression in BRCA2-mutant patient-derived xenograft models.

2. Ongoing clinical trials:

• CAPRI phase II study, Phase II trial with berzosertib and olaparib (NCT04065269),

• Phase Ib trial with ATRi and niraparib (NCT04267939).

• A phase II EFFORT trial demonstrated clinical benefit in PARPi-resistant ovarian cancer (NCT03579316). ORR for single agent adavosertib versus combination was 23% and 29%, respectively, with clinical benefit rates of 63% and 89%.

• CHk1 inhibitor Prexasertib combined with olaparib in Ovarian cancer and other solid tumours showed partial response in 4 out of 18 BRCA1 mutant, PARPi-resistant ovarian cancer patients [129].

Combining PARPi + Antiangiogenic Agents This combination leverages the synergistic effects of PARPis and antiangiogenic agents through two mechanisms: inhibition of BRCA1/2 expression and exacerbation of cellular hypoxia, leading to genetic instability. Hypoxia impairs HR and increases sensitivity to PARPis [130132]. For example, in BRCA2-mutated ovarian cancer stem-like cells, chemosensitivity was restored through VEGFR3 inhibition [133].

1. The randomised phase II ENGOT-ov24 trial (NCT02354131) showed clinical benefit with bevacizumab and niraparib in platinum-sensitive recurrent ovarian cancer.

2. A phase II study of olaparib and cediranib versus niraparib alone demonstrated significant progression-free survival (PFS) and overall survival (OS) benefits in BRCA1/2 wild-type cohorts, though a subsequent phase III trial found no clinical benefit compared to chemotherapy [134].

Combining PARPi + Epigenetic Modifiers (BET and Histone Deacetylase Inhibitors) Epigenetic modifiers involved in transcriptional regulation or chromatin remodelling can enhance PARP-related DNA damage, decrease BRCA1 levels, and increase genomic instability by disrupting replication fork progression. Preclinical data suggest that inhibiting bromodomains and histone deacetylases (HDACs) can enhance the activity of PARPis [135137].

1. Ongoing trials:

• Phase I/II study of entinostat and olaparib for recurrent ovarian cancer (NCT03924245)

• Belinostat with talazoparib for multiple solid tumours (NCT04703920).

3. Initial dose-finding studies in hematologic malignancies showed that BET inhibition correlates with thrombocytopenia [138140].

Combining PARPi + Ionising Radiation Therapy Ionising radiation sensitises PARPi in HR-proficient tumours by exporting BRCA1 from the nucleus to the cytoplasm [141, 142]. Preclinical studies show reversal of acquired drug resistance through BRCA1-independent HR restoration [143].

1. Phase I trials (NCT01589419, NCT01264432) report good tolerance and responses in advanced rectal cancer and peritoneal carcinomatosis [144, 145].

2. Ongoing trial (NCT01618357) evaluates veliparib with IR in breast cancer [146].

Combining PARPi + Proteogenomics

Proteomics, including mass spectrometry and protein arrays, identify new drug targets in BRCA mutated TNBC, improving therapy and reducing PARPi resistance [147].

TPX2, a PARP1-binding protein, enhances PARPi sensitivity by increasing PARP1 activity [148].

Tests like Oncotype DX, EndoPredict, and MammaPrint predict breast cancer recurrence risk, guiding clinical decisions [149, 150].

FoundationOne Liquid CDx trial - cfDNA-based assay predicts the efficacy of PARPi rucaparib in BRCA-mutated ovarian cancer and alpelisib in HR+/HER2−, PIK3CA-mutated breast cancer [151].

-

Moreover, cancer vaccines are an emerging immunotherapeutic strategy in the treatment of TNBC. These vaccines work by stimulating the immune system to recognise and attack tumour-specific antigens, thereby promoting an anti-tumour response [104]. Cancer vaccines can used either in combination with other conventional therapies or as a standalone treatment. Multiple current trials assessing the efficacy and safety of cancer vaccines have shown positive outcomes such as improved survival rates, tumour regression, and immune responses [105, 106]. Despite definite results, the clinical benefit of vaccines for TNBC treatment has not been conclusively established. This reinforces the need for large, randomised controlled trials providing additional efficacy and safety data.

Furthermore, combination of cancer vaccines (Fig. 2.3) with PARP inhibitors is a promising avenue, especially in the context of BRCA-mutant tumours. Dendritic cell-based vaccines may benefit from this approach. PARPi treatment of BRCA-mutant tumours activates the type I interferon (IFN) pathway via cGAS-STING signalling, which has multiple effects on the immune response, including the promotion of dendritic cell maturation, the release of interferons, and the initiation of anti-tumour T-cell responses [107]. This suggests that combining PARPis with dendritic cell-based vaccines could enhance the overall anti-tumour effect. Further, a peptide-based vaccine targeting Semaphorin 4A (Sema4A), a protein implicated in TNBC progression [108], has shown potential. Antigenic peptides derived from Sema4A have been identified and evaluated for their ability to stimulate immune responses. These peptides, combined with appropriate adjuvants and linkers, have demonstrated the ability to induce significant humoral and cellular immune responses in preclinical models [106]. These findings suggest that multi-epitope vaccines particularly personalised vaccines based on individual tumour antigens, could be a valuable addition to TNBC management [109], However, despite these advances, challenges remain in the development of effective cancer vaccines for TNBC. Key obstacles include identifying the most potent antigens, optimising vaccine formulations and delivery methods, and overcoming tumour-induced immunosuppression [110, 111]. Thus, further research is required to establish the safety and enhance the efficacy of these vaccines against TNBC.

Furthermore, Photothermal therapy (PTT) (Fig. 2.4) represents another innovative treatment approach, utilising near-infra-red (NIR) laser-absorbing agents such gold nanoparticles (AuNPs) to generate heat and ablate cancer cells upon NIR laser irradiation [112, 113]. Incorporating PTT with immunostimulatory monoclonal antibodies (such as anti-OX40 antibody), has shown substantial tumour regression in both treated and untreated tumours in preclinical models [114]. Thus, combination treatment of PTT and PARPis holds promise as the heat-mediated ablation by PTT would enhance the DNA damage induced by and drive the tumour cells into apoptosis more efficiently by PARPis [115]. This synergy is particularly helpful in surgical-resistant BRCA mutated TNBC as PTT provides a minimally invasive treatment option [112]. Overall, while there is no direct evidence of its efficacy in BRCA-mutated tumours, the potential to combine PTT with PARPis warrants investigation, particularly in BRCA-mutated TNBC cases.

Similar to PTT, Photodynamic therapy (PDT) (Fig. 2.5) has also gained attention as a minimally invasive treatment option for superficial tumours. PDT produces reactive oxygen species (ROS) in targeted cancer cells, inducing immunogenic cell death in TNBC cells through oxidative stress [115]. This stimulates dendritic cell maturation, enhances cytotoxic T lymphocyte (CTL) proliferation and intra-tumoral infiltration [116], resulting in TNBC, where immune resistance is a significant challenge, this enhanced immune activity results in ineffective inhibition of tumour growth, metastasis, consequently enhancing overall treatment efficacy [106]. Yao et al., [117, 118]. Alike PTT, PDT holds considerable potential in treating TNBC. Novel approaches such as self-cascading unimolecular prodrugs that combine PDT with chemotherapy possess the means to increase retention of photosensitizers in tumour tissues and have indicated their promise in clinical trial [119]. These developments may substantially broaden the use of PDT in combination strategies for TNBC. Further, when integrated with PARPis, the DNA damage caused by PARPis can be heightened by the oxidative stress induced through PDT, leading to an amplified therapeutic benefit. Thus, the combination of PARPis with PDT, could provide a synergistic approach to treatment for BRCA mutated TNBC [106, 119].

The efficacy and precision of non-drug treatment like PTT and PDT can be improved by the use of nanoparticle-based delivery systems such as multifunctional nanoparticles. These are designed to respond to environmental stimuli in the TME, enabling more precise targeting of tumour cells, particularly in BRCA-mutated TNBC [112, 115, 119]. Additionally, nanoparticles such as polymeric nanoparticles, liposomes, and dendrimers have revealed great potential in enhancing the delivery of PARPis while reducing systemic toxicity and off-target effects [112]. Overall, nanoparticle-based delivery systems possess great prospective in transforming treatment landscape for aggressive cancers such as TNBC and demand further research to evaluate their integration into PARPi-based treatment.

