Next Article in Journal
Electromagnetic Transduction Therapy (EMTT) Enhances Tenocyte Regenerative Potential: Evidence for Senolytic-like Effects and Matrix Remodeling
Next Article in Special Issue
PKC-ι Regulates an Oncogenic Positive Feedback Loop Between the MAPK/JNK Signaling Pathway, c-Jun/AP-1 and TNF-α in Breast Cancer
Previous Article in Journal
Unnatural Amino Acid Photo-Crosslinking Sheds Light on Gating of the Mechanosensitive Ion Channel OSCA1.2
Previous Article in Special Issue
Knockdown of Claudin-8 (CLDN8) Indicates a Link Between Breast Cancer Cell Sensitivity to Chemotherapeutics and Reveals a Potential Use of CLDN8 as a Molecular Diagnostic and Target for Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions

by
Fátima Postigo-Corrales
1,
Asunción Beltrán-Videla
2,
Antonio David Lázaro-Sánchez
3,
Ana María Hurtado
1,
Pablo Conesa-Zamora
1,2,*,
Ana Belén Arroyo
1,2,† and
Ginés Luengo-Gil
1,2,*,†
1
Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain
2
Group of Molecular Pathology and Pharmacogenetics, Pathology and Clinical Analysis Department, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, 30202 Cartagena, Spain
3
Department of Medical Oncology, Morales Meseguer General University Hospital, 30008 Murcia, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(15), 7119; https://doi.org/10.3390/ijms26157119
Submission received: 27 June 2025 / Revised: 21 July 2025 / Accepted: 22 July 2025 / Published: 23 July 2025
(This article belongs to the Special Issue Molecular Research and Cellular Biology of Breast Cancer)

Abstract

Docetaxel is a chemotherapeutic agent widely used for breast cancer treatment; however, its efficacy is often limited by drug resistance and associated toxicity. This review examines the molecular mechanisms of docetaxel resistance in breast cancer and discusses research advances and future directions for overcoming this challenge. Key resistance mechanisms include alterations in drug targets (microtubules), increased drug efflux, suppression of apoptosis, activation of survival signalling pathways, epithelial-to-mesenchymal transition (EMT), and cancer stem cell enrichment. An evolutionary perspective distinguishes between intrinsic and acquired resistance, emphasising the need for adaptive therapeutic strategies. Recent advances in genomic profiling, non-coding RNA research, novel drug combinations, and biomarker-guided therapies have also been reviewed. Emerging approaches, such as targeting the tumour microenvironment, harnessing immunotherapy, and implementing adaptive dosing schedules, have been discussed. This review emphasises the understanding of resistance as a multifactorial phenomenon that requires multipronged interventions. Research has aimed to identify predictive biomarkers, develop targeted agents to reverse resistance, and design rational combination strategies to improve patient outcomes. Progress in deciphering and targeting docetaxel resistance mechanisms holds promise for enhancing treatment responses and extending survival in patients with breast cancer.

1. Introduction

Breast cancer is the most frequently diagnosed malignancy in women worldwide, and it is a leading cause of cancer-related mortality. According to the most recent global estimates, breast cancer accounted for over 2.3 million new cases and more than 670,000 deaths annually in 2022 [1,2]. Chemotherapy has long been a cornerstone of treatment, particularly for aggressive and advanced breast cancers, and it remains the backbone of HER2-targeted therapies [3,4], and endocrine treatments [5] are often added. Among chemotherapeutic agents, taxanes, particularly docetaxel, play a central role in breast cancer management [6] (Figure 1). Docetaxel (Taxotere) is a highly lipophilic [7] semi-synthetic taxane that, in combination with other drugs, has significantly improved patient outcomes in both early-stage and metastatic breast cancer. For example, the addition of docetaxel to adjuvant regimens has yielded higher disease-free and overall survival rates in node-positive breast cancer patients than those for treatment using non-taxane regimens [8]. In the metastatic setting, the efficacy of docetaxel as a monotherapy was established in the pivotal 303 trial, which demonstrated superior response rates and time to progression compared to the results for doxorubicin [9]. Furthermore, docetaxel remains a key component of combination regimens, as in the CLEOPATRA trial, which showed improved outcomes with the addition of HER2-targeted agents [10]. These clinical successes have established docetaxel as a standard-of-care chemotherapeutic; it is frequently used either alone or in combination (e.g., with anthracyclines or platinum agents) for high-risk early breast cancer [8] and as a first-line treatment for HER2-positive recurrent or metastatic disease in combination with trastuzumab and pertuzumab [11]. In contrast, patients with luminal tumours are typically treated with endocrine therapy and CDK4/6 inhibitors, whereas single-agent paclitaxel is often preferred for triple-negative tumours. Combination chemotherapy, with the inclusion of docetaxel, is generally reserved for patients presenting with a high tumour burden or visceral crisis. In HER2-positive disease, the addition of docetaxel to HER2-targeted antibodies has shown synergistic effects, with the CLEOPATRA trial reporting a complete response rate of 12.7% and a partial response rate of 67.1% at week 9, which was associated with prolonged survival [10]. These outcomes were further validated in a Japanese cohort of patients with inoperable, recurrent, or advanced HER2-positive disease [12]. Despite its proven efficacy, the development of drug resistance is a major clinical challenge, which limits its long-term benefits [13,14,15,16].
Many breast tumours exhibit de novo (intrinsic) resistance and fail to respond to docetaxel from the outset. Even among tumours that initially respond, virtually all eventually develop acquired resistance under the selective pressure of therapy, leading to disease progression. Docetaxel resistance is associated with relapse after adjuvant chemotherapy and treatment failure in metastatic breast cancer, contributing to high mortality rates. Patients with docetaxel-resistant tumours often have limited alternative options, since cross-resistance to other chemotherapeutics is common, and resistant metastatic disease is usually incurable [15,17,18,19,20,21,22,23,24,25,26]. Docetaxel resistance is correlated with poor clinical outcomes and shorter survival [27]. Thus, understanding and overcoming resistance is critical. Elucidating the resistance mechanisms is crucial for the development of predictive biomarkers and novel therapeutic strategies to enhance patient outcomes. Table 1 summarises the predictive biomarkers of docetaxel resistance in breast cancer as obtained from recent preclinical and clinical studies.
Triple-negative breast cancer (TNBC), which lacks actionable targets, often shows poor and heterogeneous responses to taxane-based chemotherapy, partly because of the prevalence of resistance-associated biomarkers [19]. High-grade tumours may initially shrink with docetaxel treatment but then recur due to drug-resistant residual cells. Even in hormone receptor-positive cancers, docetaxel is often employed for endocrine-resistant diseases; however, cancers that have progressed despite endocrine therapy may harbour molecular alterations that confer resistance to chemotherapy [41]. Breast cancer comprises biologically distinct subtypes (luminal A/B, HER2-enriched, and triple-negative) that exhibit variable responses to docetaxel and distinct resistance mechanisms. For instance, one study found that ER-positive low-grade cell lines are more docetaxel-sensitive than are mesenchymal TNBC cell lines, which tend to exhibit higher innate resistance [22]. Clinically, this translates to variable response rates and highlights the need for biomarkers (such as tumour expression of particular genes) to predict who will benefit from docetaxel. Indeed, efforts have been made to identify such biomarkers; for example, overexpression of class III β-tubulin (encoded by TUBB3) or anti-apoptotic proteins in tumours has been linked to poor responses (as discussed later).
In summary, docetaxel is an important drug for breast cancer therapy, but its efficacy is curtailed by both intrinsic and acquired resistance [42]. This clinical problem has motivated extensive research on the biological underpinnings of drug resistance and potential solutions. The following sections discuss the molecular action of docetaxel, the known mechanisms by which breast cancer cells evade its effects, the special role of the actin-bundling protein fascin in tumour aggressiveness and drug resistance, an evolutionary perspective on how resistance emerges, and current advances aimed at combating docetaxel resistance.

2. Molecular Mechanism of Action of Taxanes (Docetaxel)

Docetaxel is a semi-synthetic chemotherapeutic agent from the taxane family, originally derived from the Taxus species, that exerts its cytotoxic effect by disrupting microtubule dynamics essential for mitosis [43,44,45]. It binds specifically to a hydrophobic pocket on β-tubulin within microtubules [44], promoting tubulin polymerisation and preventing depolymerisation, thereby “locking” microtubules in a stabilised state [45]. This suppression of dynamic instability (Figure 2) impairs mitotic spindle assembly and chromosome alignment, leading to the activation of the spindle assembly checkpoint and cell cycle arrest at G2/M [46,47,48]. As a result, cells experience mitotic catastrophe and typically undergo apoptosis through the intrinsic mitochondrial pathway, which is characterised by the activation of caspase-9, caspase-3, caspase-7, and cleavage of PARP [49,50,51,52,53]. Docetaxel also induces the phosphorylation of the anti-apoptotic protein Bcl-2, thereby inhibiting its function and further promoting cell death [44]. These effects have been validated in breast cancer models, where taxane treatment induces apoptotic cascades unless counterbalanced by pro-survival signalling [22,54].
Clinically, the cytotoxic mechanism of docetaxel results in tumour shrinkage. However, the same cellular pathways that mediate its therapeutic effects, particularly apoptosis, are frequently dysregulated in cancer cells, allowing a subset of cells to evade DTX-induced death. Furthermore, the mechanism of action underlies the side effects of several drugs. For instance, peripheral neuropathy is linked to disruption of microtubule function in neuronal cells [55,56]. Therefore, a comprehensive understanding of the mode of action of docetaxel is essential for elucidating the mechanisms by which cancer cells develop resistance. Indeed, nearly all characterised resistance pathways ultimately interfere with or bypass the key effects of taxanes, including microtubule stabilisation, mitotic arrest, and the initiation of apoptosis [57].

3. Evolutionary Perspective: Intrinsic vs. Acquired Resistance in Cancer

Cancer can be conceptualised as an evolutionary process driven by natural selection. Tumourigenesis typically originates from a single progenitor cell that undergoes clonal expansion. However, as tumours progress, they accumulate genetic mutations, epigenetic modifications, and phenotypic variability. This intratumoural heterogeneity constitutes the substrate upon which Darwinian selection can act in combination with microenvironmental plasticity, a new concept which is part of modern extended evolutionary synthesis [58].
Exposure to chemotherapy, such as docetaxel, imposes a strong selective pressure on genetically diverse tumour populations. Sensitive cells are eliminated, while resistant subclones, whether pre-existing or adaptively induced, survive and expand, ultimately leading to treatment failure (Figure 3) [59]. This mirrors the resistance dynamics observed in infectious diseases and reflects the Darwinian model of selection within heterogeneous tumour ecosystems [59]. Intrinsic resistance arises from pre-existing traits, such as TP53 mutations, quiescent stem-like phenotypes, or high expression of efflux pumps and survival proteins (for example, PI3K/Akt activation; fascin overexpression) [60,61,62], and explains the poor initial response in some subtypes, such as TNBC [61,62].
Acquired resistance develops during therapy through selection of minor resistant subclones or via adaptive rewiring (e.g., β-tubulin mutations and ABC transporter upregulation) [63,64,65]. Genomic instability in cancer cells facilitates multiple escape routes; for example, PIK3CA activation or new TP53 mutations can emerge during treatment [53]. These traits may incur fitness costs; therefore, intermittent therapy (“chemo holidays”) can allow sensitive cells to regain dominance, a concept explored in adaptive therapy [59]. Clonal shifts observed through serial biopsies or ctDNAs support this dynamic [66].
Resistance mechanisms often exhibit pathway-plasticity. Blocking one survival route can activate compensatory signalling (for example, PI3K–MAPK) [67]. Thus, tumours behave as evolving ecosystems, with interclonal competition shaped by selective pressure and microenvironmental constraints [68,69]. Docetaxel-resistant clones frequently display mesenchymal traits, high efflux transporter expression, or quiescence [70], and these features may be intrinsically present or acquired upon exposure. Overall, resistance should be viewed not as binary but as a shifting evolutionary continuum.
In summary, intrinsic resistance represents a pre-existing escape mechanism, whereas acquired resistance reflects an adaptive response that evolves during treatment. Both arise through evolutionary selection processes. Recognising this evolutionary underpinning enables researchers and clinicians to better anticipate therapeutic resistance. It must be assumed that given sufficient time and selective pressure, cancer will evolve mechanisms to evade treatment [71]. Therefore, the clinical objective is to either delay the emergence of resistance or to effectively target resistant clones once they appear. In the following section, we review the molecular mechanisms by which breast cancer cells acquire resistance to docetaxel, many of which may operate in both intrinsic and acquired contexts.

4. Mechanisms of Docetaxel Resistance in Breast Cancer

Breast cancer cells employ a variety of mechanisms to evade the cytotoxic effects of docetaxel. These mechanisms can be broadly categorised into several groups, often overlapping and co-occurring within the same tumour. Key resistance mechanisms include (1) alterations in the target of the drug (microtubules), (2) increased drug efflux and decreased drug accumulation, (3) evasion of apoptosis and cell death, (4) activation of alternative survival signalling pathways and epithelial-to-mesenchymal transition (EMT), and (5) cancer stem cell features and tumour microenvironment influences (Figure 4).

4.1. Alterations in Tubulin and Microtubule Dynamics

Taxanes, such as docetaxel, stabilise microtubules by binding to β-tubulin, preventing depolymerisation and inducing mitotic arrest. Resistance frequently arises from alterations in tubulin composition or structure that impair drug binding [19,71,72]. Overexpression of class III β-tubulin (TUBB3) has been consistently linked to taxane resistance in several cancers, including breast cancer [73,74]. Resistant cells often exhibit higher TUBB3 levels and elevated docetaxel IC50 values; for instance, MDA-MB-231 cells express four-fold more TUBB3 than luminal MCF7 cells [22,73]. Additionally, point mutations in β-tubulin (for example, A185T, A248V, and R306C) may disrupt taxane–tubulin interactions without abolishing microtubule function [71,72,73].
Post-translational modifications (PTMs) such as tubulin acetylation or detyrosination can stabilise microtubules independently of taxanes and reduce drug efficacy. Similarly, microtubule-associated proteins (MAPs) such as Tau, MAP4, and stathmin modulate polymer dynamics and influence the taxane response. High Tau expression has been associated with resistance, particularly in ER-positive breast tumours [75,76,77,78], whereas stathmin overexpression enhances microtubule turnover and opposes taxane activity [79,80].
Chronic exposure to docetaxel may also lead to adaptive changes in spindle architecture and microtubule behaviour that enable resistant cells to bypass mitotic arrest [39,81]. These multifactorial alterations, including isotype switching, PTMs, and MAP deregulation, converge to reduce the cytotoxic effects of docetaxel.

4.2. Increased Drug Efflux and Decreased Intracellular Drug Accumulation

One of the most well-characterised mechanisms of docetaxel resistance is the active efflux of cytotoxic drugs via ATP-binding cassette (ABC) transporters [82]. Among them, P-glycoprotein (P-gp), encoded by ABCB1/MDR1, has been the most studied. Its overexpression reduces intracellular docetaxel levels, thereby limiting cytotoxic efficacy. In breast cancer, ABCB1 expression may be intrinsically elevated, particularly in aggressive subtypes such as TNBC, or induced by chemotherapeutic pressure [15,83].
Functional inhibition or genetic silencing of P-gp restores docetaxel sensitivity in resistant cell lines [83]. Bufalin, a bioactive compound used in traditional Chinese medicine, downregulates ABCB1 protein levels and ATPase activity, enhancing docetaxel retention and apoptosis in vitro and reducing tumour growth in xenograft models [83]. Clinically, elevated ABCB1 levels in tumour specimens correlate with a reduced taxane response and earlier relapse [83].
In addition to P-gp, other ABC transporters such as ABCC1 (MRP1), ABCC10 (MRP7), and ABCG2 (BCRP) also mediate taxane efflux [29,84,85]. Notably, ABCC10 overexpression decreases taxane sensitivity in breast cancer, whereas genetic ablation increases susceptibility [84,85]. Although hepatic CYP3A4 enzymes contribute to taxane metabolism, they play only a minor role in efflux-mediated resistance [86].
Several pharmacological inhibitors of P-gp have been tested, including verapamil [87] and more specific MDR1 antagonists [88]; however, their clinical application is limited by poor selectivity and systemic toxicity [89]. To overcome these limitations, new approaches include nanotechnological delivery platforms designed to bypass efflux, such as albumin-bound paclitaxel (nab-paclitaxel) and liposomal or micellar formulations of docetaxel [90,91,92].
In summary, multidrug efflux transporters, particularly P-gp, constitute a central mechanism of docetaxel resistance in breast cancer. Their assessment may inform treatment decisions, whereas pharmacological inhibition and drug delivery innovations hold promise for improving taxane efficacy.