Moreover, another promising tool for TNBC therapy that has emerged is outer membrane vesicles (OMV) (Fig. 2.6). OMVs potent immunostimulatory properties and the potential for bioengineering offer an opportunity to enhance therapeutic efficacy [120]. Preclinical studies have demonstrated the potential of an OMV-based platform that combines photodynamic therapy, chemotherapy, and immunotherapy to completely eradicate TNBC in mice without observed side effects [106]. Given the immunostimulatory and tumour-targeting capabilities of OMVs, their combination with PARPis could provide a novel and effective treatment strategy, particularly in BRCA mutated TNBC. Additionally, a versatile biomimetic nanoplatform known as 4T1Mem@PGA-Ce6/Ola has been developed, combining the delivery of the photosensitizer Chlorin e6 (Ce6) and the Olaparib [121]. This platform exhibits excellent tumour-targeting capabilities and has shown potent synergistic anti-tumour effects under laser irradiation. The activation of the cGAS-STING pathway by this nanoplatform further enhances its therapeutic potential [121]. However, further preclinical and clinical research is necessary to fully establish the safety and efficacy of this approach.

Use of natural origin compounds (Fig. 2.7) for TNBC, particularly in combination with PARPis, has received significant attention due to their ability in improving efficacy and mitigating drug resistance of PARPis TNBC Compounds such as alkaloids, terpenoids, steroids, and flavonoids present a promising area of exploration for overcoming PARPi resistance [122]. Further, compounds such as cordycepin, ailanthone, polyphyllin III, and ursolic acid, possess apoptotic, anti-inflammatory and antioxidant properties, consequently showing potential in inhibiting cancer progression [123]. These compounds could also be a strong adjunct to PARPis, due to their potential to influence TME and have direct cytotoxic effects. Integration of these compounds into multimodal therapies would provide a cost-effective, low-toxicity strategy which would help reduce the adverse effects associated with PARPi treatment [123, 124]. Further, exploration in this area could create new possibilities for improving quality of life and overall survival of patient with TNBC.

Furthermore, CAR-T cell therapies (Fig. 2.8) are an inventive immunotherapeutic strategy which involves allowing T cells to recognize and target specific antigens expressed on the surface of tumour cells by genetically modifying the T cells to express a CAR. CAR-T therapy offers precise and potent therapeutic potential and is being explored as part of adoptive cell therapy advancements for treating TNBC [106]. However, to be successful, these approaches must address challenges related to tumour heterogeneity and immunosuppressive tumour microenvironments [125]. Several potential targets for BRCA mutated TNBC have been identified, some progressing to the clinical stages while others remain in the early phases of development (Table 4).

Table 4.

Potential targets for CAR-T therapy in BRCA mutated TNBC (Adapted from [152]).

Targets Mechanism of Action and Target Pathways Preclinical and Clinical Evidence
Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1) ROR1 is a type 1 membrane-spanning tyrosine kinase receptor involved in embryonic development and malignancies such as B-cell chronic lymphocytic leukaemia (B-CLL), mantle cell lymphoma, breast cancer, and ovarian cancer [153155]. ROR1-targeted CAR-T cells have shown effectiveness in preclinical models, including 3D breast cancer models, with evidence of tumour infiltration and immune response activation [155, 156]. Phase 1 trial (NCT02706392) demonstrated manageable side effects like mild cytokine release syndrome (CRS) and neurotoxicity, with some patients achieving stable disease
Mucin 1 (MUC1) MUC1 is a heavily glycosylated protein expressed on glandular epithelial cells. The tumour-associated form, tMUC1, is overexpressed in 95% of TNBCs, with no detectable expression in normal breast tissue [157159]. CAR-T cells targeting MUC1 variants have shown antigen-specific antitumor activity in TNBC models with minimal toxicity [158, 160162]. Ongoing trials (NCT04020575, NCT04025216, NCT02587689) are evaluating the safety of second-generation CAR-Ts against MUC1. Early reports suggest the need for further dose-escalation studies [152].
Disialoganglioside GD2 GD2 is a surface antigen expressed primarily in peripheral pain fibres, neurons, and melanocytes, and is linked to neuroblastoma and 60% of primary TNBC tumours [163, 164]. GD2-targeted CAR-T cells demonstrated tumour growth suppression and the ability to eradicate disseminated malignant cells in TNBC models [165]. Further research is required to validate these findings, particularly given inconsistent in vitro results [165].
Mesothelin Mesothelin, involved in cell adhesion, is overexpressed in TNBC and linked to oncogenic pathways including NF-κB, PI3K, and MAPK [166168]. Mesothelin-targeted CAR-T cells showed tumour suppression in TNBC models, particularly when combined with PD-1 checkpoint blockade strategies [169]. Clinical trials (NCT02792114, NCT02414269, NCT01355965, NCT02580747) are ongoing.
Epidermal Growth Factor Receptor (EGFR) EGFR, a member of the ERBB receptor tyrosine kinase family, is involved in cell proliferation and metastasis. EGFR is overexpressed in TNBC [170]. EGFR-targeted CAR-T cells have shown efficacy in preclinical TNBC models, with minimal off-tumour toxicity [146]. Tumour-restricted variants like EGFRvIII are being evaluated for safety. Trials are ongoing but results specific to TNBC are not yet published [171].
Folate Receptor Alpha (FRα) FRα, a GPI-anchored transmembrane protein, is involved in active folate transport and is overexpressed in TNBC, correlating with disease grade and stage [172, 173]. FRα-targeted CAR-T cells have shown tumour suppression in preclinical TNBC models, though effectiveness was lower compared to other cancers like ovarian cancer. Dual-targeting strategies are being explored to enhance efficacy [174, 175].
Receptor Tyrosine Kinase c-Met c-Met, or hepatocyte growth factor receptor (HGFR), is a tyrosine kinase involved in TNBC progression. Overexpression of c-Met correlates with poor overall survival [176, 177]. c-Met-directed CAR-T cells showed anti-tumour activity in TNBC cell lines and xenograft mouse models [178]. Phase I trials reported well-tolerated treatments, with some patients achieving stable or partial disease [178, 179].
Trophoblast Cell-Surface Antigen 2 (TROP2) TROP2, a transmembrane protein, is associated with poor prognosis in epithelial malignancies and mediates tumour-associated behaviours [88, 180]. Bi-specific TROP2/PD-L1 CAR-T cells showed higher tumoricidal activity compared to mono-specific CAR-Ts in preclinical models. However, more research is needed to validate TROP2 as a specific CAR-T target in BRCA-mutated TNBC [181, 182].
Chondroitin Sulphate Proteoglycan 4 (CSPG4) CSPG4 is a membrane-spanning glycoprotein involved in cancer progression and invasion, overexpressed in TNBC [183]. CAR-T cells targeting CSPG4 demonstrated tumour suppression, cytotoxicity, and cytokine secretion in preclinical models [184, 185]. Further clinical and preclinical data are necessary to support these findings.
Intracellular Adhesion Molecule-1 (ICAM-1) ICAM-1 is a transmembrane glycoprotein that mediates leucocyte transmigration and is upregulated in TNBC [186, 187]. Preclinical studies showed effective tumoricidal activity and a favourable safety profile, but ongoing assessments are needed to confirm ICAM-1 as a viable CAR-T target in BRCA mutated TNBC [188, 189].
Natural Killer Group 2, Member D Ligand (NKG2DL) NKG2DL is a stress-induced ligand overexpressed in TNBC, recognised by the NKG2D receptor [190]. NKG2DL-targeted CAR-T cells showed tumour growth suppression in preclinical TNBC models [43]. A Phase I clinical trial (NCT04107142) is assessing safety, with results pending.
Receptor Tyrosine Kinase AXL AXL, part of the TAM receptor tyrosine kinase family, is involved in tumour cell survival, expansion, and invasion. Abnormal AXL expression has been observed in breast cancer [191193]. CAR-T cells targeting AXL demonstrated significant tumoricidal activity in preclinical models. Enhancements like co-expressing a constitutively active IL-7 receptor further improved efficacy [194, 195].
Tumour Endothelial Marker 8 (TEM8) TEM8, also known as anthrax toxin receptor 1, is involved in endothelial cell migration and invasion, and is overexpressed in TNBC, correlating with relapse risk [196198]. TEM8-targeted CAR-T cells showed tumour suppression in preclinical TNBC models, but concerns about on-target off-tumour toxicity warrant further investigation before clinical trials [199].
Integrin Alpha V Beta 3 (αvβ3) αvβ3 integrins are adhesion receptors involved in tumour cell migration, tissue invasion, and survival, with expression in TNBC [200, 201]. CAR-T cells targeting αvβ3 showed potent tumoricidal activity in preclinical models, though their efficacy in TNBC remains uncertain. Further research is needed to assess their therapeutic potential in BRCA-mutated TNBC [202, 203].

Additionally, small interfering RNAs (siRNAs) (Fig. 2.9) also represent a promising area of exploration. Research into the regulatory signalling axis between p53, miR-34a, and PD-L1 in TNBC cells has shown that p53 can inhibit PD-L1 expression through miR-34a, leading to increased apoptosis and cytotoxicity in TNBC cells [126]. These findings suggest that targeting this axis could be a novel therapeutic strategy for BRCA-mutated TNBC, particularly when combined with PARPis.

Thus, the integration of these innovative therapies with PARPis presents a promising future direction for the treatment of TNBC, particularly in patients with BRCA mutations. Further research is required to optimise these combinations, evaluate their efficacy, and determine their potential in clinical practice.