4.3. Evasion of Apoptosis and Cell Death Programs

Docetaxel induces apoptosis, primarily via the intrinsic (mitochondrial) pathway through the activation of caspase-9, caspase-3/7, and cleavage of PARP [49,50]. However, chemoresistant breast cancer cells evade apoptosis by altering multiple molecular checkpoints. A key mechanism involves overexpression of anti-apoptotic proteins, such as BCL-2, BCL-xL, MCL-1, and IAP family members like XIAP and survivin, which inhibit caspase activation and mitochondrial membrane permeabilisation [73,93,94,95,96]. The co-expression of survivin and TUBB3 has been associated with a reduced response to docetaxel [73], and XIAP or Livin is upregulated in fascin-positive, chemoresistant cells [62,93].
Apoptosis can also be suppressed by the inactivation of pro-apoptotic factors. TP53 mutations, which are frequent in triple-negative and HER2-positive breast cancers, reduce apoptotic priming and contribute to resistance, despite taxanes being partly effective in a p53-independent manner [94,95,97]. Downregulation of BAX, BAK, and executioner caspases also impair cell death. Moreover, fascin can promote resistance by activating the PI3K/Akt and NF-κB pathways, which suppresses pro-apoptotic proteins and increases XIAP expression [49]. These pathways are often constitutively active in resistant cells, shifting the balance towards survival [93].
Mitotic slippage is another evasion mechanism by which cells escape prolonged mitotic arrest without dying, resulting in tetraploidy or senescence [49,71,98,99]. This process is facilitated by weakened spindle checkpoint signalling and the concurrent activation of survival pathways.
Clinically, tumours with high BCL-2 expression show poor pathological responses to chemotherapy, and combinations of docetaxel with BCL-2 inhibitors, such as venetoclax, are under investigation in triple-negative breast cancer [96,97]. These findings support the rational combination of cytotoxic agents with drugs targeting apoptotic checkpoints to restore cell death and overcome resistance.

4.4. Activation of Survival Pathways and EMT

Under therapeutic pressure, cancer cells activate prosurvival and developmental pathways that sustain growth and confer chemoresistance. In breast cancer, several interconnected cascades contribute to docetaxel resistance.
The PI3K/Akt pathway is one of the most frequently upregulated genes driven by mechanisms such as PTEN loss, PIK3CA mutations, and growth factor receptor activation (for example, IGF-1R; HER2) [22,100,101,102]. Akt promotes cell survival and cell cycle progression, and its phosphorylation is elevated in docetaxel-resistant breast cancer cells. In tamoxifen-resistant MCF7 models, increased Akt activity correlates with enhanced docetaxel tolerance [23,89]. Similarly, the RAS–RAF–MEK–ERK (MAPK) cascade promotes proliferation and has been linked to taxane resistance, particularly in tumours with HER2 overexpression or RAS activation. EGF stimulation of MAPK signalling induces EMT and fascin expression, thereby enhancing migration and resistance [103,104].
NF-κB, a transcription factor that regulates survival and inflammation, is constitutively active in DTX-resistant breast cancer cells. It promotes resistance by upregulating anti-apoptotic proteins (BCL-xL and XIAP) and EMT-related effectors (MMP-2, MMP-9, and uPA) [105,106]. Fascin activates NF-κB, forming a positive feedback loop that enhances cell survival and invasiveness [103,105]. These signals may be triggered by cytokines from the tumour microenvironment (for example, IL-6) or via IKK-mediated intracellular activation, linking extrinsic and intrinsic resistance mechanisms. Notably, pharmacological inhibition of NF-κB using compounds such as SN-50 or baicalin restores docetaxel sensitivity in resistant breast cancer models, both in vitro and in vivo [107,108]. These findings underscore the potential of NF-κB as a therapeutic target for overcoming chemoresistance.
EMT imparts intrinsic resistance by promoting a mesenchymal, stem cell-like phenotype associated with slow proliferation, ABC transporter upregulation, and anti-apoptotic gene expression [109,110,111]. EMT markers include E-cadherin loss and upregulation of vimentin, N-cadherin, ZEB1/2, and Snail and Twist [110]. The induction of EMT by TGF-β or EMT-TFs reduces docetaxel sensitivity, whereas EMT reversal restores chemosensitivity [111,112,113].
Non-coding RNAs modulate EMT and resistance. In lung cancer, lncRNA linc-ROR sponges miR-145, derepressing fascin and promoting EMT and resistance, which is reversible upon knockdown [114]. In breast cancer, the loss of miR-200c allows ZEB1-driven EMT and chemoresistance, while restoring miR-200c sensitises cells to docetaxel [115,116]. Additionally, miR-141 has been implicated in resistance via the regulation of EIF4E and EMT/MAPK signalling [117].
The Notch pathway promotes EMT and cancer stemness. Inhibition of Notch1/4 using γ-secretase inhibitors (e.g., PF-03084014) reverses EMT; downregulates Snail, Slug, and ABCB1; and enhances docetaxel-induced apoptosis in TNBC models [110]. Notch also induces ZEB1, N-cadherin, and fascin expression, and evidence from pancreatic cancer suggests that a Notch → Akt → fascin axis likely operates in breast cancer [118].
In summary, docetaxel-resistant breast cancer cells activate the pro-survival (PI3K/Akt, MAPK, NF-κB, and Notch) and EMT pathways to bypass cytotoxic stress. EMT imparts stemness, drug efflux, and apoptotic resistance, whereas fascin amplifies these phenotypes. These findings support combination therapies targeting both taxane vulnerability and resistance networks, such as AKT inhibitors (ipatasertib) and mTOR inhibitors (everolimus), especially in tumours with PIK3CA mutations.

4.5. Cancer Stem Cells and Tumour Heterogeneity

Cancer stem cells (CSCs) constitute a subpopulation of breast tumours that are characterised by self-renewal, quiescence, and resistance to therapy. They are commonly identified by the CD44+/CD24 phenotype and high ALDH activity, and are typically spared by chemotherapy, allowing tumour repopulation [119]. Exposure to docetaxel can enrich CSC-like cells, consistent with selective survival under cytotoxic pressure [119].
Fascin (FSCN1), which is markedly upregulated in triple-negative breast cancer (TNBC) [120], promotes CSC traits by activating the Notch and β-catenin pathways [119,121]. Fascin overexpression enhances mammosphere formation and resistance, while its silencing reduces the CD44+/CD24 fraction, impairs stem-like features, and sensitises tumours to docetaxel [119,122]. These findings highlight fascin as a convergence point linking EMT, stemness, and resistance.
The tumour microenvironment (TME) further contributes to docetaxel resistance. Hypoxic niches promote EMT, slow proliferation, and induce HIF-1α, which upregulates both MDR1 and FSCN1 [103,123,124,125]. Stromal components, such as cancer-associated fibroblasts and tumour-associated macrophages, secrete IL-6 and activate the STAT3 and NF-κB pathways in cancer cells [105,126]. IL-6/STAT3 signalling has also been shown to increase fascin expression, linking microenvironmental cues to intrinsic resistance mechanisms [103,127].
In summary, docetaxel resistance in breast cancer arises not only from tumour-intrinsic changes, but also from CSC enrichment and TME-mediated adaptation. Therapeutic strategies targeting CSCs (e.g., Notch or Wnt inhibitors) or modulation of the TME (for example, IL-6 or HIF-1α blockade) holds promise for enhancing the efficacy of taxane-based regimens.

5. Role of Fascin in Tumour Biology and Chemoresistance

Fascin, encoded by FSCN1, is an actin-bundling protein primarily expressed in dendritic cells under physiological conditions. In cancer, fascin is frequently upregulated and contributes to cellular motility and invasion by organising actin into tightly packed filopodia and invadopodia [62,128]. In breast cancer, high fascin expression correlates with aggressive histological features, triple-negative subtype, early metastasis, and reduced overall survival. Ghebeh et al. first reported that high fascin expression confers chemoresistance in breast cancer [62]. Fascin-positive tumours exhibited a poorer response to chemotherapy and reduced apoptosis compared to the results for their fascin-negative counterparts. Mechanistically, fascin enhanced PI3K/Akt activation and upregulated anti-apoptotic proteins such as XIAP and Livin, while reducing the levels of active caspase-3/9 and cleaved PARP, thereby suppressing apoptotic responses to docetaxel. A recent study in colorectal cancer demonstrated that a variant in the lncRNA CCAT1 (rs67085638) promotes paclitaxel resistance via miR-24-3p-mediated upregulation of fascin, which activates the PI3K/Akt pathway and suppresses apoptosis [129]. Fascin also modulates focal adhesion kinase (FAK) and NF-κB signalling, which promotes survival and invasive behaviour. In fascin-positive cells, increased FAK phosphorylation has been linked to downstream NF-κB activation and the transcription of pro-survival genes [122]. Moreover, fascin reinforces CSC maintenance through the Notch–integrin β1 axis and supports EMT phenotypes, contributing to both migratory potential and drug tolerance [119,121]. In docetaxel-resistant models of lung cancer, fascin was upregulated and mediated resistance via EMT; silencing fascin or restoring miR-145 expression re-sensitised cells to treatment [121]. Notably, fascin-driven resistance extends beyond breast cancer [130,131]. In cervical, prostate, and lung cancers, fascin expression has been associated with resistance to cisplatin, paclitaxel, and docetaxel, often through EMT-linked mechanisms or miRNA dysregulation [114,132,133].
Given its virtually absent expression in normal epithelia, fascin is a promising ther-apeutic target. Small-molecule fascin inhibitors such as the NP-G2-029/NP-G2-044 series have demonstrably reduced breast cancer cell invasion and metastasis, showing syner-gistic effects with paclitaxel-based chemotherapy in murine models [134,135,136]. Whether such compounds can reverse chemoresistance remains unclear.
Clinically, assessing fascin expression may offer prognostic value and help to stratify patients. Tumours with high fascin expression could benefit from more aggressive upfront therapy or inclusion in trials evaluating fascin inhibitors (for example, NP-G2-044), or PI3K/Akt pathway inhibitors. Two ongoing clinical trials (Clinical Trials: NCT05023486; EudraCT: 2021-001328-17), in which fascin inhibitors are being tested to treat different tumours, including breast cancer, will provide novel insights about mechanisms of reverting chemoresistance [135].
In summary, fascin integrates key hallmarks of cancer aggressiveness, including motility, stemness, survival, and drug resistance. Targeting this multifunctional protein may not only inhibit metastatic progression but also restore chemosensitivity, representing a dual-pronged therapeutic approach for breast cancer.

6. Current Research Advances and Future Directions

Research over the last several years has focused on unravelling the complexities of docetaxel resistance and finding ways to overcome it. Some recent advances and emerging insights include the following.

6.1. Genomic and Transcriptomic Profiling of Resistant Tumours

Modern sequencing techniques are applied to patients before and after chemotherapy to identify key genetic changes associated with resistance. These studies have reinforced the role of known players (e.g., TP53 mutations, PIK3CA pathway alterations, and MDR1 upregulation) and uncovered new candidates. For instance, 2023 integrative analyses identified a set of differentially expressed genes and non-coding RNAs in residual disease after neoadjuvant taxane therapy, suggesting new biomarkers of resistance [137]. There is also an interest in polyclonal resistance, in which different metastases or regions of a tumour employ distinct resistance mechanisms, complicating treatment.

6.2. Non-Coding RNAs (lncRNAs, miRNAs, and circRNAs)

The role of ncRNAs in chemoresistance has come to the forefront. Long non-coding RNAs (lncRNAs) can regulate gene expression at multiple levels, and have been implicated in modulating drug responses. A comprehensive 2023 review catalogued numerous lncRNAs involved in taxane resistance in breast cancer [44]. For example, LINC00680 was found to be upregulated in docetaxel-resistant breast cancer cells and promotes resistance by sponging miR-432 to upregulate TRIM14 (which affects NF-κB signalling)—this was noted as one mechanism by which lncRNAs contribute to taxane resistance [137,138]. Another lncRNA, ROR, connects to EMT and fascin via miR-145 sponging (although shown in lung cancer, similar principles apply) [114]. The loss of tumour-suppressive miRNAs (such as the miR-200 family or miR-145) or overexpression of oncogenic miRNAs (such as miR-21) has been associated with taxane resistance phenotypes. Restoration of certain miRNAs in cell models can re-sensitise cells to docetaxel by targeting the mRNAs of resistance-related genes.
Circular RNAs (circRNAs) are a novel class of non-coding RNA that have gained increasing attention. These covalently closed RNA loops often function as miRNA sponges. Recent studies have identified specific circRNAs that confer docetaxel resistance. For example, circABCB1, derived from the ABCB1 gene, has been shown to diminish sensitivity to docetaxel in breast cancer, presumably by sponging miRNAs that would normally downregulate ABCB1, thereby indirectly maintaining high P-gp levels [33]. Another circUBR5 was recently reported (2024) to promote ribosome biogenesis and docetaxel resistance in triple-negative breast cancer via the miR-340-5p/CMTM6/c-MYC axis [34]. In that study, circUBR5 acted as a sponge for miR-340-5p, leading to increased expression of CMTM6 and c-MYC, which drove protein synthesis and cell growth, ultimately rendering cells less susceptible to docetaxel-induced stress (faster proliferation can antagonise the cytostatic effect of the drug). Targeting circRNAs is still in its early stages, but these findings open up new potential molecular targets for reversing resistance.