Conclusion

TNBC has been the subject of an enormous amount of molecular characterisation and therapeutic experimentation. It has become clear that TNBC represents a spectrum of underlying molecular pathologies, of which DDR mechanistic failure is a critical hallmark in at least 50% of patients

Understanding the mechanisms of resistance and action of PARPis is crucial in treating BRCA-mutated TNBC. With PARP1 playing a key role in the DNA damage repair process, particularly through its association with the facilitation of HRR and base excision repair Its inhibition by PARPis results in the accumulation of DNA damage in tumour cells lacking functional BRCA1/2 pathways, consequently promoting cell death. Nonetheless, emerging mechanisms of resistance, such as reduced PARP1 trapping, restoration of HRR, stabilisation of the replication fork and drug efflux demand further exploration into combination strategies that may improve treatment efficacy. Additionally, insights being gained from developing combination approaches, with second and third-generation PARPis, will also aid in establishing new rational combination strategies with chemotherapeutics, targeted agents and ADCs.

Further, RNA-based drugs are the most promising therapies in the treatment of aggressive breast cancers, like TNBC. The targeted mechanism of action, reduced drug interactions, and lower overall toxicity and adverse effects of biologics highlight the potential of these therapies. Furthermore, in the future, more emphasis on combination therapies that integrate drug-based treatments with non-pharmacological approaches, such as photothermal and photodynamic therapies is anticipated. These combinations could alleviate the psychological and emotional burden on patients caused by extensive drug regimens while potentially reducing the side effects associated with multi-drug treatments. Therefore, this integrated approach seems to be a promising direction for future treatment protocols.

In the next decade, the term ‘triple-negative breast cancer’ may become obsolete as our understanding of the diverse molecular pathologies underlying this aggressive cancer subtype deepens. The current classification of TNBC is overly simplistic and insufficient for guiding effective treatment strategies. Instead, we anticipate a shift towards a more nuanced segmentation of TNBC, with patients categorised based on specific molecular aberrations within their tumours. Further, this evolution in classification could pave the way for the integration of RNA-based therapeutics. These targeted drugs, with their potential to be tailored to individual patient profiles, will not only enhance treatment efficacy but also significantly advance the field of personalised medicine. Thus, offering hope for more durable responses and improved outcomes for patients with what was once considered an intractable disease.

In conclusion, the future of TNBC treatment hinges on the development of targeted, personalised therapies that address the unique challenges of this aggressive subtype. While progress has been made in understanding TNBC’s molecular mechanisms and developing new treatments, further research is essential to optimise these strategies for clinical use. Exploring combination therapies, enhancing vaccine efficacy, and integrating multi-omics data will be crucial in advancing more effective, tailored treatments. Addressing drug resistance, especially to PARP inhibitors, remains a critical challenge, making the exploration of combination strategies and new mechanisms vital for improving treatment outcomes.

Acknowledgements

The author reports no acknowledgements for this work.

Author contributions

AJ was responsible for conducting the search, writing the report and creating the tables and figure. AB provided feedback on the report. CP contributed to the creation of Fig. 1 and provided feedback on the report.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contributed equally: Alan Barge, Christopher N. Parris.

Change history

4/1/2025

A Correction to this paper has been published: 10.1038/s41388-025-03364-6

Contributor Information

Aditi Jain, Email: jainaditik@gmail.com.

Christopher N. Parris, Email: chris.parris@aru.ac.uk

References

  • 1.WHO. Breast cancer now most common form of cancer: WHO taking action [Internet]. www.who.int. 2021 [Accessed 10 Aug 2024]. Available from: https://www.who.int/news/item/03-02-2021-breast-cancer-now-most-common-form-of-cancer-who-taking-action.
  • 2.Brown JS, O’Carrigan B, Jackson SP, Yap TA. Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov [Internet]. 2016;7:20–37. https://cancerdiscovery.aacrjournals.org/content/7/1/20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Karim AM, Eun Kwon J, Ali T, Jang J, Ullah I, Lee YG, et al. Triple-negative breast cancer: epidemiology, molecular mechanisms, and modern vaccine-based treatment strategies. Biochem Pharmacol [Internet]. 2023;212:115545. https://pubmed.ncbi.nlm.nih.gov/37044296/. [DOI] [PubMed] [Google Scholar]
  • 4.O’Reilly EA, Gubbins L, Sharma S, Tully R, Guang MHZ, Weiner-Gorzel K. et al. The fate of chemoresistance in triple-negative breast cancer (TNBC). BBA Clin [Internet].2015;3:257–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Upadhya A, Yadav KS, Misra A. Targeted drug therapy in non-small cell lung cancer: clinical significance and possible solutions-Part I. Expert Opin Drug Deliv. 2020;18:73–102. [DOI] [PubMed] [Google Scholar]
  • 6.Ghosh S, Javia A, Shetty S, Bardoliwala D, Maiti K, Banerjee S, et al. Triple negative breast cancer and non-small cell lung cancer: clinical challenges and nano-formulation approaches. J Control Release. 2021;337:27–58. [DOI] [PubMed] [Google Scholar]
  • 7.Mokhtari RB, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, et al. Combination therapy in combating cancer. Oncotarget [Internet]. 31;8. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5514969/. [DOI] [PMC free article] [PubMed]
  • 8.Li Y, Wu J, Qiu X, Dong S, He J, Liu J, et al. Bacterial outer membrane vesicles-based therapeutic platform eradicates triple-negative breast tumor by combinational photodynamic/chemo-/immunotherapy. Bioact Mater. 2023;20:548–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Metzger-Filho O, Tutt A, de Azambuja E, Saini KS, Viale G, Loi S, et al. Dissecting the heterogeneity of triple-negative breast cancer. J Clin Oncol. 2012;30:1879–87. [DOI] [PubMed] [Google Scholar]
  • 10.Zhang J, Lin Y, Sun XJ, Wang BY, Wang ZH, Luo JF, et al. Biomarker assessment of the CBCSG006 trial: a randomized phase III trial of cisplatin plus gemcitabine compared with paclitaxel plus gemcitabine as first-line therapy for patients with metastatic triple-negative breast cancer. Ann Oncol. 2018;29:1741–7. [DOI] [PubMed] [Google Scholar]
  • 11.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J Clin. 2018;68:394–424. [DOI] [PubMed] [Google Scholar]
  • 12.Liu H, Chen X, Jia Y, Chen H, Wang X, Liu G, et al. Facing inevitable PARPis resistance: mechanisms and therapeutic strategies for breast cancer treatment. Interdiscip Med. 2023;1:e20220013.
  • 13.Yin L, Duan JJ, Bian XW, Yu S. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22:61. [DOI] [PMC free article] [PubMed]
  • 14.Alexopoulos A, Karamouzis MV, Stavrinides H, Ardavanis A, Kandilis K, Stavrakakis J, et al. Phase II study of pegylated liposomal doxorubicin (Caelyx®) and docetaxel as first-line treatment in metastatic breast cancer. Ann Oncol. 2004;15:891–5. [DOI] [PubMed] [Google Scholar]
  • 15.El Gazzar B, Albakri KA, Hasan H, Badr AM, Farag AA, Saleh OM. Poly(ADP-ribose) polymerase inhibitors in the treatment landscape of triple-negative breast cancer (TNBC). J Oncol Pharm Pract. 2023;29:1467–79. [DOI] [PubMed] [Google Scholar]
  • 16.Ray Chaudhuri A, Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol [Internet]. 2017;18:610–21. https://www.nature.com/articles/nrm.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Azim HA, Kassem L, Azim Jr H. Integrating PARP inhibitors into the management of breast cancer: where are we? Chin Clin Oncol. 2021;10:50–0. [DOI] [PubMed] [Google Scholar]
  • 18.Satoh MS, Lindahl T. Role of poly(ADP-ribose) formation in DNA repair. Nature [Internet]. 1992;356:356–8. [DOI] [PubMed] [Google Scholar]
  • 19.Howard SM, Yanez DA, Stark JM. DNA damage response factors from diverse pathways, including DNA crosslink repair, mediate alternative end joining. Maizels N, editor. PLOS Genet. 2015;11:e1004943. [DOI] [PMC free article] [PubMed]
  • 20.Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1 and beyond. Nat Rev Cancer [Internet]. 2010;10:293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cong K, Peng M, Kousholt AN, Lee W, Lee S, Nayak S, et al. Replication gaps are a key determinant of PARP inhibitor synthetic lethality with BRCA deficiency. Mol Cell. 2021;81:3128–3144.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Barkauskaite E, Jankevicius G, Ahel I. Structures and mechanisms of enzymes employed in the synthesis and degradation of PARP-dependent protein ADP-ribosylation. Mol Cell. 2015;58:935–46. [DOI] [PubMed] [Google Scholar]
  • 23.Lord CJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355:1152–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Murai J, Huang SYN, Das BB, Renaud A, Zhang Y, Doroshow JH, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72:5588–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami I, Elliott R, et al. BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency. Clin Cancer Res. 2013;19:5003–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cerrato A, Morra F, Celetti A. Use of poly ADP-ribose polymerase [PARP] inhibitors in cancer cells bearing DDR defects: the rationale for their inclusion in the clinic. J Exp Clin Cancer Res. 2016;35. [DOI] [PMC free article] [PubMed]
  • 27.Roy R, Chun J, Powell SN. BRCA1 and BRCA2: different roles in a common pathway of genome protection. Nat Rev Cancer. 2012;12:68–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moynahan ME, Cui TY, Jasin M. Homology-directed dna repair, mitomycin-c resistance, and chromosome stability is restored with correction of a Brca1 mutation. PubMed. 2001;61:4842–50. [PubMed] [Google Scholar]
  • 29.Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, et al. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol. 2010;28:2512–9. [DOI] [PubMed] [Google Scholar]
  • 30.Pham MM, Hinchcliff E, Avila M, Westin SN. The clinical challenges, trials, and errors of combatting poly(ADP-Ribose) polymerase inhibitors resistance. Cancer J. 2021;27:491–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kondrashova O, Nguyen M, Shield-Artin K, Tinker AV, Teng NNH, Harrell MI, et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 2017;7:984–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Drost R, Bouwman P, Rottenberg S, Boon U, Schut E, Klarenbeek S, et al. BRCA1 RING function is essential for tumor suppression but dispensable for therapy resistance. Cancer Cell. 2011;20:797–809. [DOI] [PubMed] [Google Scholar]
  • 33.Johnson N, Johnson SF, Yao W, Li YC, Choi YE, Bernhardy AJ, et al. Stabilization of mutant BRCA1 protein confers PARP inhibitor and platinum resistance. Proc Natl Acad Sci [Internet]. 2013;110:17041–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rondinelli B, Gogola E, Yücel H, Duarte AA, van de Ven M, van der Sluijs R, et al. EZH2 promotes degradation of stalled replication forks by recruiting MUS81 through histone H3 trimethylation. Nat Cell Biol [Internet]. 2017;19:1371–8. [DOI] [PubMed] [Google Scholar]
  • 35.Ray Chaudhuri A, Callen E, Ding X, Gogola E, Duarte AA, Lee JE, et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature [Internet]. 2016;535:382–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pettitt SJ, Krastev DB, Brandsma I, Dréan A, Song F, Aleksandrov R, et al. Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat Commun [Internet]. 2018;9. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5945626/. [DOI] [PMC free article] [PubMed]
  • 37.Kanev PB, Varhoshkova S, Georgieva I, Lukarska M, Kirova D, Danovski G, et al. A unified mechanism for PARP inhibitor-induced PARP1 chromatin retention at DNA damage sites in living cells. Cell Rep. 2024;43:114234–4. [DOI] [PubMed] [Google Scholar]
  • 38.Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, et al. Whole–genome characterization of chemoresistant ovarian cancer. Nature [Internet]. 2015;521:489–94. [DOI] [PubMed] [Google Scholar]
  • 39.Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AOH, Zander SAL, et al. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci [Internet]. 2008;105:17079–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jaspers JE, Sol W, Kersbergen A, Schlicker A, Guyader C, Xu G, et al. BRCA2-deficient sarcomatoid mammary tumors exhibit multidrug resistance. Cancer Res [Internet]. 2014;75:732–41. [DOI] [PubMed] [Google Scholar]
  • 41.Han HS, Diéras V, Robson M, Palácová M, Marcom PK, Jager A, et al. Veliparib with temozolomide or carboplatin/paclitaxel versus placebo with carboplatin/paclitaxel in patients with BRCA1/2 locally recurrent/metastatic breast cancer: randomized phase II study. Ann Oncol. 2018;29:154–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Isakoff SJ, Overmoyer B, Tung NM, Gelman RS, Giranda VL, Bernhard KM, et al. A phase II trial of the PARP inhibitor veliparib (ABT888) and temozolomide for metastatic breast cancer. J Clin Oncol. 2010;28:1019–9. [Google Scholar]