6.3. Novel Therapeutic Agents and Combinations

There is ongoing research on drugs that can overcome the known resistance mechanisms or kill cells via alternative lethal pathways. Table 2, Table 3, Table 4, Table 5 and Table 6 summarise clinical trials incorporating docetaxel as part of the therapeutic intervention in breast cancer. Some avenues to explore include the following:
  • Next-generation taxanes: Analogues such as cabazitaxel (a semisynthetic taxane) have been developed to evade P-gp efflux and exhibit activity in docetaxel-resistant tumours (cabazitaxel is approved in prostate cancer after docetaxel failure). Trials on breast cancer are not ongoing, but cabazitaxel and other drugs (e.g., larotaxel) may offer options for taxane-resistant cases [139,140,141].
  • Tubulin inhibitors with different mechanisms: Epothilones (such as ixabepilone) also stabilise microtubules but can retain activity in some taxane-resistant tumours, particularly those overexpressing TUBB3, because epothilones bind β-tubulin at different sites. Ixabepilone has been tested in breast cancer and approved for use in certain resistant metastatic cases, highlighting how drugs in the same functional class can sometimes overcome specific resistance, such as P-gp (ixabepilone is a poor P-gp substrate) or β-tubulin alterations [142,143].
  • Targeted pathway inhibitors: Combining docetaxel with targeted inhibitors is a major research area. PI3K inhibitors (e.g., buparlisib and alpelisib) and AKT inhibitors have been used to overcome resistance mediated by the PI3K/Akt pathway [144]. mTOR inhibitors (everolimus) combined with taxanes have shown some synergy in preclinical models, and a clinical trial of everolimus with weekly paclitaxel showed improved pathological complete response in HER2-negative breast cancer, suggesting that a similar strategy might improve docetaxel efficacy [145]. Notch pathway inhibitors (gamma secretase inhibitors) and hedgehog inhibitors are being studied to deplete cancer stem cells and potentially improve the chemo response [146,147]. There is also interest in inhibiting anti-apoptotic proteins; for example, a phase I trial combining venetoclax (BCL-2 inhibitor) with pegylated liposomal doxorubicin in TNBC could be envisioned with docetaxel, if the preclinical rationale is strong.
  • Immunotherapy combinations: While not directly reversing a resistance mechanism in the classical sense, the use of immunotherapy (such as checkpoint inhibitors) with chemotherapy can provide an alternative way to kill tumour cells by harnessing the immune system. In TNBC, the addition of atezolizumab (anti-PD-L1) to nab-paclitaxel improves outcomes in PD-L1+ patients. Trials have added checkpoint inhibitors to docetaxel. The idea is that even if some cells resist the direct effect of docetaxel, they may become more immunogenic (chemotherapy can cause immunogenic cell death in some cases), and immunotherapy can then eliminate those cells. In addition, chemotherapy may modulate the immune environment to make immunotherapy more effective. This multipronged kill approach may circumvent individual cellular resistance mechanisms.
Table 2. Clinical trials in early breast cancer HR+ Her2—recruiting, active, not recruiting—with docetaxel interventions.
Table 2. Clinical trials in early breast cancer HR+ Her2—recruiting, active, not recruiting—with docetaxel interventions.
NTC RegistryStudy TitlePhaseInterventionsDesignStart DateReferences
NCT05165225Phase II Neoadjuvant Pyrotinib Combined with Neoadjuvant Chemotherapy in HER2-Low-Expressing and HR Positive Early or Locally Advanced Breast Cancer: a Single-Arm, Non-Randomized, Single-Center, Open Label Trial2pyrotinib + epirubicin and cyclophosphamide followed by docetaxelNA; single group13 July 2021[148]
NCT04293393Neoadjuvant Study Chemotherapy vs. Letrozole + Abemaciclib in HR+/HER2− High/Intermediate Risk Breast Cancer Patients2doxorubicin + cyclophosphamide + docetaxel vs. letrozole +abermaciclib +/− LHRHRCT; open label1 October 2020[149]
NCT03201861Addition of Cisplatin to Adjuvant Chemotherapy for Early-Stage Breast Cancer in High-Risk Women3wpirubicin + cyclophospamide to docetaxel or paclitaxel vs. paclitaxel + cisplatinRCT; open label27 July 2017[150]
NCT06107673Dalpiciclib Plus AI (Neoadjuvant Endocrine Therapy) Compared with Neoadjuvant Chemotherapy in Early Breast Cancer (EBC)2NACT-dalpiciclib vs. ciclophosphamide + docetaxelRCT; masking triple30 September 2023[151]
NACT: neoadjuvant chemotherapy; RCT: randomized controlled trial; NA: no allocation.
Table 3. Clinical trials in breast cancer HR+ Her2—recruiting, active, not recruiting—with docetaxel interventions.
Table 3. Clinical trials in breast cancer HR+ Her2—recruiting, active, not recruiting—with docetaxel interventions.
NTC RegistryStudy TitlePhaseInterventionsDesignStart DateReferences
NCT06009627Study of Neoadjuvant Endocrine Therapy in HR Positive and HER2 Negative Premenopausal Breast Cancer Patients2/3darxil + exenestane + goserelin vs. docetaxel + doxorubicin + cyclophosphamideR; open label11 April 2023[152]
NCT03701334A Trial to Evaluate Efficacy and Safety of Ribociclib with Endocrine Therapy as Adjuvant Treatment in Patients With HR+/HER2- Early Breast Cancer (NATALEE)3ribociclib + endocrine therapy (ET) vs. ETRCT; open label10 October 2018[153]
NCT06375707Efficacy and Safety of Ribociclib in Combination with NSAI vs. Physician’s Choice of Chemotherapy Sequential Endocrine Therapy in HR+/HER2− Advanced Breast Cancer2docetaxel or paclitaxel + vinorelbine + capecitabine: sequential ribociclib: 600mg/d, 3 weeks continuous oral withdrawal for 1-week NSAI: anastrozole 1mg, 1 time/d, oral or letrozole: 2.5mg, 1 time/d, oralRCT; open label9 January 2024N/A
NCT04872985Pyrotinib in Combination with Neoadjuvant Chemotherapy in HR+/HER2-, HER4 High Expression Breast Cancer Patients: a Phase II Trial2pyrotinib + doxorubicin/epirubicin + cyclophosphamide followed by docetaxel/nab-paclitaxelRCT; DB20 April 2021[148]
NCT05296746Neoadjuvant and Adjuvant Ribociclib and ET for Clinically High-Risk ER+ and HER2- Breast Cancer2NACT–ACT ribociclib + letrozole
(responder) vs. (non-responder
NACT: ribociclib + letrozole
ACT—Arm1: doxorubicin + cyclophosphamide + docetaxel;
Arm2: docetaxel + cyclophosphamide;
Arm3: paclitaxel + doxorubicin + cyclophosphamide. All patients receive ribociclib + letrozole or AI
NR; parallel assigned3 May 2022[154]
DB: double-blind; NACT: neoadjuvant chemotherapy; ACT: adjuvant chemotherapy; NR: non-randomized; RCT: randomized controlled trial.
Table 4. Clinical trials in breast cancer HR+/- Her2+—recruiting, active, not recruiting—with docetaxel interventions.
Table 4. Clinical trials in breast cancer HR+/- Her2+—recruiting, active, not recruiting—with docetaxel interventions.
NTC RegistryStudy TitlePhaseInterventionsDesignStart DateReferences
NCT05638594Pyrotinib Combined with Trastuzumab, Dalpiciclib, Letrozole vs. TCbHP (Trastuzumab Plus Pertuzumab with Docetaxel and Carboplatin) as Neoadjuvant Treatment in HR +/HER2 + Breast Cancer2pyrotinib + trastuzumab + dalpiciclib + letrozole vs. trastuzumab + pertuzumab + docetaxel + carboplatinRCT; open label20 December 2022[155]
NCT05346224A Study to Evaluate the Efficacy and Safety of HLX11 vs. EU-Perjeta® in the Neoadjuvant Therapy of HER2-Positive and HR-Negative Early-Stage or Locally Advanced Breast Cancer3NACT: HLX11 + trastuzumab docetaxel; ACT: doxorubicin + cyclophosphamide + trastuzumab +HLX11 vs.
NACT: EU-Perjeta® + trastuzumab docetaxel; ACT: doxorubicin + cyclophosphamide + trastuzumab + EU-Perjeta®
RCT; open label17 April 2022[156]
NCT05319873Ribociclib, Tucatinib, and Trastuzumab for the Treatment of HER2 Positive Breast Cancer1/2Phase 1b; ribociclib + tucatinib + trastuzumab, if no progression diseases or unacceptable toxicity, allowed to Phase 2 Arm A: ribociclib + tucatinib + trastuzumab + fulvestrant;
Arm B: docetaxel + carboplatin + trastuzumab;
Arm C: ribociclib + tucatinib + trastuzumab
RCT; sequential assignment7 April 2022[157]
NCT05900206Trastuzumab Deruxtecan vs. Standard Neoadjuvant Treatment for HER2-Positive Breast Cancer2Arm 1: trastuzumab deruxtecan;
Arm 2: docetaxel/paclitaxel + carboplatin + trastuzumab + pertuzumab;
Arm 3: (ER + and luminal) ribociclib + letrozole;
Arm 4: (ER- and luminal)
epirubicin vs. cyclosporine
Arm 5: trastuzumab deruxtecan or docetaxel/paclitaxel + carboplatin + trastuzumab + pertuzumab
RCT; parallel assigment; open label 26 October 2023Link
NCT06770296The Dosing Regimen of Pyrotinib in HER2-Positive Advanced First-Line Breast Cancer: a Phase I Clinical Study1pyrotinib low dose + trastuzumab + docetaxel vs. pyrotinib normal dose + trastuzumab + docetaxelRCT; sequential assignment; open label1 November 2024N/A
NCT05704829NeoAdjuvant Therapy With Trastuzumab-deruxtecan vs. Chemotherapy+Trastuzumab+Pertuzumab in HER2+ Early Breast Cancer2T-DXd iv NACT-ACT vs. pacli-/docetacel + carboplatin + trastuzumab + pertuzumabRCT; crossover assignment; open label5 February 2024[158]
NCT05720026Study to Evaluate the Efficacy and Safety of SYSA1901 vs. Perjeta® of HER2-Positive Breast Cancer3SYSA1901 + trastuzumab + docetaxel vs. Perjeta® + trastuzumab + docetaxelRCT; DB; parallel-controlled9 January 2023N/A
NCT06278870Disitamab Vedotin + Pyrotinib vs. THP in the First-Line Treatment for HER2+ Advanced Breast Cancer Clinical Trial3disitamab vedotin + pyrotinib + trastuzumab vs.
trastuzumab + pertuzumab + docetaxel/paclitaxel/paclitaxel liposomal/paclitaxel alb.
RCT; quadruple; parallel assignment30 June 2023[159]
NCT06747338A Phase III Study of KN026 in Combination with HB1801 ± Carboplatin as Neoadjuvant Treatment for Early or Locally Advanced HER2-Positive Breast Cancer3KN026 + HB1801 + carboplatin vs. trastuzumab + pertuzumab + docetaxel + carboplatinRCT; parallel assignment; open label16 December 2024[160]
NCT06038539Efficacy and Safety of the Proposed Biosimilar Pertuzumab (PERT-IJS) vs. EU-Perjeta® Along with Trastuzumab and Chemotherapy (Carboplatin and Docetaxel) as Neoadjuvant Treatment in Chemotherapy naïve Patients with Early Stage or Locally Advanced HR Negative and HER2 Positive Breast Cancer3PERT-IJS + trastuzumab + carboplatin and docetaxel vs. EU-Perjeta® + trastuzumab + carboplatin and docetaxelRCT; DB; parallel assignment31 January 2025[161]
DB: double-blind; RCT: randomized controlled trial; NACT: neoadjuvant chemotherapy; ACT: adjuvant chemotherapy.
Table 5. Clinical trials in metastatic breast cancer HR+/- Her2+—recruiting, active, not recruiting—with docetaxel interventions.
Table 5. Clinical trials in metastatic breast cancer HR+/- Her2+—recruiting, active, not recruiting—with docetaxel interventions.
NTC RegistryStudy TitlePhaseInterventionsDesignStart DateReferences
NCT04760431TKIs vs. Pertuzumab in HER2+ Breast Cancer Patients with Active Brain Metastases (HER2BRAIN)2trastuzumab docetaxel pyrotinib vs. trastuzumab + docetaxel + pertuzumabRCT; 1:125 January 2021N/A
NCT05621434A Study to Evaluate Inetetamab + Pyrotinib + Chemotherapy in Previously Untreated HER2-Positive Metastatic Breast Cancer2inetetamab + pyrotinib and (taxane, vinorelbine, capecitabine, eribulin, and other agents (physicians choices).NR10 December 2022[162]
NCT05296798A Study to Evaluate the Efficacy and Safety of Giredestrant in Combination with Phesgo (Pertuzumab, Trastuzumab, and Hyaluronidase-zzxf) vs. Phesgo in Participants with Locally Advanced or Metastatic Breast Cancer (heredERA Breast Cancer)3Induction: giredestrant + pertuzumab + trastuzumab + taxane. Maintenance Arm A: giredestrant + pertuzumab + trastuzumab or Arm B: giredestrant + pertuzumab + trastuzumab + ETRCT; open label18 July 2022[163]
NCT06057610A Phase III Study of SHR-A1811 Injection with or Without Pertuzumab in HER2-Positive Recurrent or Metastatic Breast Cancer3A: SHR-A1811;
B: SHR-A1811 + pertuzumab;
C: trastuzumab + pertuzumab + docetaxel
RCT; open label16 October 2023[164]
NCT05698186Thero2-01S22 in HER2-Positive Breast Cancer3thero2-01S22/placebo + docetaxel or vinorelbine + pertuzumab + trastuzumabRCT; DB; placebo-controlled15 May 2023N/A
NCT06135714Metastasis-Directed Therapy for Oligometastases of Breast Cancer3Luminal BC: CDK4/6 inhibitors + ET;
HER2+: trastuzumab + pertuzumab + docetaxel;
TNBC: immune checkpoint inhibitors expressing PD-L1;
Arm A continues systemic chemotherapy alone; Arm B followed the same treatment.
RCT; parallel assignment; open label11 August 2023[165]
NCT06439693The SAPPHO Study: Sequential Therapy with Curative Intent in de Novo HER2+ Metastatic Breast Cancer2taxane + trastuzumab + pertuzumab followed by trastuzumab deruxtecan, followed by tucatinib + ado-trastuzumab emtansine, followed by trastuzumab + pertuzumab + tucatinibNA; single group sequential treatment8 August 2024N/A
NCT06445400A Study of BL-M07D1, BL-M07D1+Pertuzumab and BL-M07D1+Pertuzumab+Docetaxel in Patients with Unresectable Locally Advanced or Metastatic HER2-Positive Breast Cancer2Arm A: BL-M07D1 + pertuzumab;
Arm B: BL-M07D1 + pertuzumab + docetaxel
NA; single group; open label19 June 2024N/A
NCT07003074A Clinical Study of TQB2102 vs. Docetaxel Plus Trastuzumab and Pertuzumab in the Treatment of HER2 Positive Recurrent or Metastatic Breast Cancer3Arm A: TQB2102;
Arm B: docetaxel + trastuzumab + pertuzumab
RCT; open label; parallel-controlledAugust 2025[166]
ET: endocrine therapy; DB: double-blind; NR: non-randomized; RCT: randomized controlled trial; NA: no allocation.
Table 6. Clinical trials in triple negative breast cancer (TNBC)—recruiting, active, not recruiting—with docetaxel interventions.
Table 6. Clinical trials in triple negative breast cancer (TNBC)—recruiting, active, not recruiting—with docetaxel interventions.
NTC RegistryStudy TitlePhaseInterventionsDesignStart DateReferences
NCT04836156Neoadjuvant Therapy Study Guided by Drug Screening in Vitro for Human Epidermal Growth Factor Receptor 2 (HER2) Negative Early Breast Cancer Patients1/2docetaxel + carboplatin
docetaxel + epirubicin
NR; single group assignment; open label2 April 2021[167]
NCT05475678Clinical Study of Camrelizumab Combined with TCb vs. TCb in Neoadjuvant Treatment of Triple-Negative Breast Cancer2carrelizumab + docetaxel + carboplatin vs. docetaxel + carboplatinRCT; parallel assignment; open label19 July 2022[168]
NCT05645380Neoadjuvant TIL- and Response-Adapted Chemoimmunotherapy for TNBC2Arm A: carboplatin + docetaxel + pembrolizumab;
Arm B: carboplatin + docetaxel + doxorubicin + cyclophosphamide + pembrolizumab
NR; parallel assignment; open label5 December 2022[169]
NCT04947189Seviteronel in Combination with Chemotherapy in Androgen-receptor Positive Metastatic Triple-Negative Breast Cancer1/2seviteronel + dexamethasone + docetaxelNR; single group; open label1 November 2021[170]
NCT05076760MEM-288 Oncolytic Virus Alone and in Combination with Standard of Care Therapy in Advanced Solid Tumours1MEM-288 vs. nivolumab + docetaxelNR; single group; open label21 April 2022[171]
NCT05929768Shorter Anthracycline-Free Chemo Immunotherapy Adapted to Pathological Response in Early Triple Negative Breast Cancer (SCARLET), A Randomized Phase III Study3paclitaxel + carboplatin + pembrolizumab, followed by doxorubicin + cyclophosphamide + pembrolizumab;
ACT: pembrolizumab vs.
docetaxel + carboplatin + pembrolizumab;
ACT: pembrolizumab
RCT; parallel assignment; open label15 September 2023N/A
NCT05978648Trilaciclib in Patients with Early-Stage HR-Negative Breast Cancer Receiving Adjuvant Chemotherapy2trilaciclib + epirubicin + cyclophosphamide + paclitaxelNR; single group; open label20 September 2023N/A
NCT06225284Neoadjuvant Chemotherapy with or Without GnRH Agonist for Premenopausal Triple-negative Early Breast Cancer Patients2GnRH: goserelin or leuprolide or triptorelin + anthracycline + cyclophosphamide, followed by taxane and optional pembrolizumab vs. anthracycline + cyclophosphamide, followed by taxane and optional pembrolizumabRCT; parallel assignment; open label22 August 2024N/A
NCT06795503Non-Inferiority Study on MRNA-lncRNA Model in Low-Risk Triple-Negative Breast Cancer Patients3docetaxel + cyclophosphamide vs. epirubicin + cyclophosphamide, followed by paclitaxelRCT; parallel assignment; open label27 January 2025N/A
RCT: randomized controlled trial; ACT: adjuvant chemotherapy.

6.4. Adaptive Therapy and Dosing Strategies

Inspired by evolutionary principles, researchers have tested non-traditional dosing schedules to manage resistance. Instead of the maximum tolerated dose (MTD) given on a fixed schedule, adaptive therapy adjusts the dosing based on tumour response to maintain a stable tumour burden and keep sensitive cells alive to suppress resistant cells [59]. For example, an adaptive regimen might provide docetaxel until a certain shrinkage is achieved, then stop or lower doses to allow sensitive cells to recover and outnumber resistant cells, and then resume treatment. This method has shown promise in prostate cancer models with hormone therapy, and trials for other cancers are in progress. Such strategies for breast cancer would be highly experimental; however, mathematically, they could delay the emergence of dominant–resistant populations. Additionally, dose-dense schedules (providing the same total dose in a shorter interval) have been tested clinically to outpace resistance. In early breast cancer, dose-dense taxane schedules improved survival in some studies, potentially by reducing the regrowth of resistant clones between cycles.

6.5. Biomarker-Guided Therapy

There has been a push towards personalised chemotherapy. Rather than assuming that all patients receive the same regimen, new trials should incorporate biomarkers of likely resistance to decide on therapy. For example, patients whose tumours show high TUBB3 or high MDR1 might be initially triaged away from docetaxel to an alternative (maybe to an epothilone or a platinum, etc.), or an added agent might be administered to counteract the specific resistance (e.g., a P-gp inhibitor in a clinical trial context or a PI3K inhibitor for a PIK3CA-mutant, Akt-activated tumour). Tests such as Oncotype DX currently guide the use of chemotherapy in general; future refinements may guide the selection of the chemotherapy type or treatment pairings. Researchers are validating assays for measuring expression of resistance-related genes (like an “MDR1 score” or a “stemness signature”) to predict chemo response. Interesting biomarkers include circulating tumour cells (CTCs) and circulating tumour DNA. A high burden of CTCs after a few cycles of chemotherapy can indicate early resistance, prompting the switch to alternate therapies before clinical progression.