  • 43.Han Y, Xie W, Song DG, Powell DJ. Control of triple-negative breast cancer using ex vivo self-enriched, costimulated NKG2D CAR T cells. J Hematol Oncol. 2018b;11. [DOI] [PMC free article] [PubMed]
  • 44.Isakoff S, Overmoyer B, Tung N, Gelman R, Habin K, Qian J, et al. P3-16-05: a phase II trial expansion cohort of the PARp inhibitor veliparib (ABT888) and temozolomide in BRCA1/2 associated metastatic breast cancer. Cancer Res [Internet]. 2011;71:P3–1605. https://aacrjournals.org/cancerres/article/71/24_Supplement/P3-16-05/568922/P3-16-05-A-Phase-II-Trial-Expansion-Cohort-of-the. [Google Scholar]
  • 45.Diéras V, Han HS, Kaufman B, Wildiers H, Friedlander M, Ayoub JP. et al. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer(BROCADE3): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet Oncology. 2020;21:1269–82. [DOI] [PubMed] [Google Scholar]
  • 46.Kunwor R, Silver DP, Abu-Khalaf M. PARP Inhibitors for treatment of BRCA1/2-mutated metastatic breast cancer- a systematic review and meta-analysis. Hematol/Oncol Stem Cell Ther. 2022;16. [DOI] [PubMed]
  • 47.Sharma P, Rodler E, Barlow WE, Gralow J, Huggins-Puhalla SL, Anders CK, et al. Results of a phase II randomized trial of cisplatin +/- veliparib in metastatic triple-negative breast cancer (TNBC) and/or germline BRCA-associated breast cancer (SWOG S1416). J Clin Oncol. 2020;38:1001–1. [Google Scholar]
  • 48.Bouberhan S, Pujade-Lauraine E, Cannistra SA. Advances in the management of platinum-sensitive relapsed ovarian cancer. J Clin Oncol. 2019;37:2424–36. [DOI] [PubMed] [Google Scholar]
  • 49.Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J Clin Oncol [Internet]. 2011;29:3008–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.O’Shaughnessy J, Osborne C, Pippen JE, Yoffe M, Patt D, Rocha C. et al. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N Engl J Med [Internet].2011;364:205–14. https://pubmed.ncbi.nlm.nih.gov/21208101/. [DOI] [PubMed] [Google Scholar]
  • 51.Pothuri B, Brodsky AL, Sparano JA, Blank SV, Kim M, Hershman DL, et al. Phase I and pharmacokinetic study of veliparib, a PARP inhibitor, and pegylated liposomal doxorubicin (PLD) in recurrent gynecologic cancer and triple-negative breast cancer with long-term follow-up. Cancer Chemother Pharmacol [Internet]. 2020;85:741–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.LaFargue CJ, Dal Molin GZ, Sood AK, Coleman RL. Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol [Internet]. 2019;20:e15–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yap TA, Im SA, Schram AM, Sharp A, Balmana J, Baird RD, et al. Abstract CT007: PETRA: first in class, first in human trial of the next generation PARP1-selective inhibitor AZD5305 in patients (pts) with BRCA1/2, PALB2 or RAD51C/D mutations. Cancer Res. 2022;82:CT007–7. [Google Scholar]
  • 54.Sharma P, López-Tarruella S, García-Saenz JA, Ward C, Connor CS, Gómez HL, et al. Efficacy of neoadjuvant carboplatin plus docetaxel in triple-negative breast cancer: combined analysis of two cohorts. Clin Cancer Res. 2016;23:649–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Schneeweiss A, Möbus V, Tesch H, Hanusch C, Denkert C, Lübbe K, et al. Intense dose-dense epirubicin, paclitaxel, cyclophosphamide versus weekly paclitaxel, liposomal doxorubicin (plus carboplatin in triple-negative breast cancer) for neoadjuvant treatment of high-risk early breast cancer (GeparOcto—GBG 84): a randomised phase III trial. Eur J Cancer. 2019;106:181–92. [DOI] [PubMed] [Google Scholar]
  • 56.Loibl S, O’Shaughnessy J, Untch M, Sikov WM, Rugo HS, McKee MD, 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 [Internet]. 2018;19:497–509. [DOI] [PubMed] [Google Scholar]
  • 57.Darlix A, Louvel G, Fraisse J, Jacot W, Brain E, Debled M, et al. Impact of breast cancer molecular subtypes on the incidence, kinetics and prognosis of central nervous system metastases in a large multicentre real-life cohort. Br J Cancer. 2019;121:991–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lee JS, Yost SE, Yuan Y. Neoadjuvant treatment for triple negative breast cancer: recent progresses and challenges. Cancers. 2020;12:1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Q, Bergholz J, Ding L, Lin Z, Kabraji S, Hughes ME, et al. STING agonism reprograms tumor-associated macrophages and overcomes resistance to PARP inhibition in BRCA1-deficient models of breast cancer. Nat Commun. 2022;13. [DOI] [PMC free article] [PubMed]
  • 60.Savas P, Loi S. Metastatic breast cancer: TIL it is too late. Clin Cancer Res. 2019;26:526–8. [DOI] [PubMed] [Google Scholar]
  • 61.Hutchinson KE, Yost SE, Chang CW, Johnson RM, Carr AR, McAdam PR, et al. Comprehensive profiling of poor-risk paired primary and recurrent triple-negative breast cancers reveals immune phenotype shifts. Clin Cancer Res. 2019;26:657–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pantelidou C, Sonzogni O, De Oliveria Taveira M, Mehta AK, Kothari A, Wang D, et al. PARP inhibitor efficacy depends on CD8+ T-cell recruitment via intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast cancer. Cancer Discov [Internet]. 2019;9:722–37. https://cancerdiscovery.aacrjournals.org/content/9/6/722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Staniszewska AD, Armenia J, King M, Michaloglou C, Reddy A, Singh M, et al. PARP inhibition is a modulator of anti-tumor immune response in BRCA-deficient tumors. Oncoimmunology [Internet]. 2022;11:2083755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chabanon RM, Muirhead G, Krastev DB, Adam J, Morel D, Garrido M, et al. PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non–small cell lung cancer. J Clin Investig. 2019;129:1211–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mosely SIS, Prime JE, Sainson RCA, Koopmann JO, Wang DYQ, Greenawalt DM, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res. 2017;5:29–41. [DOI] [PubMed] [Google Scholar]
  • 66.Stewart RA, Pilié PG, Yap TA. Development of PARP and immune-checkpoint inhibitor combinations. Cancer Res. 2018;78:6717–25. [DOI] [PubMed] [Google Scholar]
  • 67.Drew Y, de Jonge M, Hong SH, Park YH, Wolfer A, Brown J, et al. An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): results in germline BRCA -mutated (gBRCA m) platinum-sensitive relapsed (PSR) ovarian cancer (OC). Gynecol Oncol. 2018;149:246–7. [Google Scholar]
  • 68.Konstantinopoulos PA, Waggoner S, Vidal GA, Mita M, Moroney JW, Holloway R, et al. Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncol [Internet]. 2019;5:1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Boussios S, Karihtala P, Moschetta M, Karathanasi A, Sadauskaite A, Rassy E, et al. Combined strategies with poly (ADP-Ribose) polymerase (PARP) inhibitors for the treatment of ovarian cancer: a literature review. Diagnostics [Internet]. 2019;9:87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Färkkilä A, Gulhan DC, Casado J, Jacobson CA, Nguyen H, Kochupurakkal B, et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun. 2020;11:1459 [DOI] [PMC free article] [PubMed]
  • 71.Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Investig. 2015;125:3413–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS–STING pathway of cytosolic DNA sensing. Nat Immunol [Internet]. 2016;17:1142–9. https://www.nature.com/articles/ni.3558. [DOI] [PubMed] [Google Scholar]
  • 73.Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Peyraud F, Italiano A. Combined PARP inhibition and immune checkpoint therapy in solid tumors. Cancers. 2020;12:1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Domchek S, Postel-Vinay S, Im S-A, Hee Park Y, Delord J-P, Italiano A, et al. Abstract PD5-04: An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): updated results in patients with germline BRCA mutated (gBRCAm) metastatic breast cancer (MBC). Cancer Res. 2019;79:PD5-04PD5-04.
  • 76.Domchek SM, Postel-Vinay S, Im SA, Park YH, Delord JP, Italiano A, 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;21:1155–64. [DOI] [PubMed] [Google Scholar]
  • 77.Pilié PG, Gay CM, Byers LA, O’Connor MJ, Yap TA. PARP Inhibitors: extending benefit beyond BRCA-mutant cancers. Clin Cancer Res [Internet]. 2019;25:3759–71. [DOI] [PubMed] [Google Scholar]
  • 78.Drew Y, Kaufman B, Banerjee S, Lortholary A, Hong SH, Park YH, et al. 1190PD - Phase II study of olaparib + durvalumab (MEDIOLA): updated results in germline BRCA-mutated platinum-sensitive relapsed (PSR) ovarian cancer (OC). Ann Oncol [Internet] 2019;30:v485–6. [Google Scholar]
  • 79.Jiao S, Xia W, Yamaguchi H, Wei Y, Chen MK, Hsu JM, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res. 2017;23:3711–20. Feb 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sen T, Rodriguez BL, Chen L, Corte CMD, Morikawa N, Fujimoto J, et al. Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung cancer. Cancer Discov. 2019;9:646–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Revythis A, Limbu A, Mikropoulos C, Ghose A, Sanchez E, Sheriff M, et al. Recent Insights into PARP and immuno-checkpoint inhibitors in epithelial ovarian cancer. Int J Environ Res Public Health [Internet]. 2022;19:8577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jalalvand M, Darbeheshti F, Rezaei N. Immune checkpoint inhibitors: review of the existing evidence and challenges in breast cancer. Immunotherapy. 2021;13:587–603. [DOI] [PubMed] [Google Scholar]
  • 83.Lyons TG. Targeted therapies for triple-negative breast cancer. Curr Treat Options Oncol. 2019;20. [DOI] [PubMed]
  • 84.Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 2017;16:315–37. [DOI] [PubMed]
  • 85.Baah S, Laws M, Rahman KM. Antibody–drug conjugates—a tutorial review. Molecules. 2021;26:2943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Conilh L, Sadilkova L, Viricel W, Dumontet C. Payload diversification: a key step in the development of antibody–drug conjugates. J Hematol Oncol. 2023;16. [DOI] [PMC free article] [PubMed]
  • 87.Keskinkilic M, Sacks R. Antibody-Drug Conjugates in Triple Negative Breast Cancer. Clinical Breast Cancer. 2024;24:163–74. [DOI] [PubMed] [Google Scholar]
  • 88.Goldenberg D, Stein R, Sharkey R. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget [Internet]. 2018; Available from: https://www.semanticscholar.org/paper/The-emergence-of-trophoblast-cell-surface-antigen-2-Goldenberg-Stein/f5e70445dd34cc5d737dd26dd33715a2bda421e4. [DOI] [PMC free article] [PubMed]
  • 89.Keskinkilic M, Sacks R. Antibody-drug conjugates in triple negative breast cancer. Clin Breast Cancer. 2024;24:163–74. [DOI] [PubMed] [Google Scholar]
  • 90.Kumar A, Fernandez-Capetillo O, Carrera AC. Nuclear phosphoinositide 3-kinase controls double-strand break DNA repair. Proc Natl Acad Sci. 2010;107:7491–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Juvekar A, Burga LN, Hu H, Lunsford EP, Ibrahim YH, Balmañà J, et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov [Internet]. 2012;2:1048–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Rehman FL, Lord CJ, Ashworth A. The promise of combining inhibition of PI3K and PARP as cancer therapy. Cancer Discov. 2012;2:982–4. [DOI] [PubMed] [Google Scholar]
  • 93.Ibrahim YH, García-García C, Serra V, He L, Torres-Lockhart K, Prat A, et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov [Internet]. 2012;2:1036–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Labrie M, Ju Z, Litton JK, Kim TB, Lee S, Chen K, et al. Abstract 2070: exploration of markers of synergistic lethality of PARP and PI3K-Akt-mTOR inhibitors in women’s cancers. Cancer Res. 2019;79:2070–0. [Google Scholar]
  • 95.Childers CP, Childers KK, Maggard-Gibbons M, Macinko J. National estimates of genetic testing in women with a history of breast or ovarian cancer. J Clin Oncol [Internet]. 2017;35:3800–6. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5707208/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schmid P, Cortes J, Dent R, Pusztai L, McArthur H, Kümmel S, et al. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386:556–67. [DOI] [PubMed] [Google Scholar]
  • 97.Morganti S, Bychkovsky BL, Poorvu PD, Garrido-Castro AC, Weiss A, Block CC, et al. Adjuvant olaparib for germline BRCA carriers with HER2-negative early breast cancer: evidence and controversies. Oncologist [Internet]. 2023;28:565–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Morganti S, Marra A, De Angelis C, Toss A, Licata L, Giugliano F, et al. PARP inhibitors for breast cancer treatment: a review. JAMA Oncol [Internet]. 2024;10:658–70. [DOI] [PubMed] [Google Scholar]
  • 99.Diéras V, Han HS, Kaufman B, Wildiers H, Friedlander M, Ayoub JP, et al. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol [Internet]. 2020;21:1269–82. [DOI] [PubMed] [Google Scholar]
  • 100.Geyer CE, Sikov WM, Huober J, Rugo HS, Wolmark N, O’Shaughnessy J, et al. Long-term efficacy and safety of addition of carboplatin with or without veliparib to standard neoadjuvant chemotherapy in triple-negative breast cancer: 4-year follow-up data from BrighTNess, a randomized phase III trial. Ann Oncol. 2022;33:384–94. [DOI] [PubMed] [Google Scholar]
  • 101.Fasching PA, Link T, Hauke J, Seither F, Jackisch C, Klare P, et al. Neoadjuvant paclitaxel/olaparib in comparison to paclitaxel/carboplatinum in patients with HER2-negative breast cancer and homologous recombination deficiency (GeparOLA study). Ann Oncol. 2021;32:49–57. [DOI] [PubMed] [Google Scholar]
  • 102.Sammons SL, Tan TJ, Im YH, Traina TA, Anders C, Renzulli E, et al. Abstract PD11-12: PD11-12 DORA: a phase II, multicenter, international, non-comparator study of olaparib (O) +/- durvalumab (D) as a chemotherapy-free maintenance strategy in platinum-treated advanced triple-negative breast cancer (aTNBC). Cancer Res. 2023;83:PD11–12. [Google Scholar]
  • 103.Arnold MR, Langelier MF, Gartrell J, Kirby IT, Sanderson DJ, Bejan DS, et al. Allosteric regulation of DNA binding and target residence time drive the cytotoxicity of phthalazinone-based PARP-1 inhibitors. Cell Chem Biol [Internet]. 2022;29:1694–1708.e10. [DOI] [PubMed] [Google Scholar]
  • 104.Greten TF, Jaffee EM. Cancer vaccines. J Clin Oncol. 1999;17:1047. [DOI] [PubMed] [Google Scholar]
  • 105.Toh U, Sakurai S, Saku S, Takao Y, Okabe M, Iwakuma N, et al. Early phase II study of mixed 19‐peptide vaccine monotherapy for refractory triple‐negative breast cancer. Cancer Sci. 2020;111:2760–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Novel immunotherapies for breast cancer: Focus on 2023 findings. Int Immunopharmacol. 2024;128:111549–9. [DOI] [PubMed] [Google Scholar]
  • 107.Polgár C, Ott OJ, Hildebrandt G, Kauer-Dorner D, Knauerhase H, Major T, et al. Late side-effects and cosmetic results of accelerated partial breast irradiation with interstitial brachytherapy versus whole-breast irradiation after breast-conserving surgery for low-risk invasive and in-situ carcinoma of the female breast: 5-year results of a randomised, controlled, phase 3 trial. Lancet Oncol. 2017;18:259–68. [DOI] [PubMed] [Google Scholar]
  • 108.Chen H, Gao C, Wang L, Zhao W, Zhang B, Zhang J. The expression and clinical significance of Sema4A in triple-negative breast cancer. J Clin Nurs Res. 2020;4.
  • 109.Dhanushkumar T, Kamaraj B, Vasudevan K, Gopikrishnan M, Dasegowda KR, Rambabu M, et al. Structural immunoinformatics approach for rational design of a multi-epitope vaccine against triple-negative breast cancer. Int J Biol Macromol. 2023;243:125209–9. [DOI] [PubMed] [Google Scholar]
  • 110.Zhu SY, Yu KD. Breast cancer vaccines: disappointing or promising? Front Immunol. 2022;13. [DOI] [PMC free article] [PubMed]
  • 111.Behravan J, Razazan A, Behravan G. Towards breast cancer vaccines, progress and challenges. Curr Drug Discov Technol [Internet]. 2019;16:251–8. [DOI] [PubMed] [Google Scholar]
  • 112.Nag S, Mitra O, Tripathi G, Adur I, Mohanto S, Nama M, et al. Nanomaterials-assisted photothermal therapy for breast cancer: State-of-the-art advances and future perspectives. Photodiagn Photodyn Ther [Internet]. 2024;45:103959 . [DOI] [PubMed] [Google Scholar]
  • 113.Zou L, Wang H, He B, Zeng L, Tan T, Cao H, et al. Current approaches of photothermal therapy in treating cancer metastasis with nanotherapeutics. Theranostics. 2016;6:762–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Stecklein SR, Yoder R, Salgado R, Staley JM, O’Dea A, Nye L, et al. Abstract PD1-06: black patients with triple negative breast cancer (TNBC) have enriched stromal tumor infiltrating lymphocytes (sTILs) and receive preferential benefit from neoadjuvant immunotherapy. Cancer Res. 2023;83:PD1-06PD1-06.
  • 115.Li Y, Miao W, Yuan C, Tang J, Zhong N, Jin Y, et al. PARP inhibitors boost the efficacy of photothermal therapy to TNBC through enhanced DNA damage and inhibited homologous recombination repair. Drug Deliv Transl Res [Internet]. 2024 [Accessed 27 Sep 2024]; Available from: 10.1007/s13346-024-01650-6. [DOI] [PubMed]
  • 116.Hwang HS, Shin H, Han J, Na K. Combination of photodynamic therapy (PDT) and anti-tumor immunity in cancer therapy. J Pharm Investig. 2018;48:143–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Dolmans D, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer. 2003;3:380–7. [DOI] [PubMed] [Google Scholar]
  • 118.Yao D, Wang Y, Bian K, Zhang B, Wang D. A self-cascaded unimolecular prodrug for pH-responsive chemotherapy and tumor-detained photodynamic-immunotherapy of triple-negative breast cancer. Biomaterials. 2023;292:121920–0. [DOI] [PubMed] [Google Scholar]
  • 119.Thakur V, Kutty RV Recent advances in nanotheranostics for triple-negative breast cancer treatment. J Exp Clin Cancer Res [Internet]. 2019;38. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819447/. [DOI] [PMC free article] [PubMed]
  • 120.Turner KB, Walper SA. Bacterial outer membrane vesicles: an emerging tool in vaccine development, as adjuvants, and for therapeutic delivery. Drug Deliv Lett.. 2017;7:83–91.
  • 121.Wu H, Du X, Xu J, Kong X, Li Y, Liu D, et al. Multifunctional biomimetic nanoplatform based on photodynamic therapy and DNA repair intervention for the synergistic treatment of breast cancer. Acta Biomater. 2023;157:551–65. [DOI] [PubMed] [Google Scholar]
  • 122.Ramasamy T, Priyankha S, Kannan M, Prakash M, Selvam C. Compounds from diverse natural origin against triple‐negative breast cancer: a comprehensive review. Chem Biol Drug Des. 2022;101. [DOI] [PubMed]
  • 123.Kumar S, Gupta S, Gupta SC. Editorial: targeting triple-negative breast cancer by natural compounds. Front Pharmacol. 2023;14. [DOI] [PMC free article] [PubMed]