6.6. Targeting the Tumour Microenvironment

Because the environment contributes to resistance (e.g., fibrosis-limiting drug delivery and hypoxia-inducing resistance genes), some approaches aim to modify the microenvironment. For instance, trials on TGF-β inhibitors (to prevent fibrosis/EMT) and CXCR4 inhibitors (to disrupt protective niches) are ongoing. Normalising tumour vasculature with agents such as bevacizumab (anti-VEGF) might improve docetaxel drug delivery, although in practice, the benefit of bevacizumab in breast cancer has been debated. Nevertheless, understanding that resistance is not purely tumour cell-autonomous will lead to holistic strategies.

6.7. Emerging Drug Targets from Omics

High-throughput screening identifies novel mediators of resistance. A recent CRISPR knockout screen in breast cancer cells revealed that the loss of certain epigenetic regulators makes cells more sensitive to docetaxel, indicating that these regulators are potential drug targets. Similarly, metabolomic studies have shown that resistant cells exhibit distinct metabolic pathways (e.g., greater dependence on oxidative phosphorylation). Inhibiting mitochondrial respiration with drugs such as metformin or specific OXPHOS inhibitors can preferentially kill chemoresistant cells that rely on that pathway [121]. Fascin itself was identified through unbiased approaches as a top hit connecting metastasis and resistance [62], validating the approach.
In the realm of fascin and actin dynamics, small molecules that disrupt cytoskeletal adaptations in resistant cells have been actively investigated. Compounds that target actin regulators or mitotic spindle assembly checkpoints (such that resistant cells that slip through mitosis can be caught by different mechanisms) are currently being studied.

6.8. Clinical Rechallenge and Sequencing

For patients, a pragmatic question is whether a cancer becomes resistant to docetaxel or whether it is permanently resistant. Interestingly, some data suggest that after a “drug holiday”, tumours might regain sensitivity (if the resistant population wanes because of fitness costs). A small clinical study on docetaxel rechallenge in metastatic breast cancer showed that a subset of patients responded to the reintroduction of docetaxel after having progressed previously [172]. The responses were not as high as those of the initial therapy; however, this raises the point that resistance is dynamic. Clinicians are also exploring alternative therapies to maintain tumour off-balance (e.g., alternating taxanes with another agent every few cycles). Such strategies are still experimental but underscore the evolving mindset in the treatment of drug-resistant cancers.
In conclusion, the period 2020–2025 saw significant progress in decoding docetaxel resistance. We appreciate this as a multidimensional problem involving genetics, epigenetics, non-coding RNAs, cellular phenotypes (such as EMT/CSC), and microenvironmental factors. Combating resistance requires combination approaches that target the structure of cancer cells (microtubules), their survival signals, and their interactions with the environment. Trials are becoming increasingly biomarker-driven to test these combinations in patient subgroups. It is hoped that by integrating these new insights, future therapies will either prevent resistance from developing or convert resistant diseases into chronic, manageable conditions. Ongoing research, including clinical trials of novel agents and treatment strategies, will determine how closely this goal is achieved (Table 1, Table 2, Table 3, Table 4 and Table 5). As our understanding deepens, the outlook for overcoming docetaxel resistance in breast cancer appears cautiously optimistic, moving us closer to more durable responses and improved survival in patients facing this challenge.

6.9. Nanoparticle-Based Strategies to Overcome Taxane Resistance: Advances, Benefits, and Current Challenges

Nanoparticle-based drug delivery systems offer promising solutions to enhance taxane efficacy, reverse resistance, and reduce systemic toxicity. Compared to conventional formulations, nanoparticles improve solubility and eliminate the need for toxic solvents, such as Cremophor EL or polysorbate 80, which are associated with hypersensitivity and require corticosteroid premedication. Systems such as nab-paclitaxel [173] and RNA-based nanoassemblies [174] increase the aqueous solubility and improve pharmacokinetics. Table 7 provides a comparative overview of docetaxel delivery systems in breast cancer.
In order to overcome failures encountered in clinical trials, nanoformulations offer a promising approach, as they can bypass drug efflux pumps, improve cellular uptake, and co-deliver agents that modulate drug resistance pathways. For instance, folate-targeted paclitaxel–fisetin nanoparticles downregulated ABCG2 and triggered apoptosis in resistant ovarian cells [178], whereas NanoOrl, an orlistat-loaded formulation, restored taxane sensitivity via fatty acid synthase inhibition, independent of P-gp in prostate cancer [179]. Docetaxel–resveratrol micelles demonstrated synergistic cytotoxicity against MCF-7 breast cancer cells, enhanced drug uptake, and prolonged circulation time in vivo, compared with the results for free drugs [180,181], and docetaxel–curcumin nanoparticles increased uptake, ROS generation, and apoptosis in MCF-7/Adr cells [182].
Controlled-release formulations further prolong intratumoural drug exposure. Hybrid PLGA-lipid nanoparticles reduced paclitaxel IC50 > 300-fold in anoikis-resistant lung cancer cells [183], and rod-shaped PLGA nanoparticles loaded with docetaxel improved survival and tumour inhibition in taxane-resistant TNBC mouse models [184].
Despite these advances, most nanoformulations remain in the preclinical stage. The challenges include poor scalability, limited in vivo validation, and regulatory uncertainty. As Patel et al. noted, many in vitro models fail to recapitulate tumour complexity, limiting clinical translation [185]. Even approved systems such as nab-paclitaxel offer only modest clinical benefits and raise cost-effectiveness concerns [173].
Nevertheless, innovations such as stimuli-responsive nanoparticles, surface-functionalized systems, and biodegradable carriers have expanded the therapeutic potential of these platforms [186]. Figure 5 illustrates the key strategies by which targeted nanoparticles overcome resistance, and Table 8 and Table 9 compare delivery systems currently under investigation in preclinical and clinical trials, respectively.

7. Conclusions and Perspectives

Docetaxel has indisputably improved the outcomes of breast cancer therapy; however, the emergence of resistance remains a formidable hurdle that limits its full potential. Resistance to docetaxel is a multifactorial phenomenon; tumour cells evade the effects of the drug through alterations in the drug target (tubulin/microtubules), active efflux of the drug, suppression of apoptotic pathways, adoption of an EMT and stem-like state, and various genetic/epigenetic adaptations. The actin-bundling protein fascin exemplifies how a single molecular change can simultaneously drive tumour aggressiveness and therapy resistance, making it an attractive target for next-generation treatments. An evolutionary perspective sheds light on how resistance develops, why a one-dimensional treatment approach is often inadequate, and the adaptability of cancer cells to adjustable therapeutic strategies.
The battle against docetaxel resistance is being waged on multiple fronts. Advances in genomic and transcriptomic profiling have pinpointed new mediators of resistance, particularly in the non-coding RNA realm. Therapeutic innovations, from novel taxane analogues to combinations of chemotherapy with targeted inhibitors or immunotherapy, are under active investigation in outsmart-resistant cancer cells. Early results suggest that tackling resistance mechanisms (such as using a PI3K inhibitor to counter Akt-driven survival, or a drug like bufalin to inhibit P-gp) can restore docetaxel sensitivity in preclinical trials [83]. In parallel, clinical strategies, such as personalised treatment regimens based on tumour biomarkers and adaptive dosing schedules, are being explored to delay or prevent evolution of resistance in patients.
Therefore, further research is required. Key areas for future studies include the discovery of reliable biomarkers that predict resistance (so therapy can be tailored accordingly), the development of safe inhibitors of proteins, such as fascin or survivin, that cancer cells use to resist apoptosis, and the integration of mathematical oncology models to optimise treatment sequencing. Moreover, understanding how the tumour microenvironment and host immune system contribute to chemoresistance may open new avenues (for example, using agents to modulate the stroma or combining chemotherapy with immunotherapies to target residual resistant cells).
In our view, targeting fascin holds exceptional therapeutic promise, as it functions both as a marker of tumour aggressiveness and as a mechanistic driver of resistance. Furthermore, incorporating dynamic biomarker monitoring (e.g., via serial liquid biopsies or ctDNA) may enable real-time treatment adaptation in clinical settings. We also advocate greater integration of evolutionary principles, such as adaptive dosing or temporally modulated combinations, into trial design, which may delay or prevent the emergence of resistant subclones.
In writing this narrative review, we underscored not only established concepts but also the most recent (2020–2025) findings in this fast-evolving field. The goal for clinicians and researchers is to convert our growing mechanistic knowledge into effective therapies that prevent or overcome docetaxel resistance, thereby prolonging remission and saving lives. With multidisciplinary efforts spanning molecular biology, pharmacology, and clinical trials, there is legitimate hope that what is now common clinical frustration (taxane resistance) will be a more manageable or even reversible condition in the future. The story of docetaxel resistance in breast cancer, once a seemingly intractable problem, is steadily being rewritten with chapters on innovation and improved understanding, moving us closer to the day when we can outsmart cancer resilience.

Author Contributions

F.P.-C. and G.L.-G. jointly conceived the study. F.P.-C. and G.L.-G. designed the study. F.P.-C., A.B.-V., A.D.L.-S., A.M.H., P.C.-Z., A.B.A. and G.L.-G. drafted the manuscript. A.M.H., P.C.-Z., A.B.A. and G.L.-G. offered important intellectual input, provided critical resources or funding, and critically reviewed the study design. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded through the Project MEFASCINAS (Análisis de Eficacia de Terapias Anti Fascina para el Tratamiento del Cáncer de Mama Triple Negativo), by Ursus Investigaciones Agrupación de Interés Económico and its participants, under the call “Financiación Estructurada de Proyectos de I+D por Agrupaciones de Interés Económico (A.I.E.)”, in collaboration with INVENTIUM.

Acknowledgments

The authors acknowledge the staff of the Pathology Department at the Hospital General Universitario Santa Lucía for their support in carrying out this project. During the preparation of this work, the author(s) used chatGPT (https://www.chatgpt.com/) and Paperpal (https://www.paperpal.com/): AI Academic Writing Tool to improve the language, readability, and image generation. After using this tool/service, the author(s) reviewed and edited the content as needed and take full responsibility for the content of the publication. Figures created in BioRender (https://www.biorender.com/).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AKTProtein Kinase B
ALDHAldehyde Dehydrogenase
ATPAdenosine Triphosphate
BADBCL2 Associated Agonist of Cell Death
BAKBCL2 Antagonist/Killer
BAXBCL2 Associated X Protein
BCLB-Cell Lymphoma
BCRPBreast Cancer Resistance Protein
CRComplete Response
CRISPRClustered Regularly Interspaced Short Palindromic Repeats
CSCCancer Stem Cell
DNADeoxyribonucleic Acid
DTXDocetaxel
EGFEpidermal Growth Factor
EREstrogen Receptor
ERKExtracellular Signal-Regulated Kinase
HER2Human Epidermal Growth Factor Receptor 2
HIFHypoxia-Inducible Factor
IGFInsulin-like Growth Factor
IKKIκB Kinase
JAKJanus Kinase
MAPMicrotubule-Associated Protein
MAPKMitogen-Activated Protein Kinase
MEKMitogen-Activated Protein Kinase
MMPMatrix Metalloproteinase
MYCMyelocytomatosis Viral Oncogene
NFNuclear Factor
OXPHOSOxidative Phosphorylation
PARPPoly (ADP-Ribose) Polymerase
PDProgressive Disease
PFProgression-Free
PRProgesterone Receptor
PTENPhosphatase and Tensin Homolog
RAFRapidly Accelerated Fibrosarcoma
RASRat Sarcoma Viral Oncogene Homolog
RNARibonucleic Acid
RORRegulator of Reprogramming
SMACSecond Mitochondria-Derived Activator of Caspases
TGFTransforming Growth Factor
VEGFVascular Endothelial Growth Factor
XIAPX-linked Inhibitor of Apoptosis Protein