  • 124.Kushwaha PP, Vardhan PS, Kapewangolo P, Shuaib M, Prajapati SK, Singh AK, et al. Bulbine frutescens phytochemical inhibits notch signaling pathway and induces apoptosis in triple-negative and luminal breast cancer cells. Life Sci [Internet]. 2019;234:116783. [DOI] [PubMed] [Google Scholar]
  • 125.Toulouie S, Johanning G, Shi Y. Chimeric antigen receptor T-cell immunotherapy in breast cancer: development and challenges. J Cancer. 2021;12:1212–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Deng S, Wang M, Wang C, Zeng Y, Xue Q, Tan Y, et al. p53 downregulates PD-L1 expression via miR-34a to inhibit the growth of triple-negative breast cancer cells: a potential clinical immunotherapeutic target. Mol Biol Rep. 2022;50:577–87. [DOI] [PubMed] [Google Scholar]
  • 127.Lee EK, Matulonis UA. PARP inhibitor resistance mechanisms and implications for post-progression combination therapies. Cancers. 2020;12:2054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Haynes B, Murai J, Lee JM. Restored replication fork stabilization, a mechanism of PARP inhibitor resistance, can be overcome by cell cycle checkpoint inhibition. Cancer Treat Rev. 2018;71:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Do KT, Kochupurakkal B, Kelland S, de Jonge A, Hedglin J, Powers A, et al. Phase 1 combination study of the CHK1 inhibitor prexasertib and the PARP inhibitor olaparib in high-grade serous ovarian cancer and other solid tumors. Clin Cancer Res. 2021;27:4710–6. [DOI] [PubMed] [Google Scholar]
  • 130.Kaplan AR, Gueble SE, Liu Y, Oeck S, Kim H, Yun Z, et al. Cediranib suppresses homology-directed DNA repair through down-regulation of BRCA1/2 and RAD51. Sci Transl Med. 2019;11:eaav4508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chan N, Koritzinsky M, Zhao H, Bindra R, Glazer PM, Powell S, et al. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance. Cancer Res. 2008;68:605–14. [DOI] [PubMed] [Google Scholar]
  • 132.Bristow RG, Hill RP. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer [Internet]. 2008;8:180–92. https://www.ncbi.nlm.nih.gov/pubmed/18273037. [DOI] [PubMed] [Google Scholar]
  • 133.Lim J, Yang K, Taylor-Harding B, Wiedemeyer WR, Buckanovich RJ. VEGFR3 inhibition chemosensitizes ovarian cancer stemlike cells through down-regulation of BRCA1 and BRCA2. Neoplasia. 2014;16:343–353.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Mirza MR, Åvall Lundqvist E, Birrer MJ, dePont Christensen R, Nyvang GB, Malander S, et al. Niraparib plus bevacizumab versus niraparib alone for platinum-sensitive recurrent ovarian cancer (NSGO-AVANOVA2/ENGOT-ov24): a randomised, phase 2, superiority trial. Lancet Oncol [Internet]. 2019;20:1409–19. [DOI] [PubMed] [Google Scholar]
  • 135.Sun C, Yin J, Fang Y, Chen J, Jeong KJ, Chen X, et al. BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell. 2018;33:401–416.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zhang B, Lyu J, Yang EJ, Liu Y, Wu C, Pardeshi L, et al. Class I histone deacetylase inhibition is synthetic lethal with BRCA1 deficiency in breast cancer cells. Acta Pharm Sin B. 2020;10:615–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gupta V, Hirst J, Petersen S, Roby KF, Kusch M, Zhou H, et al. Entinostat, a selective HDAC1/2 inhibitor, potentiates the effects of olaparib in homologous recombination proficient ovarian cancer. Gynecol Oncol. 2021;162:163–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Abramson JS, Blum KA, Flinn IW, Gutierrez M, Goy A, Maris MB, et al. BET inhibitor CPI-0610 is well tolerated and induces responses in diffuse large B-cell lymphoma and follicular lymphoma: preliminary analysis of an ongoing phase 1 study. Blood. 2015;126:1491–1. [Google Scholar]
  • 139.Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol [Internet]. 2016;3:e196–204. [DOI] [PubMed] [Google Scholar]
  • 140.Cochran AG, Conery AR, Sims RJ. Bromodomains: a new target class for drug development. Nat Rev Drug Discov. 2019;18:609–28. [DOI] [PubMed] [Google Scholar]
  • 141.Jiang J, Yang ES, Jiang G, Nowsheen S, Wang H, Wang T, et al. p53-dependent BRCA1 nuclear export controls cellular susceptibility to DNA damage. Cancer Res. 2011;71:5546–57. [DOI] [PubMed] [Google Scholar]
  • 142.Yang YH, Liu JW, Lu C, Wei JF. CAR-T cell therapy for breast cancer: from basic research to clinical application. Int J Biol Sci. 2022;18:2609–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Barazas M, Gasparini A, Huang Y, Küçükosmanoğlu A, Annunziato S, Bouwman P, et al. Radiosensitivity is an acquired vulnerability of PARPi-resistant BRCA1-deficient tumors. Cancer Res [Internet]. 2019;79:452–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Reiss KA, Herman JM, Zahurak M, Brade A, Dawson LA, Scardina A, et al. A phase I study of veliparib (ABT-888) in combination with low-dose fractionated whole abdominal radiation therapy in patients with advanced solid malignancies and peritoneal carcinomatosis. Clin Cancer Res. 2015;21:68–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Czito BG, Deming DA, Jameson GS, Mulcahy MF, Vaghefi H, Dudley MW, et al. Safety and tolerability of veliparib combined with capecitabine plus radiotherapy in patients with locally advanced rectal cancer: a phase 1b study. Lancet Gastroenterol Hepatol. 2017;2:418–26. [DOI] [PubMed] [Google Scholar]
  • 146.Liu Y, Zhou Y, Huang KH, Li Y, Fang X, An L. et al. EGFR-specific CAR-T cells trigger cell lysis in EGFR-positive TNBC. Aging [Internet]. 2019;11:11054–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ghose A, Gullapalli SVN, Chohan N, Bolina A, Moschetta M, Rassy E, et al. Applications of proteomics in ovarian cancer: dawn of a new era. Proteomes. 2022;10:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Mosler T, Baymaz HI, Gräf JF, Mikicic I, Blattner G, Bartlett E, et al. PARP1 proximity proteomics reveals interaction partners at stressed replication forks. Nucleic Acids Res. 2022;50:11600–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Cardoso F, van’t Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med. 2016;375:717–29. [DOI] [PubMed] [Google Scholar]
  • 150.Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med [Internet]. 2018;379:111–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Woodhouse R, Li M, Hughes J, Delfosse D, Skoletsky J, Ma P, et al. Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay for cancers of solid tumor origin. PLoS ONE [Internet]. 2020;15. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7518588/. [DOI] [PMC free article] [PubMed]
  • 152.Nasiri F, Kazemi M, Mirarefin SMJ, Mahboubi Kancha M, Ahmadi Najafabadi M, Salem F, et al. CAR-T cell therapy in triple-negative breast cancer: hunting the invisible devil. Front Immunol. 2022;13:1018786. [DOI] [PMC free article] [PubMed]
  • 153.Matsuda T, Nomi M, Ikeya M, Kani S, Oishi I, Terashima T, et al. Expression of the receptor tyrosine kinase genes, Ror1 and Ror2, during mouse development. Mech Dev. 2001;105:153–6. [DOI] [PubMed] [Google Scholar]
  • 154.Hudecek M, Schmitt TM, Baskar S, Lupo-Stanghellini MT, Nishida T, Yamamoto TN, et al. The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor. Blood. 2010;116:4532–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Hudecek M, Lupo-Stanghellini MT, Kosasih PL, Sommermeyer D, Jensen MC, Rader C, et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res. 2013;19:3153–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wallstabe L, Göttlich C, Nelke LC, Kühnemundt J, Schwarz T, Nerreter T, et al. ROR1-CAR T cells are effective against lung and breast cancer in advanced microphysiologic 3D tumor models. JCI Insight [Internet]. 2019 [Accessed 21 Aug 2020]; 4. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6795380/pdf/jciinsight-4-126345.pdf. [DOI] [PMC free article] [PubMed]
  • 157.Gendler SJ, Taylor-Papadimitriou J, Duhig T, Rothbard JB, Burchell J. A highly immunogenic region of a human polymorphic epithelial mucin expressed by carcinomas is made up of tandem repeats. J Biol Chem. 1988;263:12820–3. [PubMed] [Google Scholar]
  • 158.Zhou R, Yazdanifar M, Roy LD, Whilding LM, Gavrill A, Maher J, et al. CAR T cells targeting the tumor MUC1 glycoprotein reduce triple-negative breast cancer growth. Front Immunol [Internet]. 2019;10:1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, Duhig T, Peat N, Burchell J, et al. Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem. 1990;265:15286–93. [PubMed] [Google Scholar]
  • 160.Safarzadeh Kozani P, Rahbarizadeh F. CAR T cells redirected against tumor-specific antigen glycoforms: can low-sugar antigens guarantee a sweet success? Front Med. 2022;16:322–38. [DOI] [PubMed] [Google Scholar]
  • 161.Posey Avery D, Schwab Robert D, Boesteanu Alina C, Steentoft C, Mandel U, Engels B, et al. Engineered CAR T cells targeting the cancer-associated tn-glycoform of the membrane Mucin MUC1 control adenocarcinoma. Immunity. 2016;44:1444–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zhai X, You F, Xiang S, Jiang L, Chen D, Li Y, et al. MUC1-Tn-targeting chimeric antigen receptor-modified Vγ9Vδ2 T cells with enhanced antigen-specific anti-tumor activity. Am J Cancer Res. 2021;11:79–91. [PMC free article] [PubMed] [Google Scholar]