References

  1. Filho, A.M.; Laversanne, M.; Ferlay, J.; Colombet, M.; Piñeros, M.; Znaor, A.; Parkin, D.M.; Soerjomataram, I.; Bray, F. The GLOBOCAN 2022 cancer estimates: Data sources, methods, and a snapshot of the cancer burden worldwide. Int. J. Cancer 2024, 156, 1336–1346. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA. Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  3. Moja, L.; Tagliabue, L.; Balduzzi, S.; Parmelli, E.; Pistotti, V.; Guarneri, V.; D’Amico, R. Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst. Rev. 2012, 2021, CD006243. [Google Scholar] [CrossRef] [PubMed]
  4. Swain, S.M.; Shastry, M.; Hamilton, E. Targeting HER2-positive breast cancer: Advances and future directions. Nat. Rev. Drug Discov. 2023, 22, 101–126. [Google Scholar] [CrossRef] [PubMed]
  5. Bui, K.T.; Willson, M.L.; Goel, S.; Beith, J.; Goodwin, A. Ovarian suppression for adjuvant treatment of hormone receptor-positive early breast cancer. Cochrane Database Syst. Rev. 2020, 2020, CD013538. [Google Scholar] [CrossRef] [PubMed]
  6. Willson, M.L.; Burke, L.; Ferguson, T.; Ghersi, D.; Nowak, A.K.; Wilcken, N. Taxanes for adjuvant treatment of early breast cancer. Cochrane Database Syst. Rev. 2019, 2019, CD013538. [Google Scholar] [CrossRef] [PubMed]
  7. Aranda, E.; Teruel, J.A.; Ortiz, A.; Pérez-Cárceles, M.D.; Aranda, F.J. Interaction of Docetaxel with Phosphatidylcholine Membranes: A Combined Experimental and Computational Study. J. Membr. Biol. 2022, 255, 277–291. [Google Scholar] [CrossRef] [PubMed]
  8. Braybrooke, J.; Bradley, R.; Gray, R.; Hills, R.K.; Pan, H.; Peto, R.; Dodwell, D.; McGale, P.; Taylor, C.; Aihara, T.; et al. Anthracycline-containing and taxane-containing chemotherapy for early-stage operable breast cancer: A patient-level meta-analysis of 100,000 women from 86 randomised trials. Lancet 2023, 401, 1277–1292. [Google Scholar] [CrossRef] [PubMed]
  9. Chan, S.; Friedrichs, K.; Noel, D.; Pintér, T.; Van Belle, S.; Vorobiof, D.; Duarte, R.; Gil, M.G.; Bodrogi, I.; Murray, E.; et al. Prospective randomized trial of docetaxel versus doxorubicin in patients with metastatic breast cancer. J. Clin. Oncol. 1999, 17, 2341–2354. [Google Scholar] [CrossRef] [PubMed]
  10. Debien, V.; Agostinetto, E.; Bruzzone, M.; Ceppi, M.; Martins-Branco, D.; Molinelli, C.; Jacobs, F.; Nader-Marta, G.; Lambertini, M.; de Azambuja, E. The Impact of Initial Tumor Response on Survival Outcomes of Patients with HER2-Positive Advanced Breast Cancer Treated With Docetaxel, Trastuzumab, and Pertuzumab: An Exploratory Analysis of the CLEOPATRA Trial. Clin. Breast Cancer 2024, 24, 421–430.e3. [Google Scholar] [CrossRef] [PubMed]
  11. Nabholtz, J.M.; Senn, H.J.; Bezwoda, W.R.; Melnychuk, D.; Deschênes, L.; Douma, J.; Vandenberg, T.A.; Rapoport, B.; Rosso, R.; Trillet-Lenoir, V.; et al. Prospective randomized trial of docetaxel versus mitomycin plus vinblastine in patients with metastatic breast cancer progressing despite previous anthracycline-containing chemotherapy. J. Clin. Oncol. 1999, 17, 1413–1424. [Google Scholar] [CrossRef] [PubMed]
  12. Takahashi, M.; Ohtani, S.; Nagai, S.E.; Takashima, S.; Yamaguchi, M.; Tsuneizumi, M.; Komoike, Y.; Osako, T.; Ito, Y.; Ikeda, M.; et al. The efficacy and safety of pertuzumab plus trastuzumab and docetaxel as a first-line therapy in Japanese patients with inoperable or recurrent HER2-positive breast cancer: The COMACHI study. Breast Cancer Res. Treat. 2021, 185, 125–134. [Google Scholar] [CrossRef] [PubMed]
  13. Chang, J.C.; Wooten, E.C.; Tsimelzon, A.; Hilsenbeck, S.G.; Gutierrez, M.C.; Tham, Y.L.; Kalidas, M.; Elledge, R.; Mohsin, S.; Osborne, C.K.; et al. Patterns of resistance and incomplete response to docetaxel by gene expression profiling in breast cancer patients. J. Clin. Oncol. 2005, 23, 1169–1177. [Google Scholar] [CrossRef] [PubMed]
  14. Chang, J.C.; Wooten, E.C.; Tsimelzon, A.; Hilsenbeck, S.G.; Gutierrez, M.C.; Elledge, R.; Mohsin, S.; Osborne, C.K.; Chamness, G.C.; Allred, D.C.; et al. Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet 2003, 362, 362–369. [Google Scholar] [CrossRef] [PubMed]
  15. Hansen, S.N.; Westergaard, D.; Thomsen, M.B.H.; Vistesen, M.; Do, K.N.; Fogh, L.; Belling, K.C.; Wang, J.; Yang, H.; Gupta, R.; et al. Acquisition of docetaxel resistance in breast cancer cells reveals upregulation of ABCB1 expression as a key mediator of resistance accompanied by discrete upregulation of other specific genes and pathways. Tumor Biol. 2015, 36, 4327–4338. [Google Scholar] [CrossRef] [PubMed]
  16. Hansen, S.N.; Ehlers, N.S.; Zhu, S.; Thomsen, M.B.H.; Nielsen, R.L.; Liu, D.; Wang, G.; Hou, Y.; Zhang, X.; Xu, X.; et al. The stepwise evolution of the exome during acquisition of docetaxel resistance in breast cancer cells. BMC Genom. 2016, 17, 442. [Google Scholar] [CrossRef] [PubMed]
  17. Ingham, J.; Ruan, J.-L.; Coelho, M.A. Breaking barriers: We need a multidisciplinary approach to tackle cancer drug resistance. BJC Rep. 2025, 3, 11. [Google Scholar] [CrossRef] [PubMed]
  18. Khan, S.U.; Fatima, K.; Aisha, S.; Malik, F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun. Signal. 2024, 22, 109. [Google Scholar] [CrossRef] [PubMed]
  19. Murray, S.; Briasoulis, E.; Linardou, H.; Bafaloukos, D.; Papadimitriou, C. Taxane resistance in breast cancer: Mechanisms, predictive biomarkers and circumvention strategies. Cancer Treat. Rev. 2012, 38, 890–903. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, J.; Meng, H.; Zhang, M.; Zhang, C.; Huang, M.; Yan, C.; Wang, Z.; Hou, L.; Yang, L.; Ling, R. Regulation of docetaxel chemosensitivity by NR2F6 in breast cancer. Endocr. Relat. Cancer 2020, 27, 309–323. [Google Scholar] [CrossRef] [PubMed]
  21. Ma, J.; Wang, J.; Xu, T.; Ouyang, Q.; Wang, X.; Wang, J.; Gan, L.; Ouyang, Z.; Lin, D.; Sun, T.; et al. Efficacy and safety of KN026 and docetaxel for HER2-positive breast cancer: A phase II clinical trial. Cancer Commun. 2025, 45, 476–485. [Google Scholar] [CrossRef] [PubMed]
  22. Scherbakov, A.M.; Basharina, A.A.; Sorokin, D.V.; Mikhaevich, E.I.; Mizaeva, I.E.; Mikhaylova, A.L.; Bogush, T.A.; Krasil’nikov, M.A. Targeting hormone-resistant breast cancer cells with docetaxel: A look inside the resistance. Cancer Drug Resist. 2023, 6, 103–115. [Google Scholar] [CrossRef] [PubMed]
  23. Lim, W.; Hwang, I.; Zhang, J.; Chen, Z.; Han, J.; Jeon, J.; Koo, B.K.; Kim, S.; Lee, J.E.; Kim, Y.; et al. Exploration of drug resistance mechanisms in triple negative breast cancer cells using a microfluidic device and patient tissues. Elife 2024, 12, RP88830. [Google Scholar] [CrossRef] [PubMed]
  24. Imran, M.; Saleem, S.; Chaudhuri, A.; Ali, J.; Baboota, S. Docetaxel: An update on its molecular mechanisms, therapeutic trajectory and nanotechnology in the treatment of breast, lung and prostate cancer. J. Drug Deliv. Sci. Technol. 2020, 60, 101959. [Google Scholar] [CrossRef]
  25. Bin Emran, T.; Shahriar, A.; Mahmud, A.R.; Rahman, T.; Abir, M.H.; Siddiquee, M.F.R.; Ahmed, H.; Rahman, N.; Nainu, F.; Wahyudin, E.; et al. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front. Oncol. 2022, 12, 891652. [Google Scholar] [CrossRef] [PubMed]
  26. Kinnel, B.; Singh, S.K.; Oprea-Ilies, G.; Singh, R. Targeted Therapy and Mechanisms of Drug Resistance in Breast Cancer. Cancers 2023, 15, 1320. [Google Scholar] [CrossRef] [PubMed]
  27. Gomez-Miragaya, J.; Díaz-Navarro, A.; Tonda, R.; Beltran, S.; Palomero, L.; Palafox, M.; Dobrolecki, L.E.; Huang, C.; Vasaikar, S.; Zhang, B.; et al. Chromosome 12p amplification in triple-negative/BRCA1-mutated breast cancer associates with emergence of docetaxel resistance and carboplatin sensitivity. Cancer Res. 2019, 79, 4258–4270. [Google Scholar] [CrossRef] [PubMed]
  28. Modi, A.; Roy, D.; Sharma, S.; Vishnoi, J.R.; Pareek, P.; Elhence, P.; Sharma, P.; Purohit, P. ABC transporters in breast cancer: Their roles in multidrug resistance and beyond. J. Drug Target. 2022, 30, 927–947. [Google Scholar] [CrossRef] [PubMed]
  29. Marques, A.V.L.; Ruginsk, B.E.; Prado, L.d.O.; de Lima, D.E.; Daniel, I.W.; Moure, V.R.; Valdameri, G. The association of ABC proteins with multidrug resistance in cancer. Biochim. Biophys. Acta-Mol. Cell Res. 2025, 1872, 119878. [Google Scholar] [CrossRef] [PubMed]
  30. Abd El-Aziz, Y.S.; Spillane, A.J.; Jansson, P.J.; Sahni, S. Role of ABCB1 in mediating chemoresistance of triple-negative breast cancers. Biosci. Rep. 2021, 41, BSR20204092. [Google Scholar] [CrossRef] [PubMed]
  31. Huang, P.; Li, F.; Li, L.; You, Y.; Luo, S.; Dong, Z.; Gao, Q.; Wu, S.; Brünner, N.; Stenvang, J. lncRNA profile study reveals the mRNAs and lncRNAs associated with docetaxel resistance in breast cancer cells. Sci. Rep. 2018, 8, 17970. [Google Scholar] [CrossRef] [PubMed]
  32. Li, J.; Kang, J.; Liu, W.; Liu, J.; Pan, G.; Mao, A.; Zhang, Q.; Lu, J.; Ding, J.; Li, H. Docetaxel-resistant triple-negative breast cancer cell-derived exosomal lncrna linc00667 reduces the chemosensitivity of breast cancer cells to docetaxel via targeting mir-200b-3p/bcl-2 axis. Eur. J. Histochem. 2022, 66, 3529. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, J.; Kong, L.; Bian, W.; Lin, X.; Wei, F.; Chu, J. CircRNA CircABCB1 Diminishes the Sensitivity of Breast Cancer Cells to Docetaxel by Sponging MiR-153-3p. Tohoku J. Exp. Med. 2023, 261, 25–33. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, X.; Wang, X.; Gu, J.; Wei, Y.; Wang, Y. circUBR5 promotes ribosome biogenesis and induces docetaxel resistance in triple-negative breast cancer cell lines via the miR-340-5p/CMTM6/c-MYC axis. Neoplasia 2025, 59, 101062. [Google Scholar] [CrossRef] [PubMed]
  35. Zong, B.; Sun, L.; Peng, Y.; Wang, Y.; Yu, Y.; Lei, J.; Zhang, Y.; Guo, S.; Li, K.; Liu, S. HORMAD1 promotes docetaxel resistance in triple negative breast cancer by enhancing DNA damage tolerance. Oncol. Rep. 2021, 46, 138. [Google Scholar] [CrossRef] [PubMed]
  36. Li, J.; Sun, P.; Huang, T.; He, S.; Li, L.; Xue, G. Individualized chemotherapy guided by the expression of ERCC1, RRM1, TUBB3, TYMS and TOP2A genes versus classic chemotherapy in the treatment of breast cancer: A comparative effectiveness study. Oncol. Lett. 2020, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  37. Jing, L.; Lan, L.; Zhang, M.; Zhang, Z. METTL3/LINC00662/miR-186-5p feedback loop regulates docetaxel resistance in triple negative breast cancer. Sci. Rep. 2022, 12, 16715. [Google Scholar] [CrossRef] [PubMed]
  38. Ou, X.; Tan, Y.; Xie, J.; Yuan, J.; Deng, X.; Shao, R.; Song, C.; Cao, X.; Xie, X.; He, R.; et al. Methylation of GPRC5A promotes liver metastasis and docetaxel resistance through activating mTOR signaling pathway in triple negative breast cancer. Drug Resist. Updat. 2024, 73, 101063. [Google Scholar] [CrossRef] [PubMed]
  39. Mann, J.; Yang, N.; Montpetit, R.; Kirschenman, R.; Lemieux, H.; Goping, I.S. BAD sensitizes breast cancer cells to docetaxel with increased mitotic arrest and necroptosis. Sci. Rep. 2020, 10, 355. [Google Scholar] [CrossRef] [PubMed]
  40. Kang, J.; Xiong, H.; Jiang, X.; Huang, Z.; Guo, Y.; Cao, Y.; Ding, J. Factors influencing pathological complete response to neoadjuvant chemotherapy in resectable breast cancer: A retrospective study. Oncol. Lett. 2025, 30, 366. [Google Scholar] [CrossRef] [PubMed]
  41. Mavroudis, D.; Papakotoulas, P.; Ardavanis, A.; Syrigos, K.; Kakolyris, S.; Ziras, N.; Kouroussis, C.; Malamos, N.; Polyzos, A.; Christophyllakis, C.; et al. Randomized phase III trial comparing docetaxel plus epirubicin versus docetaxel plus capecitabine as first-line treatment in women with advanced breast cancer. Ann. Oncol. 2009, 21, 48–54. [Google Scholar] [CrossRef] [PubMed]
  42. de Hoon, J.P.J.; Veeck, J.; Vriens, B.E.P.J.; Calon, T.G.A.; van Engeland, M.; Tjan-Heijnen, V.C.G. Taxane resistance in breast cancer: A closed HER2 circuit? Biochim. Biophys. Acta-Rev. Cancer 2012, 1825, 197–206. [Google Scholar] [CrossRef] [PubMed]
  43. Field, J.J.; Kanakkanthara, A.; Miller, J.H. Microtubule-targeting agents are clinically successful due to both mitotic and interphase impairment of microtubule function. Bioorganic Med. Chem. 2015, 22, 5050–5059. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, H.; Zhang, M.; Deng, Y. Long Noncoding RNAs in Taxane Resistance of Breast Cancer. Int. J. Mol. Sci. 2023, 24, 12253. [Google Scholar] [CrossRef] [PubMed]
  45. Fromes, Y.; Gounon, P.; Veitia, R.; Bissery, M.C.; Fellous, A. Influence of microtubule-associated proteins on the differential effects of paclitaxel and docetaxel. J. Protein Chem. 1996, 15, 377–388. [Google Scholar] [CrossRef] [PubMed]
  46. Verweij, J.; Clavel, M.; Chevalier, B. Paclitaxel (TaxolTM) and docetaxel (TaxotereTM): Not simply two of a kind. Ann. Oncol. 1994, 5, 495–505. [Google Scholar] [CrossRef] [PubMed]
  47. Morse, D.L.; Gray, H.; Payne, C.M.; Gillies, R.J. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol. Cancer Ther. 2005, 4, 1495–1504. [Google Scholar] [CrossRef] [PubMed]
  48. McGrogan, B.T.; Gilmartin, B.; Carney, D.N.; McCann, A. Taxanes, microtubules and chemoresistant breast cancer. Biochim. Biophys. Acta-Rev. Cancer 2008, 1785, 96–132. [Google Scholar] [CrossRef] [PubMed]
  49. Han, T.L.; Sha, H.; Ji, J.; Li, Y.T.; Wu, D.S.; Lin, H.; Hu, B.; Jiang, Z.X. Depletion of Survivin suppresses docetaxel-induced apoptosis in HeLa cells by facilitating mitotic slippage. Sci. Rep. 2021, 11, 2283. [Google Scholar] [CrossRef] [PubMed]
  50. Xu, A.P.; Xu, L.B.; Smith, E.R.; Fleishman, J.S.; Chen, Z.S.; Xu, X.X. Cell death in cancer chemotherapy using taxanes. Front. Pharmacol. 2023, 14, 1338633. [Google Scholar] [CrossRef] [PubMed]
  51. Alalawy, A.I. Key genes and molecular mechanisms related to Paclitaxel Resistance. Cancer Cell Int. 2024, 24, 244. [Google Scholar] [CrossRef] [PubMed]
  52. Jeon, Y.W.; Kim, O.H.; Shin, J.S.; Hong, H.E.; Kim, C.H.; Kim, S.J. Potentiation of the Anticancer Effects by Combining Docetaxel with Ku-0063794 against Triple-Negative Breast Cancer Cells. Cancer Res. Treat. 2022, 54, 157–173. [Google Scholar] [CrossRef] [PubMed]