  • 163.Ly S, Anand V, El-Dana F, Nguyen K, Cai Y, Cai S, et al. Anti-GD2 antibody dinutuximab inhibits triple-negative breast tumor growth by targeting GD2+ breast cancer stem-like cells. J Immunother Cancer. 2021;9:e001197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Voeller J, Sondel PM. Advances in anti-GD2 immunotherapy for treatment of high-risk neuroblastoma. J Pediatr Hematol/Oncol. 2019;41:163–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Seitz CM, Schroeder S, Knopf P, Krahl AC, Hau J, Schleicher S, et al. GD2-targeted chimeric antigen receptor T cells prevent metastasis formation by elimination of breast cancer stem-like cells. OncoImmunology. 2019;9:1683345. [DOI] [PMC free article] [PubMed]
  • 166.Tozbikian G, Brogi E, Kadota K, Catalano J, Akram M, Patil S, et al. Mesothelin expression in triple-negative breast carcinomas correlates significantly with basal-like phenotype, distant metastases and decreased survival. Coleman WB, editor. PLoS ONE. 2014;9:e114900. [DOI] [PMC free article] [PubMed]
  • 167.Parinyanitikul N, Blumenschein GR, Wu Y, Lei X, Chavez-MacGregor M, Smart M, et al. Mesothelin expression and survival outcomes in triple receptor-negative breast cancer. Clin Breast Cancer. 2013;13:378–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Klampatsa A, Dimou V, Albelda SM. Mesothelin-targeted CAR-T cell therapy for solid tumors. Expert Opin Biol Ther. 2020;21:1–14. [DOI] [PubMed] [Google Scholar]
  • 169.Hu W, Zi Z, Jin Y, Li G, Shao K, Cai Q, et al. CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions. Cancer Immunol, Immunother. 2018;68:365–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Xia L, Zheng Z, Liu J, Chen Y, Ding J, Xia N, et al. EGFR‐targeted CAR‐T cells are potent and specific in suppressing triple‐negative breast cancer both in vitro and in vivo. Clin Transl Immunol [Internet]. 2020 [Accessed 16 Apr 2023];9. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7196685/. [DOI] [PMC free article] [PubMed]
  • 171.Yoon D, Osborn M, Tolar J, Kim C. Incorporation of immune checkpoint blockade into chimeric antigen receptor T Cells (CAR-Ts): combination or built-In CAR-T. Int J Mol Sci. 2018;19:340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Salazar MD, Ratnam M. The folate receptor: What does it promise in tissue-targeted therapeutics? Cancer Metastasis Rev. 2007;26:141–52. [DOI] [PubMed] [Google Scholar]
  • 173.Boogerd LSF, Boonstra MC, Beck AJ, Charehbili A, Hoogstins CES, Prevoo HAJM, et al. Concordance of folate receptor-α expression between biopsy, primary tumor and metastasis in breast cancer and lung cancer patients. Oncotarget. 2016;7:17442–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Song DG, Ye Q, Poussin M, Chacon JA, Figini M, Powell DJ. Effective adoptive immunotherapy of triple-negative breast cancer by folate receptor-alpha redirected CAR T cells is influenced by surface antigen expression level. J Hematol Oncol. 2016;9:56. [DOI] [PMC free article] [PubMed]
  • 175.Lanitis E, Poussin M, Klattenhoff AW, Song DG, Sandaltzopoulos R, June CH, et al. Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Resist. 2013;1:43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Zhao X, Qu J, Hui Y, Zhang H, Sun Y, Liu X, et al. Clinicopathological and prognostic significance of c-Met overexpression in breast cancer. Oncotarget. 2017;8:56758–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kim YJ, Choi JS, Seo J, Song JY, Lee SE, Kwon MJ, et al. MET is a potential target for use in combination therapy with EGFR inhibition in triple-negative/basal-like breast cancer. Int J Cancer [Internet]. 2014;134:2424–36. [DOI] [PubMed] [Google Scholar]
  • 178.Tchou J, Zhao Y, Levine BL, Zhang PJ, Davis MM, Melenhorst JJ, et al. Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol Res [Internet]. 2017;5:1152–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Shah PD, Huang AC, Xu X, Zhang PJ, Orlowski R, Matlawski T, et al. Phase I trial of autologous cMET-directed CAR-t cells administered intravenously in patients with melanoma & breast carcinoma. J Clin Oncol. 2020;38:10035–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/ therapeutic implications. Genes Cancer. 2014;6:84–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Zhao W, Jia L, Zhang M, Huang X, Qian P, Tang Q, et al. The killing effect of novel bi-specific Trop2/PD-L1 CAR-T cell targeted gastric cancer. PubMed. 2019;9:1846–56. [PMC free article] [PubMed] [Google Scholar]
  • 182.Bedoya DM, King T, Posey AD. Generation of CART cells targeting oncogenic TROP2 for the elimination of epithelial malignancies. Cytotherapy. 2019;21:S11–2. [Google Scholar]
  • 183.Ilieva KM, Cheung A, Mele S, Chiaruttini G, Crescioli S, Griffin M, et al. Chondroitin sulfate proteoglycan 4 and its potential as an antibody immunotherapy target across different tumor types. Front Immunol [Internet]. 2018;8. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5767725/. [DOI] [PMC free article] [PubMed]
  • 184.Geldres C, Savoldo B, Hoyos V, Caruana I, Zhang M, Yvon E, et al. T lymphocytes redirected against the chondroitin sulfate proteoglycan-4 control the growth of multiple solid tumors both in vitro and in vivo. Clin Cancer Res: Off J Am Assoc Cancer Res [Internet]. 2014;20:962–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Beard RE, Zheng Z, Lagisetty KH, Burns WR, Tran E, Hewitt SM, et al. Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J Immunother Cancer [Internet]. 2014;2:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Müller N. The role of intercellular adhesion molecule-1 in the pathogenesis of psychiatric disorders. Front Pharmacol. 2019;10:1251. [DOI] [PMC free article] [PubMed]
  • 187.Guo P, Huang J, Wang L, Jia D, Yang J, Dillon DA, et al. ICAM-1 as a molecular target for triple-negative breast cancer. Proc Natl Acad Sci. 2014;111:14710–5. Sep 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Vedvyas Y, McCloskey JE, Yang Y, Min IM, Fahey TJ, Zarnegar R, et al. Manufacturing and preclinical validation of CAR T cells targeting ICAM-1 for advanced thyroid cancer therapy. Sci Rep. 2019;9:10634. [DOI] [PMC free article] [PubMed]
  • 189.Yang Y, Vedvyas Y, McCloskey JE, Min IM, Jin MM. Abstract 2322: ICAM-1 targeting CAR T cell therapy for triple-negative breast cancer. Cancer Res. 2019;79:2322–2. [Google Scholar]
  • 190.Morisaki T, Onishi H, Katano M. Cancer immunotherapy using NKG2D and DNAM-1 systems. PubMed. 2012;32:2241–7. [PubMed] [Google Scholar]
  • 191.Feneyrolles C, Spenlinhauer A, Guiet L, Fauvel B, Daydé-Cazals B, Warnault P, et al. Axl kinase as a key target for oncology: focus on small molecule inhibitors. Mol Cancer Ther. 2014;13:2141–8. [DOI] [PubMed] [Google Scholar]
  • 192.Barata PC, Rini BI. Treatment of renal cell carcinoma: current status and future directions. CA: A Cancer J Clin. 2017;67:507–24. [DOI] [PubMed] [Google Scholar]
  • 193.Holland SJ, Pan A, Franci C, Hu Y, Chang B, Li W, et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 2010;70:1544–54. [DOI] [PubMed] [Google Scholar]
  • 194.Wei J, Sun H, Zhang A, Wu X, Li Y, Liu J, et al. A novel AXL chimeric antigen receptor endows T cells with anti-tumor effects against triple-negative breast cancers. Cell Immunol. 2018;331:49–58. [DOI] [PubMed] [Google Scholar]
  • 195.Zhao Z, Li Y, Liu W, Li X. Engineered IL-7 receptor enhances the therapeutic effect of AXL-CAR-T cells on triple-negative breast cancer. BioMed Res Int. 2020;2020:1–13. Jan 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Fu S, Tong X, Cai C, Zhao Y, Wu Y, Li Y, et al. The structure of tumor endothelial marker 8 (TEM8) extracellular domain and implications for its receptor function for recognizing anthrax toxin. Xu W, editor. PLoS ONE. 2010;5:e11203 [DOI] [PMC free article] [PubMed]
  • 197.Opoku-Darko M, Yuen C, Gratton K, Sampson E, Bathe OF. Tumor endothelial marker 8 overexpression in breast cancer cells enhances tumor growth and metastasis. Cancer Investig. 2011;29:676–82. [DOI] [PubMed] [Google Scholar]
  • 198.Davies G, Rmali K, Watkins G, Mansel R, Mason M, Jiang W. Elevated levels of tumour endothelial marker-8 in human breast cancer and its clinical significance. Int J Oncol. 2006;29:1311–7. [PubMed]
  • 199.Petrovic K, Robinson J, Whitworth K, Jinks E, Shaaban A, Lee SP. TEM8/ANTXR1-specific CAR T cells mediate toxicity in vivo. PLOS ONE. 2019;14:e0224015–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Schnell O, Krebs B, Wagner E, Romagna A, Beer AJ, Grau SJ, et al. Expression of Integrin αvβ3in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Pathol. 2008;18:378–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Wallstabe L, Mades A, Frenz S, Einsele H, Rader C, Hudecek M. CAR T cells targeting αvβ3 integrin are effective against advanced cancer in preclinical models. Adv Cell Gene Ther. 2018;1:e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Cobb DA, Rossi J, de, Liu L, An E. Targeting of the alphav beta3 integrin complex by CAR-T cells leads to rapid regression of diffuse intrinsic pontine glioma and glioblastoma. J Immunother Cancer. 2022;10:e003816–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Paes Dias M, Tripathi V, van der Heijden I, Cong K, Manolika EM, Bhin J, et al. Loss of nuclear DNA ligase III reverts PARP inhibitor resistance in BRCA1/53BP1 double-deficient cells by exposing ssDNA gaps. Mol Cell. 2021;81:4692–4708.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Oncogene are provided here courtesy of Nature Publishing Group

RESOURCES