  53. Abal, M.; Andreu, J.; Barasoain, I. Taxanes: Microtubule and Centrosome Targets, and Cell Cycle Dependent Mechanisms of Action. Curr. Cancer Drug Targets 2005, 3, 193–203. [Google Scholar] [CrossRef] [PubMed]
  54. Jordan, M.A.; Wilson, L. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 2004, 4, 253–265. [Google Scholar] [CrossRef] [PubMed]
  55. Carlson, K.; Ocean, A.J. Peripheral neuropathy with microtubule-targeting agents: Occurrence and management approach. Clin. Breast Cancer 2011, 11, 73–81. [Google Scholar] [CrossRef] [PubMed]
  56. Kudlowitz, D.; Muggia, F. Clinical features of taxane neuropathy. Anticancer. Drugs 2014, 25, 495–501. [Google Scholar] [CrossRef] [PubMed]
  57. Assaraf, Y.G.; Brozovic, A.; Gonçalves, A.C.; Jurkovicova, D.; Linē, A.; Machuqueiro, M.; Saponara, S.; Sarmento-Ribeiro, A.B.; Xavier, C.P.R.; Vasconcelos, M.H. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist. Updat. 2019, 46, 100645. [Google Scholar] [CrossRef] [PubMed]
  58. Savy, T.; Flanders, L.; Karpanasamy, T.; Sun, M.; Gerlinger, M. Cancer evolution: From Darwin to the Extended Evolutionary Synthesis. Trends Cancer 2025, 11, 204–215. [Google Scholar] [CrossRef] [PubMed]
  59. Gatenby, R.A.; Brown, J.S. The evolution and ecology of resistance in cancer therapy. Cold Spring Harb. Perspect. Med. 2020, 10, a040972. [Google Scholar] [CrossRef] [PubMed]
  60. Kalli, M.; Poskus, M.D.; Stylianopoulos, T.; Zervantonakis, I.K. Beyond matrix stiffness: Targeting force-induced cancer drug resistance. Trends Cancer 2023, 9, 937–954. [Google Scholar] [CrossRef] [PubMed]
  61. Mansur, M.B.; deSouza, N.M.; Natrajan, R.; Abegglen, L.M.; Schiffman, J.D.; Greaves, M. Evolutionary determinants of curability in cancer. Nat. Ecol. Evol. 2023, 7, 1761–1770. [Google Scholar] [CrossRef] [PubMed]
  62. Ghebeh, H.; Al-Khaldi, S.; Olabi, S.; Al-Dhfyan, A.; Al-Mohanna, F.; Barnawi, R.; Tulbah, A.; Al-Tweigeri, T.; Ajarim, D.; Al-Alwan, M. Fascin is involved in the chemotherapeutic resistance of breast cancer cells predominantly via the PI3K/Akt pathway. Br. J. Cancer 2014, 111, 1552–1561. [Google Scholar] [CrossRef] [PubMed]
  63. Stubbs, N.M.; Roady, T.J.; Schwermann, M.P.; Eteshola, E.O.; MacDonald, W.J.; Purcell, C.; Ryspayeva, D.; Verovkina, N.; Tajiknia, V.; Ghandali, M.; et al. Acquired resistance to molecularly targeted therapies for cancer. Cancer Drug Resist. 2025, 8, 27. [Google Scholar] [CrossRef] [PubMed]
  64. Vander Velde, R.; Yoon, N.; Marusyk, V.; Durmaz, A.; Dhawan, A.; Miroshnychenko, D.; Lozano-Peral, D.; Desai, B.; Balynska, O.; Poleszhuk, J.; et al. Resistance to targeted therapies as a multifactorial, gradual adaptation to inhibitor specific selective pressures. Nat. Commun. 2020, 11, 2393. [Google Scholar] [CrossRef] [PubMed]
  65. Friedman, R. Drug resistance in cancer: Molecular evolution and compensatory proliferation. Oncotarget 2016, 7, 11746–11755. [Google Scholar] [CrossRef] [PubMed]
  66. Ferrari, P.; Scatena, C.; Ghilli, M.; Bargagna, I.; Lorenzini, G.; Nicolini, A. Molecular Mechanisms, Biomarkers and Emerging Therapies for Chemotherapy Resistant TNBC. Int. J. Mol. Sci. 2022, 23, 1665. [Google Scholar] [CrossRef] [PubMed]
  67. Seidel, E.; von Karstedt, S. Extrinsic cell death pathway plasticity: A driver of clonal evolution in cancer? Cell Death Discov. 2022, 8, 465. [Google Scholar] [CrossRef] [PubMed]
  68. Gerlinger, M.; Swanton, C. How Darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine. Br. J. Cancer 2010, 103, 1139–1143. [Google Scholar] [CrossRef] [PubMed]
  69. Langhammer, S. Rationale for the design of an oncology trial using a generic targeted therapy multi-drug regimen for NSCLC patients without treatment options (Review). Oncol. Rep. 2013, 30, 1535–1541. [Google Scholar] [CrossRef] [PubMed]
  70. Engle, K.; Kumar, G. Cancer multidrug-resistance reversal by ABCB1 inhibition: A recent update. Eur. J. Med. Chem. 2022, 239, 114542. [Google Scholar] [CrossRef] [PubMed]
  71. Mosca, L.; Ilari, A.; Fazi, F.; Assaraf, Y.G.; Colotti, G. Taxanes in cancer treatment: Activity, chemoresistance and its overcoming. Drug Resist. Updat. 2021, 54, 100742. [Google Scholar] [CrossRef] [PubMed]
  72. Yin, S.; Bhattacharya, R.; Cabral, F. Human mutations that confer paclitaxel resistance. Mol. Cancer Ther. 2010, 9, 327–335. [Google Scholar] [CrossRef] [PubMed]
  73. Yuan, S.F.; Zhu, L.J.; Zheng, W.E.; Chen, H.; Wu, L.L.; Zhang, W.; Sun, H.Y.; Chen, W.J. Expression of β-tubulin III and survivin in advance stage breast cancer correlates with chemotheraputic effects of docetaxel. Asian Pac. J. Cancer Prev. 2012, 13, 361–365. [Google Scholar] [CrossRef] [PubMed]
  74. Lebok, P.; Öztürk, M.; Heilenkötter, U.; Jaenicke, F.; Müller, V.; Paluchowski, P.; Geist, S.; Wilke, C.; Burandt, E.; Lebeau, A.; et al. High levels of class III β-tubulin expression are associated with aggressive tumor features in breast cancer. Oncol. Lett. 2016, 11, 1987–1994. [Google Scholar] [CrossRef] [PubMed]
  75. Rouzier, R.; Rajan, R.; Wagner, P.; Hess, K.R.; Gold, D.L.; Stec, J.; Ayers, M.; Ross, J.S.; Zhang, P.; Buchholz, T.A.; et al. Microtubule-associated protein tau: A marker of paclitaxel sensitivity in breast cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 8315–8320. [Google Scholar] [CrossRef] [PubMed]
  76. Mimori, K.; Sadanaga, N.; Yoshikawa, Y.; Ishikawa, K.; Hashimoto, M.; Tanaka, F.; Sasaki, A.; Inoue, H.; Sugimachi, K.; Mori, M. Reduced tau expression in gastric cancer can identify candidates for successful Paclitaxel treatment. Br. J. Cancer 2006, 94, 1894–1897. [Google Scholar] [CrossRef] [PubMed]
  77. Smoter, M.; Bodnar, L.; Duchnowska, R.; Stec, R.; Grala, B.; Szczylik, C. The role of Tau protein in resistance to paclitaxel. Cancer Chemother. Pharmacol. 2011, 68, 553–557. [Google Scholar] [CrossRef] [PubMed]
  78. Spicakova, T.; O’Brien, M.M.; Duran, G.E.; Sweet-Cordero, A.; Sikic, B.I. Expression and silencing of the microtubule-associated protein Tau in breast cancer cells. Mol. Cancer Ther. 2010, 9, 2970–2981. [Google Scholar] [CrossRef] [PubMed]
  79. Baquero, M.T.; Hanna, J.A.; Neumeister, V.; Cheng, H.; Molinaro, A.M.; Harris, L.N.; Rimm, D.L. Stathmin expression and its relationship to microtubule-associated protein tau and outcome in breast cancer. Cancer 2012, 118, 4660–4669. [Google Scholar] [CrossRef] [PubMed]
  80. Das, T.; Anand, U.; Pandey, S.K.; Ashby, C.R.; Assaraf, Y.G.; Chen, Z.S.; Dey, A. Therapeutic strategies to overcome taxane resistance in cancer. Drug Resist. Updat. 2021, 55, 100754. [Google Scholar] [CrossRef] [PubMed]
  81. Galletti, G.; Zhang, C.; Gjyrezi, A.; Cleveland, K.; Zhang, J.; Powell, S.; Thakkar, P.V.; Betel, D.; Shah, M.A.; Giannakakou, P. Microtubule Engagement with Taxane Is Altered in Taxane-Resistant Gastric Cancer. Clin. Cancer Res. 2020, 26, 3771–3783. [Google Scholar] [CrossRef] [PubMed]
  82. Choi, Y.; Yu, A.-M. ABC Transporters in Multidrug Resistance and Pharmacokinetics, and Strategies for Drug Development. Curr. Pharm. Des. 2014, 20, 793–807. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, D.; Jia, T.; Chen, X.; Jiang, H.; Guo, T.; Dong, J.; Zeng, H.; Wang, Y.; Yuan, Y. Bufalin reverses ABCB1-mediated resistance to docetaxel in breast cancer. Heliyon 2023, 9, e13840. [Google Scholar] [CrossRef] [PubMed]
  84. Domanitskaya, N.; Wangari-Talbot, J.; Jacobs, J.; Peiffer, E.; Mahdaviyeh, Y.; Paulose, C.; Malofeeva, E.; Foster, K.; Cai, K.Q.; Zhou, Y.; et al. Abcc10 status affects mammary tumour growth, metastasis, and docetaxel treatment response. Br. J. Cancer 2014, 111, 696–707. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, B.; Fatayer, H.; Hanby, A.M.; Horgan, K.; Perry, S.L.; Valleley, E.M.A.; Verghese, E.T.; Williams, B.J.; Thorne, J.L.; Hughes, T.A. Neoadjuvant Chemotherapy Induces Expression Levels of Breast Cancer Resistance Protein That Predict Disease-Free Survival in Breast Cancer. PLoS ONE 2013, 8, e62766. [Google Scholar] [CrossRef] [PubMed]
  86. Hirth, J.A.; Watkins, P.B.; Strawderman, M.; Schott, A.; Bruno, R.; Baker, L.H. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin. Cancer Res. 2000, 6, 1255–1258. [Google Scholar] [PubMed]
  87. Guo, Y.; He, W.; Yang, S.; Zhao, D.; Li, Z.; Luan, Y. Co-delivery of docetaxel and verapamil by reduction-sensitive PEG-PLGA-SS-DTX conjugate micelles to reverse the multi-drug resistance of breast cancer. Colloids Surf. B Biointerfaces 2017, 151, 119–127. [Google Scholar] [CrossRef] [PubMed]
  88. Mi, Y.; Lou, L. ZD6474 reverses multidrug resistance by directly inhibiting the function of P-glycoprotein. Br. J. Cancer 2007, 97, 934–940. [Google Scholar] [CrossRef] [PubMed]
  89. Pusztai, L.; Wagner, P.; Ibrahim, N.; Rivera, E.; Theriault, R.; Booser, D.; Symmans, F.W.; Wong, F.; Blumenschein, G.; Fleming, D.R.; et al. Phase II study of tariquidar, a selective P-glycoprotein inhibitor, in patients with chemotherapy-resistant, advanced breast carcinoma. Cancer 2005, 104, 682–691. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, D.; Hung, N.; Hung, T.; Eden, K.; Chan, W.K.; Kwan, R.; Qin, A.; Chang, C.; Duffull, S.; Glue, P.; et al. Oral docetaxel plus encequidar—A phase 1 clinical trial. Cancer Chemother. Pharmacol. 2024, 94, 475–481. [Google Scholar] [CrossRef] [PubMed]
  91. Zhu, S.; Sun, C.; Cai, Z.; Li, Y.; Liu, W.; Luan, Y.; Wang, C. Effective therapy of advanced breast cancer through synergistic anticancer by paclitaxel and P-glycoprotein inhibitor. Mater. Today Bio 2024, 26, 101029. [Google Scholar] [CrossRef] [PubMed]
  92. Zhao, M.; Lei, C.; Yang, Y.; Bu, X.; Ma, H.; Gong, H.; Liu, J.; Fang, X.; Hu, Z.; Fang, Q. Abraxane, the nanoparticle formulation of paclitaxel can induce drug resistance by up-regulation of P-gp. PLoS ONE 2015, 10, e0131429. [Google Scholar] [CrossRef] [PubMed]
  93. Pluta, P.; Jesionek-Kupnicka, D.; Pluta, A.; Brzozowski, K.; Braun, M.; Kubicka-Wołkowska, J.; Piekarski, J. Prognostic value of XIAP and survivin expression in locally advanced breast cancer patients treated with anthracycline-based neoadjuvant chemotherapy. Arch. Med. Sci. 2023, 19, 343–354. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, D.; Chen, M.B.; Wang, L.Q.; Yang, L.; Liu, C.Y.; Lu, P.H. Bcl-2 expression predicts sensitivity to chemotherapy in breast cancer: A systematic review and meta-analysis. J. Exp. Clin. Cancer Res. 2013, 32, 105. [Google Scholar] [CrossRef] [PubMed]
  95. Tewari, M.; Pradhan, S.; Singh, U.; Singh, T.B.; Shukla, H.S. Assessment of predictive markers of response to neoadjuvant chemotherapy in breast cancer. Asian J. Surg. 2010, 33, 157–167. [Google Scholar] [CrossRef] [PubMed]
  96. Cory, S.; Roberts, A.W.; Colman, P.M.; Adams, J.M. Targeting BCL-2-like Proteins to Kill Cancer Cells. Trends Cancer 2016, 2, 443–460. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, K.M.; Lee, H.; Han, D.; Moon, W.K.; Kim, K.; Oh, H.J.; Choi, J.; Hwang, E.H.; Kang, S.E.; Im, S.A.; et al. Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer. Breast Cancer Res. 2020, 22, 130. [Google Scholar] [CrossRef] [PubMed]
  98. Bonaiuti, P.; Chiroli, E.; Gross, F.; Corno, A.; Vernieri, C.; Štefl, M.; Cosentino Lagomarsino, M.; Knop, M.; Ciliberto, A. Cells Escape an Operational Mitotic Checkpoint through a Stochastic Process. Curr. Biol. 2018, 28, 28–37.e7. [Google Scholar] [CrossRef] [PubMed]
  99. Lok, T.M.; Wang, Y.; Xu, W.K.; Xie, S.; Ma, H.T.; Poon, R.Y.C. Mitotic slippage is determined by p31comet and the weakening of the spindle-assembly checkpoint. Oncogene 2020, 39, 2819–2834. [Google Scholar] [CrossRef] [PubMed]
  100. Dey, G.; Bharti, R.; Das, A.K.; Sen, R.; Mandal, M. Resensitization of Akt Induced Docetaxel Resistance in Breast Cancer by ‘Iturin A’ a Lipopeptide Molecule from Marine Bacteria Bacillus megaterium. Sci. Rep. 2017, 7, 17324. [Google Scholar] [CrossRef] [PubMed]
  101. Kaboli, P.J.; Imani, S.; Jomhori, M.; Ling, K.-H. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs. the current chemotherapy. Am. J. Cancer Res. 2021, 11, 5155–5183. [Google Scholar] [PubMed]
  102. Dong, M.; Shan, B.; Han, X.; Zhao, X.; Wang, F.; Zhu, L.; Ou, Q.; Ma, X.; Pan, Y. Baseline Mutations and Up-Regulation of PI3K-AKT Pathway Serve as Potential Indicators of Lack of Response to Neoadjuvant Chemotherapy in Stage II/III Breast Cancer. Front. Oncol. 2022, 11, 784985. [Google Scholar] [CrossRef] [PubMed]
  103. Al-Alwan, M.; Olabi, S.; Ghebeh, H.; Barhoush, E.; Tulbah, A.; Al-Tweigeri, T.; Ajarim, D.; Adra, C. Fascin is a key regulator of breast cancer invasion that acts via the modification of metastasis-associated molecules. PLoS ONE 2011, 6, e27339. [Google Scholar] [CrossRef] [PubMed]
  104. Safi, A.; Heidarian, E.; Ahmadi, R. Quercetin Synergistically Enhances the Anticancer Efficacy of Docetaxel through Induction of Apoptosis and Modulation of PI3K/AKT, MAPK/ERK, and JAK/STAT3 Signaling Pathways in MDA-MB-231 Breast Cancer Cell Line. Int. J. Mol. Cell. Med. 2021, 10, 11. [Google Scholar] [CrossRef] [PubMed]
  105. Snyder, M.; Huang, J.; Huang, X.Y.; Zhang, J.J. A signal transducer and activator of transcription 3·nuclear factor κB (Stat3·NFκB) complex is necessary for the expression of fascin in metastatic breast cancer cells in response to interleukin (IL)-6 and tumor necrosis factor (TNF)-α. J. Biol. Chem. 2014, 289, 30082–30089. [Google Scholar] [CrossRef] [PubMed]
  106. Mussa, A.; Afolabi, H.A.; Syed, N.H.; Talib, M.; Murtadha, A.H.; Hajissa, K.; Mokhtar, N.F.; Mohamud, R.; Hassan, R. The NF-κB Transcriptional Network Is a High-Dose Vitamin C-Targetable Vulnerability in Breast Cancer. Biomedicines 2023, 11, 1060. [Google Scholar] [CrossRef] [PubMed]
  107. Sprowl, J.A.; Reed, K.; Armstrong, S.R.; Lanner, C.; Guo, B.; Kalatskaya, I.; Stein, L.; Hembruff, S.L.; Tam, A.; Parissenti, A.M. Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: A study in isogenic resistant tumor cells. Breast Cancer Res. 2012, 14, R2. [Google Scholar] [CrossRef] [PubMed]
  108. Zeng, A.; Liang, X.; Zhu, S.; Liu, C.; Luo, X.; Zhang, Q.; Song, L. Baicalin, a Potent Inhibitor of NF-κB Signaling Pathway, Enhances Chemosensitivity of Breast Cancer Cells to Docetaxel and Inhibits Tumor Growth and Metastasis Both In Vitro and In Vivo. Front. Pharmacol. 2020, 11, 879. [Google Scholar] [CrossRef] [PubMed]
  109. Ashrafizadeh, M.; Mirzaei, S.; Hashemi, F.; Zarrabi, A.; Zabolian, A.; Saleki, H.; Sharifzadeh, S.O.; Soleymani, L.; Daneshi, S.; Hushmandi, K.; et al. New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomed. Pharmacother. 2021, 141, 111824. [Google Scholar] [CrossRef] [PubMed]
  110. Zhang, C.C.; Yan, Z.; Zong, Q.; Fang, D.D.; Painter, C.; Zhang, Q.; Chen, E.; Lira, M.E.; John-Baptiste, A.; Christensen, J.G. Synergistic Effect of the γ-Secretase Inhibitor PF-03084014 and Docetaxel in Breast Cancer Models. Stem Cells Transl. Med. 2013, 2, 233–242. [Google Scholar] [CrossRef] [PubMed]
  111. Hanrahan, K.; O’Neill, A.; Prencipe, M.; Bugler, J.; Murphy, L.; Fabre, A.; Puhr, M.; Culig, Z.; Murphy, K.; Watson, R.W. The role of epithelial-mesenchymal transition drivers ZEB1 and ZEB2 in mediating docetaxel-resistant prostate cancer. Mol. Oncol. 2017, 11, 251–265. [Google Scholar] [CrossRef] [PubMed]
  112. Ren, J.; Chen, Y.; Song, H.; Chen, L.; Wang, R. Inhibition of ZEB1 reverses EMT and chemoresistance in docetaxel-resistant human lung adenocarcinoma cell line. J. Cell. Biochem. 2013, 114, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
  113. He, W.; Sun, Z.; Liu, Z. Silencing of TGM2 reverses epithelial to mesenchymal transition and modulates the chemosensitivity of breast cancer to docetaxel. Exp. Ther. Med. 2015, 10, 1413–1418. [Google Scholar] [CrossRef] [PubMed]
  114. Pan, Y.; Chen, J.; Tao, L.; Zhang, K.; Wang, R.; Chu, X.; Chen, L. Long noncoding RNA ROR regulates chemoresistance in docetaxelresistant lung adenocarcinoma cells via epithelial mesenchymal transition pathway. Oncotarget 2017, 8, 33144–33158. [Google Scholar] [CrossRef] [PubMed]
  115. Damiano, V.; Brisotto, G.; Borgna, S.; di Gennaro, A.; Armellin, M.; Perin, T.; Guardascione, M.; Maestro, R.; Santarosa, M. Epigenetic silencing of miR-200c in breast cancer is associated with aggressiveness and is modulated by ZEB1. Genes Chromosom. Cancer 2017, 56, 147–158. [Google Scholar] [CrossRef] [PubMed]
  116. Khajah, M.A.; Al-Ateyah, A.; Luqmani, Y.A. MicroRNA expression profiling of endocrine sensitive and resistant breast cancer cell lines. Biochem. Biophys. Rep. 2022, 31, 101316. [Google Scholar] [CrossRef] [PubMed]
  117. Yao, Y.S.; Qiu, W.S.; Yao, R.Y.; Zhang, Q.; Zhuang, L.K.; Zhou, F.; Sun, L.B.; Yue, L. MiR-141 confers docetaxel chemoresistance of breast cancer cells via regulation of EIF4E expression. Oncol. Rep. 2015, 33, 2504–2512. [Google Scholar] [CrossRef] [PubMed]
  118. Edwards, A.; Brennan, K. Notch Signalling in Breast Development and Cancer. Front. Cell Dev. Biol. 2021, 9, 692173. [Google Scholar] [CrossRef] [PubMed]
  119. Barnawi, R.; Al-Khaldi, S.; Majed Sleiman, G.; Sarkar, A.; Al-Dhfyan, A.; Al-Mohanna, F.; Ghebeh, H.; Al-Alwan, M. Fascin Is Critical for the Maintenance of Breast Cancer Stem Cell Pool Predominantly via the Activation of the Notch Self-Renewal Pathway. Stem Cells 2016, 34, 2799–2813. [Google Scholar] [CrossRef] [PubMed]
  120. Sánchez-Espinosa, A.; García-Rodríguez, J.; Alonso-Aguirre, V.; Acosta-Ortega, J.M.; Conesa-Zamora, P.; García-Solano, J.; Luengo-Gil, G. Expression of Fascin and DNA Topoisomerase 2-Alpha in Breast Carcinoma: Correlation with Histological Subtypes and Other Prognostic Markers. Int. J. Mol. Sci. 2025, 26, 3076. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, H.; Zhang, Y.; Li, L.; Cao, J.; Guo, Y.; Wu, Y.; Gao, W. Fascin actin-bundling protein 1 in human cancer: Promising biomarker or therapeutic target? Mol. Ther.-Oncolytics 2021, 20, 240–264. [Google Scholar] [CrossRef] [PubMed]
  122. Barnawi, R.; Al-Khaldi, S.; Bakheet, T.; Fallatah, M.; Alaiya, A.; Ghebeh, H.; Al-Alwan, M. Fascin Activates β-Catenin Signaling and Promotes Breast Cancer Stem Cell Function Mainly Through Focal Adhesion Kinase (FAK): Relation With Disease Progression. Front. Oncol. 2020, 10, 440. [Google Scholar] [CrossRef] [PubMed]
  123. Zhao, X.; Gao, S.; Sun, W.; Zhang, H.; Sun, J.; Yang, S.; Hao, J. Hypoxia-Inducible factor-1 promotes pancreatic ductal adenocarcinoma invasion and metastasis by activating transcription of the actin-Bundling protein fascin. Cancer Res. 2014, 74, 2455–2464. [Google Scholar] [CrossRef] [PubMed]
  124. Bu, M.; Liu, X.; Liu, X.; Xu, W. Upregulation of fascin-1 is involved in HIF-1α-dependent invasion and migration of hypopharyngeal squamous cell carcinoma. Int. J. Oncol. 2019, 55, 488–498. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, W.; Wei, X.; Jiao, Y.; Bai, Y.; Sam, W.N.; Yan, Q.; Sun, X.; Li, G.; Ma, J.; Wei, W.; et al. STAT3/HIF-1α/fascin-1 axis promotes RA FLSs migration and invasion ability under hypoxia. Mol. Immunol. 2022, 142, 83–94. [Google Scholar] [CrossRef] [PubMed]
  126. Snyder, M.; Huang, X.Y.; Zhang, J.J. Signal Transducers and Activators of Transcription 3 (STAT3) directly regulates cytokine-induced fascin expression and is required for breast cancer cell migration. J. Biol. Chem. 2011, 286, 38886–38893. [Google Scholar] [CrossRef] [PubMed]
  127. Huang, B.; Lang, X.; Li, X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front. Oncol. 2022, 12, 1023177. [Google Scholar] [CrossRef] [PubMed]
  128. Li, A.; Dawson, J.C.; Forero-Vargas, M.; Spence, H.J.; Yu, X.; König, I.; Anderson, K.; Machesky, L.M. The Actin-Bundling Protein Fascin Stabilizes Actin in Invadopodia and Potentiates Protrusive Invasion. Curr. Biol. 2010, 20, 339–345. [Google Scholar] [CrossRef] [PubMed]
  129. Xiao, Z.S.; Zhao, L.; Zhang, X.N.; Li, H.X.; Yin, Z.H. Effect of rs67085638 in long non-coding RNA (CCAT1) on colon cancer chemoresistance to paclitaxel through modulating the microRNA-24-3p and FSCN1. J. Cell. Mol. Med. 2021, 25, 3744–3753. [Google Scholar] [CrossRef] [PubMed]
  130. Abdullah, A.R.; Gamal El-Din, A.M.; El-Mahdy, H.A.; Ismail, Y.; El-Husseiny, A.A. The crucial role of fascin-1 in the pathogenesis, metastasis, and chemotherapeutic resistance of breast cancer. Pathol. Res. Pract. 2024, 254, 155079. [Google Scholar] [CrossRef] [PubMed]
  131. Min, K.W.; Chae, S.W.; Kim, D.H.; Do, S.I.; Kim, K.; Lee, H.J.; Sohn, J.H.; Pyo, J.S.; Kim, D.H.; Oh, S.; et al. Fascin expression predicts an aggressive clinical course in patients with advanced breast cancer. Oncol. Lett. 2015, 10, 121–130. [Google Scholar] [CrossRef] [PubMed]
  132. Bi, J.; Chen, X.; Zhang, Y.; Li, B.; Sun, J.; Shen, H.; Kong, C. Fascin is a predictor for invasiveness and recurrence of urothelial carcinoma of bladder. Urol. Oncol. Semin. Orig. Investig. 2012, 30, 688–694. [Google Scholar] [CrossRef] [PubMed]
  133. Belnap, C.; Divis, T.; Kingsley, K.; Howard, K.M. Differential Expression of MicroRNA MiR-145 and MiR-155 Downstream Targets in Oral Cancers Exhibiting Limited Chemotherapy Resistance. Int. J. Mol. Sci. 2024, 25, 2167. [Google Scholar] [CrossRef] [PubMed]
  134. Huang, J.; Dey, R.; Wang, Y.; Jakoncic, J.; Kurinov, I.; Huang, X.Y. Structural Insights into the Induced-fit Inhibition of Fascin by a Small-Molecule Inhibitor. J. Mol. Biol. 2018, 430, 1324–1335. [Google Scholar] [CrossRef] [PubMed]
  135. Asensi-Cantó, A.; Rodríguez-Braun, E.; Beltrán-Videla, A.; Hurtado, A.M.; Conesa-Zamora, P. Effects of imipramine on cancer patients over-expressing Fascin1; description of the HITCLIF clinical trial. Front. Oncol. 2023, 13, 1238464. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Y.; Jillian Zhang, J.; Huang, X.Y. Anti-metastasis fascin inhibitors decrease the growth of specific subtypes of cancers. Cancers 2020, 12, 2287. [Google Scholar] [CrossRef] [PubMed]
  137. Koo, D.H.; Lee, H.J.; Ahn, J.H.; Yoon, D.H.; Kim, S.B.; Gong, G.; Son, B.H.; Ahn, S.H.; Jung, K.H. Tau and PTEN status as predictive markers for response to trastuzumab and paclitaxel in patients with HER2-positive breast cancer. Tumor Biol. 2015, 36, 5865–5871. [Google Scholar] [CrossRef] [PubMed]
  138. Alkhathami, A.G.; Pallathadka, H.; Shah, S.; Ganesan, S.; Sharma, A.; Devi, S.; Mustafa, Y.F.; Alasheqi, M.Q.; Kadhim, A.J.; Zwamel, A.H. Mechanisms behind the LncRNAs-mediated regulation of paclitaxel (PTX) resistance in human malignancies. Exp. Cell Res. 2025, 445, 114434. [Google Scholar] [CrossRef] [PubMed]
  139. Ren, S.; Wang, Y.; Wang, J.; Gao, D.; Zhang, M.; Ding, N.; Li, Y. Synthesis and biological evaluation of novel larotaxel analogues. Eur. J. Med. Chem. 2018, 156, 692–710. [Google Scholar] [CrossRef] [PubMed]
  140. Valsalakumari, R.; Feith, M.; Pettersen, S.; Åslund, A.K.O.; Mørch, Ý.; Skotland, T.; Sandvig, K.; Mælandsmo, G.M.; Iversen, T.G. Combinatorial Effects of Free and Nanoencapsulated Forms of Cabazitaxel and RAS-Selective Lethal 3 in Breast Cancer Cells. Pharmaceutics 2025, 17, 657. [Google Scholar] [CrossRef] [PubMed]
  141. Meyer-Wilmes, P.; Huober, J.; Untch, M.; Blohmer, J.U.; Janni, W.; Denkert, C.; Klare, P.; Link, T.; Rhiem, K.; Bayer, C.; et al. Long-term outcomes of a randomized, open-label, phase II study comparing cabazitaxel versus paclitaxel as neoadjuvant treatment in patients with triple-negative or luminal B/HER2-negative breast cancer (GENEVIEVE). ESMO Open 2024, 9, 103009. [Google Scholar] [CrossRef] [PubMed]
  142. Erciyestepe, M.; Ekinci, Ö.B.; Seçmeler, Ş.; Selvi, O.; Öztürk, A.E.; Aydin, O.; Büyükkuşcu, A.; Atasever, T.; Çelik, E.; Ertürk, K.; et al. Evaluation of ixabepilone efficacy and tolerability in metastatic breast cancer. Medicine 2024, 103, e40649. [Google Scholar] [CrossRef] [PubMed]
  143. Roskoski, R. Targeted and cytotoxic inhibitors used in the treatment of lung cancers. Pharmacol. Res. 2024, 209, 107465. [Google Scholar] [CrossRef] [PubMed]
  144. James, N.; Owusu, E.; Rivera, G.; Bandyopadhyay, D. Small Molecule Therapeutics in the Pipeline Targeting for Triple-Negative Breast Cancer: Origin, Challenges, Opportunities, and Mechanisms of Action. Int. J. Mol. Sci. 2024, 25, 6285. [Google Scholar] [CrossRef] [PubMed]
  145. Hurvitz, S.A.; Dalenc, F.; Campone, M.; O’Regan, R.M.; Tjan-Heijnen, V.C.; Gligorov, J.; Llombart, A.; Jhangiani, H.; Mirshahidi, H.R.; Tan-Chiu, E.; et al. A phase 2 study of everolimus combined with trastuzumab and paclitaxel in patients with HER2-overexpressing advanced breast cancer that progressed during prior trastuzumab and taxane therapy. Breast Cancer Res. Treat. 2013, 141, 437–446. [Google Scholar] [CrossRef] [PubMed]
  146. Feng, M.; Santhanam, R.K.; Xing, H.; Zhou, M.; Jia, H. Inhibition of γ-secretase/Notch pathway as a potential therapy for reversing cancer drug resistance. Biochem. Pharmacol. 2024, 220, 115991. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, X.; Wang, T.; Song, X.; Gao, J.; Xu, G.; Ma, Y.; Song, G. Current Status of Hedgehog Signaling Inhibitors. Curr. Top. Med. Chem. 2024, 24, 243–258. [Google Scholar] [CrossRef] [PubMed]
  148. Gong, C.; Xia, Y.; Zhu, Y.; Yang, Y.; Lin, Q.; Liu, Q.; Yang, W.; Ling, L.; Zhong, J.; Duan, Z.; et al. Preclinical study and phase 2 trial of neoadjuvant pyrotinib combined with chemotherapy in luminal/HER2-low breast cancer: PILHLE-001 study. Cell Rep. Med. 2024, 5, 101807. [Google Scholar] [CrossRef] [PubMed]
  149. Jimenez, M.M.; Zotano, A.L.G.; Perez, M.E.; Borrego, M.R.; Martinez, N.; Lopez-Muniz, J.I.C.; Gil, M.J.; Conejero, R.A.; Bermejo, B.; Sanchez-Rovira, P.; et al. 112MO Neoadjuvant study of 12 months of abemaciclib plus letrozole vs. chemotherapy in HR+/HER2– highly proliferative (Ki67≥20%) breast cancer: CARABELA (GEICAM/2019-01) trial. ESMO Open 2024, 9, 103100. [Google Scholar] [CrossRef]
  150. Sparano, J.A.; Zhao, F.; Martino, S.; Ligibel, J.A.; Perez, E.A.; Saphner, T.; Wolff, A.C.; Sledge, G.W.; Wood, W.C.; Davidson, N.E. Long-term follow-up of the E1199 phase III trial evaluating the role of taxane and schedule in operable breast cancer. J. Clin. Oncol. 2015, 33, 2353–2360. [Google Scholar] [CrossRef] [PubMed]
  151. Zhang, L.; Geng, C.; Liu, Y.; Han, J.; Han, M.; Huang, R.; Li, X.; Luo, R.; Li, Y.; Ma, J.; et al. A phase II trial comparing dalpiciclib in combination with letrozole versus standard chemotherapy as neoadjuvant therapy in patients with high-risk HR- positive HER-2 negative breast cancer: DARLING-02. J. Clin. Oncol. 2024, 42, TPS626. [Google Scholar] [CrossRef]
  152. Yang, Z.; Wang, X.; Chen, A.; Meng, R.; Zhao, Y.; Liu, H.; Ge, J.; Huang, R.; Bai, J.; Cao, X. Neoadjuvant dalpiciclib, exemestane, and goserelin in premenopausal women with HR-positive, HER2-negative breast cancer: A prospective, multicenter, randomized two-group, non-controlled phase 2 clinical trial. J. Clin. Oncol. 2024, 42, TPS625. [Google Scholar] [CrossRef]
  153. Slamon, D.J.; Fasching, P.A.; Hurvitz, S.; Chia, S.; Crown, J.; Martín, M.; Barrios, C.H.; Bardia, A.; Im, S.A.; Yardley, D.A.; et al. Rationale and trial design of NATALEE: A Phase III trial of adjuvant ribociclib + endocrine therapy versus endocrine therapy alone in patients with HR+/HER2− early breast cancer. Ther. Adv. Med. Oncol. 2023, 15, 17588359231178125. [Google Scholar] [CrossRef] [PubMed]
  154. Pascual, T.; Chic, N.; Fernandez-Martinez, A.; González-Farré, B.; Paré, L.; Saura, C.; Hernando, C.; Muñoz, M.; Arumí, M.; Galván, P.; et al. Abstract PD17-07: Cell-Cycle Inhibition and Immune Microenvironment in HR+/HER2- Breast Cancer During and After preoperative ribociclib and letrozole vesus chemotherapy: A correlative analysis of the 1402-SOLTI/CORALLEEN phase 2 trial. Cancer Res. 2023, 83, PD17-07. [Google Scholar] [CrossRef]
  155. Huo, S.; Xue, J.; Wang, S.; Shan, H.; Chen, G.; Niu, N.; Wang, Y.; Qiu, F.; Zhao, Y.; Xing, F.; et al. A pilot trial of neoadjuvant pyrotinib plus trastuzumab, dalpiciclib, and letrozole for triple-positive breast cancer. MedComm 2024, 5, e505. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, J.; Lin, L.; Long, Q.; Zhang, Q.; Sun, G.; Zhou, L.; Wang, Q.; Zhu, J.; Li, F.; Hu, W. HLX11, a Proposed Pertuzumab Biosimilar: Pharmacokinetics, Immunogenicity, and Safety Profiles Compared to Three Reference Biologic Products (US-, EU-, and CN-Approved Pertuzumab) Administered to Healthy Male Subjects. BioDrugs 2022, 36, 393–409. [Google Scholar] [CrossRef] [PubMed]
  157. McAndrew, N.P.; Hurvitz, S.A.; Tetef, M.L.; Kivork, C.; Ikenouye, L.; Slamon, D.J. UCLA B-13: A phase 1b trial evaluating the safety of ribociclib, tucatinib, and trastuzumab in patients with metastatic, HER2+ breast cancer and a multicenter, randomized, open-label, phase 2 study of preoperative treatment with ribociclib, trastuzumab, t. J. Clin. Oncol. 2023, 41, TPS1116. [Google Scholar] [CrossRef]
  158. Harbeck, N.; Braun, M.; Gluz, O.; Schmid, P.; Graeser, M.K.; Hartkopf, A.D.; Hoffmann, O.; Kostara, A.; Polata, S.; Volmer, L.L.; et al. Neoadjuvant dynamic marker-adjusted personalized therapy comparing trastuzumab-deruxtecan versus pacli-/docetaxel + carboplatin + trastuzumab + pertuzumab in HER2+ early breast cancer: WSG-ADAPT-HER2-IV. J. Clin. Oncol. 2024, 42, TPS631. [Google Scholar] [CrossRef]
  159. Liang, X.; Zhang, N.; Zhou, X. Abstract P5-05-24: RC48-ADC (Disitamab vedotin, a HER2-directed antibody-drug conjugate) in Combination with Pyrotinib for Trastuzumab-treated HER2-Positive Recurrent or Metastatic Breast Cancer: A Phase Ib/II Study. Clin. Cancer Res. 2025, 31, P5-05-24. [Google Scholar] [CrossRef]
  160. Liu, J.; Song, C.; Yang, Y.; Wang, X.; Ni, M.; Wang, X.; Chen, L.; Yang, H.; Zhao, R.; Xu, T.; et al. Safety and Efficacy of KN046 in Combination with KN026 in Patients with Advanced HER2-Positive Breast Cancer: A Phase II Trial. Clin. Cancer Res. 2025, 31, 2379–2385. [Google Scholar] [CrossRef] [PubMed]
  161. Loganathan, S.; Klein, J.; Haggag, A.Z.; Shaw, A.A.; Lawrence, T.E.; Liu, M.S.; Rao Betha, M. A randomized, double-blind, three-arm, parallel group, phase 1 study to assess the pharmacokinetics, safety, and tolerability of PERT-IJS following a single dose of 420 mg intravenous infusion compared to the EU-and US-marketed drug product (pertuzumab; PERT) in healthy male volunteers. J. Clin. Oncol. 2025, 43, e13009. [Google Scholar] [CrossRef]
  162. Liu, B.; Xie, N.; Tian, C.; Feng, R.; Hu, Z.Y.; Li, J.; Liu, L.; Xiao, H.; Yang, X.; Zeng, M.; et al. Exploring the clinical outcomes and safety profile of inetetamab treatment in metastatic breast cancer patients: A multicenter assessment of a Chinese-origin recombinant Anti-HER2 monoclonal antibody. Breast 2023, 72, 103597. [Google Scholar] [CrossRef] [PubMed]
  163. Kuemmel, S.; Harper-Wynne, C.; Park, Y.H.; Franke, F.; de Laurentiis, M.; Schumacher-Wulf, E.; Eiger, D.; Heeson, S.; Cardona, A.; Özyilkan, Ö.; et al. heredERA Breast Cancer: A phase III, randomized, open-label study evaluating the efficacy and safety of giredestrant plus the fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection in patients with previously untreated HER2-positi. BMC Cancer 2024, 24, 641. [Google Scholar] [CrossRef] [PubMed]
  164. Li, J.; Wang, Z.H.; Chen, L.; Zhang, W.J.; Ma, L.X.X.; Wu, J.; Liu, G.Y.; Hou, Y.F.; Yu, K.D.; Di, G.H.; et al. Efficacy and safety of neoadjuvant SHR-A1811 with or without pyrotinib in women with locally advanced or early HER2-positive breast cancer: A randomized, open-label, phase II trial. Ann. Oncol. 2025, 36, 651–659. [Google Scholar] [CrossRef] [PubMed]
  165. Ishiba, T.; Nishibuchi, I.; Hara, F.; Sasaki, K.; Sadate, R.; Sekino, Y.; Machida, R.; Fukuda, H.; Kogawa, T.; Fujisawa, T.; et al. Abstract PO3-19-01: A Randomized Controlled Trial of Metastasis-directed Therapy for Oligometastases in Breast Cancer (OLIGAMI trial; JCOG2110). Cancer Res. 2024, 84, PO3-19-01. [Google Scholar] [CrossRef]
  166. Xu, R.-H.; Wang, S.; Ruan, D.; Lin, S.; Liu, F.-R.; Wu, H.-X.; Huang, J.J.; Zheng, Q.; Jiang, K.; Du, X.; et al. Safety and efficacy of TQB2102, a novel bispecific anti-HER2 antibody–drug conjugate, in patients with advanced solid tumors: Preliminary data from the first-in-human phase 1 trial. J. Clin. Oncol. 2025, 43, 3003. [Google Scholar] [CrossRef]
  167. Wang, C.; Zhang, X.; Yin, S.; Liu, M.; Yang, H.; Xie, F.; Peng, Y.; Yang, Y.; Wang, S.; Du, W.; et al. A prospective phase II study of neoadjuvant therapy guided by personalized drug-testing on patient-derived tumor-like cell clusters for early breast cancer. J. Clin. Oncol. 2024, 42, e12625. [Google Scholar] [CrossRef]
  168. Zheng, C.; Liu, Y.; Wang, X.; Bi, Z.; Qiu, P.; Qiao, G.; Bi, X.; Shi, Z.; Zhang, Z.; Chen, P.; et al. Clinical efficacy and biomarker analysis of neoadjuvant camrelizumab plus chemotherapy for early-stage triple-negative breast cancer: A experimental single-arm phase II clinical trial pilot study. Int. J. Surg. 2024, 110, 1527–1536. [Google Scholar] [CrossRef] [PubMed]
  169. Stecklein, S.R.; Aripoli, A.; Salgado, R.; O’Dea, A.; Nye, L.E.; Larson, K.E.; Satelli, D.; Rader, R.; Madan, R.; Yoder, R.; et al. NeoTRACT: Phase II trial of neoadjuvant tumor infiltrating lymphocyte- and response-adapted chemoimmunotherapy for triple-negative breast cancer (TNBC). J. Clin. Oncol. 2024, 42, TPS629. [Google Scholar] [CrossRef]
  170. Bardia, A.; Gucalp, A.; DaCosta, N.; Gabrail, N.; Danso, M.; Ali, H.; Blackwell, K.L.; Carey, L.A.; Eisner, J.R.; Baskin-Bey, E.S.; et al. Phase 1 study of seviteronel, a selective CYP17 lyase and androgen receptor inhibitor, in women with estrogen receptor-positive or triple-negative breast cancer. Breast Cancer Res. Treat. 2018, 171, 111–120. [Google Scholar] [CrossRef] [PubMed]
  171. Saltos, A.N.; Arrowood, C.; Beasley, G.; Ronald, J.; El-Haddad, G.; Khan, U.; Guerra-Guevara, L.; Wolf, S.; Gu, L.; Wang, X.F.; et al. Abstract CT103: A phase 1 first-in-human study of MEM-288 oncolytic virus in solid tumors including non-small cell lung cancer (NSCLC): Impact on tumor and systemic T cell immunity. Cancer Res. 2023, 83, CT103. [Google Scholar] [CrossRef]
  172. Zeng, H.A.; Lv, H.M.; Zhang, M.W.; Niu, L.M.; Wang, J.; Sun, H.H.; Liu, Z.Z.; Yan, M. Docetaxel rechallenge in HER2-negative metastatic breast cancer: A real-world study of previously discontinued patients for non-progression reasons. J. Cancer Res. Clin. Oncol. 2025, 151, 89. [Google Scholar] [CrossRef] [PubMed]
  173. He, F.; Liu, J.; Shen, X.; Wang, Z.; Li, Q.; Li, G. Adverse Event Profile for Nanoparticle Albumin-Bound Paclitaxel Compared With Solvent-Based Taxanes in Solid-Organ Tumors: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Ann. Pharmacother. 2022, 56, 898–909. [Google Scholar] [CrossRef] [PubMed]
  174. Guo, S.; Vieweger, M.; Zhang, K.; Yin, H.; Wang, H.; Li, X.; Li, S.; Hu, S.; Sparreboom, A.; Evers, B.M.; et al. Ultra-thermostable RNA nanoparticles for solubilizing and high-yield loading of paclitaxel for breast cancer therapy. Nat. Commun. 2020, 11, 972. [Google Scholar] [CrossRef] [PubMed]
  175. Teixeira, S.; Carvalho, M.A.; Castanheira, E.M.S. Functionalized Liposome and Albumin-Based Systems as Carriers for Poorly Water-Soluble Anticancer Drugs: An Updated Review. Biomedicines 2022, 10, 486. [Google Scholar] [CrossRef] [PubMed]
  176. Niza, E.; Ocaña, A.; Castro-Osma, J.A.; Bravo, I.; Alonso-Moreno, C. Polyester polymeric nanoparticles as platforms in the development of novel nanomedicines for cancer treatment. Cancers 2021, 13, 3387. [Google Scholar] [CrossRef] [PubMed]
  177. Junnuthula, V.; Kolimi, P.; Nyavanandi, D.; Sampathi, S.; Vora, L.K.; Dyawanapelly, S. Polymeric Micelles for Breast Cancer Therapy: Recent Updates, Clinical Translation and Regulatory Considerations. Pharmaceutics 2022, 14, 1860. [Google Scholar] [CrossRef] [PubMed]
  178. McFadden, M.; Singh, S.K.; Kinnel, B.; Varambally, S.; Singh, R. The effect of paclitaxel- and fisetin-loaded PBM nanoparticles on apoptosis and reversal of drug resistance gene ABCG2 in ovarian cancer. J. Ovarian Res. 2023, 16, 220. [Google Scholar] [CrossRef] [PubMed]
  179. Souchek, J.J.; Davis, A.L.; Hill, T.K.; Holmes, M.B.; Qi, B.; Singh, P.K.; Kridel, S.J.; Mohs, A.M. Combination treatment with orlistat-containing nanoparticles and taxanes is synergistic and enhances microtubule stability in taxane-resistant prostate cancer cells. Mol. Cancer Ther. 2017, 16, 1819–1830. [Google Scholar] [CrossRef] [PubMed]
  180. Guo, X.; Zhao, Z.; Chen, D.; Qiao, M.; Wan, F.; Cun, D.; Sun, Y.; Yang, M. Co-delivery of resveratrol and docetaxel via polymeric micelles to improve the treatment of drug-resistant tumors. Asian J. Pharm. Sci. 2019, 14, 78–85. [Google Scholar] [CrossRef] [PubMed]
  181. Jurczyk, M.; Kasperczyk, J.; Wrześniok, D.; Beberok, A.; Jelonek, K. Nanoparticles Loaded with Docetaxel and Resveratrol as an Advanced Tool for Cancer Therapy. Biomedicines 2022, 10, 1187. [Google Scholar] [CrossRef] [PubMed]
  182. Dian, C.; Qian, Z.; Ran, M.; Yan, X.; Dian, L. Co-Delivery of Docetaxel and Curcumin Functionalized Mixed Micelles for the Treatment of Drug-Resistant Breast Cancer by Oral Administration. Int. J. Nanomed. 2024, 19, 8603–8620. [Google Scholar] [CrossRef] [PubMed]
  183. Pramual, S.; Lirdprapamongkol, K.; Atjanasuppat, K.; Chaisuriya, P.; Niamsiri, N.; Svasti, J. PLGA-Lipid Hybrid Nanoparticles for Overcoming Paclitaxel Tolerance in Anoikis-Resistant Lung Cancer Cells. Molecules 2022, 27, 8295. [Google Scholar] [CrossRef] [PubMed]
  184. Bowerman, C.J.; Byrne, J.D.; Chu, K.S.; Schorzman, A.N.; Keeler, A.W.; Sherwood, C.A.; Perry, J.L.; Luft, J.C.; Darr, D.B.; Deal, A.M.; et al. Docetaxel-loaded PLGA nanoparticles improve efficacy in taxane-resistant triple-negative breast cancer. Nano Lett. 2017, 17, 242–248. [Google Scholar] [CrossRef] [PubMed]
  185. Patel, N.R.; Pattni, B.S.; Abouzeid, A.H.; Torchilin, V.P. Nanopreparations to overcome multidrug resistance in cancer. Adv. Drug Deliv. Rev. 2013, 65, 1748–1762. [Google Scholar] [CrossRef] [PubMed]
  186. Feng, L.; Mumper, R.J. A critical review of lipid-based nanoparticles for taxane delivery. Cancer Lett. 2013, 334, 157–175. [Google Scholar] [CrossRef] [PubMed]
  187. Swetha, K.L.; Paul, M.; Maravajjala, K.S.; Kumbham, S.; Biswas, S.; Roy, A. Overcoming drug resistance with a docetaxel and disulfiram loaded pH-sensitive nanoparticle. J. Control. Release 2023, 356, 93–114. [Google Scholar] [CrossRef] [PubMed]
  188. Ye, X.; Chen, X.; He, R.; Meng, W.; Chen, W.; Wang, F.; Meng, X. Enhanced anti-breast cancer efficacy of co-delivery liposomes of docetaxel and curcumin. Front. Pharmacol. 2022, 13, 969611. [Google Scholar] [CrossRef] [PubMed]
  189. Di Gregorio, E.; Romiti, C.; Di Lorenzo, A.; Cavallo, F.; Ferrauto, G.; Conti, L. RGD_PLGA Nanoparticles with Docetaxel: A Route for Improving Drug Efficiency and Reducing Toxicity in Breast Cancer Treatment. Cancers 2023, 15, 8. [Google Scholar] [CrossRef] [PubMed]
  190. Tran, P.; Nguyen, T.N.; Lee, Y.; Tran, P.N.; Park, J.S. Docetaxel-loaded PLGA nanoparticles to increase pharmacological sensitivity in MDA-MB-231 and MCF-7 breast cancer cells. Korean J. Physiol. Pharmacol. 2021, 25, 479–488. [Google Scholar] [CrossRef] [PubMed]
  191. Da Rocha, M.C.O.; Da Silva, P.B.; Radicchi, M.A.; Andrade, B.Y.G.; De Oliveira, J.V.; Venus, T.; Merker, C.; Estrela-Lopis, I.; Longo, J.P.F.; Báo, S.N. Docetaxel-loaded solid lipid nanoparticles prevent tumor growth and lung metastasis of 4T1 murine mammary carcinoma cells. J. Nanobiotechnol. 2020, 18, 43. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, Y.; Wang, Q.; Wang, X.; Yao, P.; Dai, Q.; Qi, X.; Yang, M.; Zhang, X.; Huang, R.; Yang, J.; et al. Docetaxel-loaded pH/ROS dual-responsive nanoparticles with self-supplied ROS for inhibiting metastasis and enhancing immunotherapy of breast cancer. J. Nanobiotechnol. 2023, 21, 286. [Google Scholar] [CrossRef] [PubMed]
  193. Basak, M.; Kulkarni, M.; Narisepalli, S.; Chitkara, D.; Mittal, A. Exosomal fragment enclosed polyamine-salt nano-complex for co-delivery of docetaxel and mir-34a exhibits higher cytotoxicity and apoptosis in breast cancer cells. Sci. Rep. 2024, 14, 21669. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, Y.; Cheng, W.Y.; Zhu, J.; He, L.; Ren, W.Y.; Bao, D.; Piao, J.G. Programmed Co-delivery of tamoxifen and docetaxel using lipid-coated mesoporous silica nanoparticles for overcoming CYP3A4-mediated resistance in triple-negative breast cancer treatment. Biomed. Pharmacother. 2024, 170, 116084. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of treatment schemes used in breast cancer management. This schematic summarises the standard treatment strategies for early and advanced breast cancer across molecular subtypes, highlighting that taxane-based regimens, including docetaxel, are typically integrated in neoadjuvant, adjuvant, and metastatic settings.
Figure 1. Overview of treatment schemes used in breast cancer management. This schematic summarises the standard treatment strategies for early and advanced breast cancer across molecular subtypes, highlighting that taxane-based regimens, including docetaxel, are typically integrated in neoadjuvant, adjuvant, and metastatic settings.
Ijms 26 07119 g001
Figure 2. Schematic representation of the mechanisms of action of docetaxel. Docetaxel and paclitaxel bind to the β-subunit of tubulin within the microtubules, preventing depolymerisation and disrupting mitotic spindle dynamics. This leads to cell cycle arrest at the G2/M phase and triggers apoptosis in proliferating cancer cells.
Figure 2. Schematic representation of the mechanisms of action of docetaxel. Docetaxel and paclitaxel bind to the β-subunit of tubulin within the microtubules, preventing depolymerisation and disrupting mitotic spindle dynamics. This leads to cell cycle arrest at the G2/M phase and triggers apoptosis in proliferating cancer cells.
Ijms 26 07119 g002
Figure 3. Overview of intrinsic and acquired mechanisms contributing to resistance to docetaxel. This figure illustrates how pre-existing (intrinsic) resistance and treatment-induced (acquired) adaptations enable tumour cells to survive docetaxel exposure, leading to clonal selection and tumour relapse driven by resistant subpopulations.
Figure 3. Overview of intrinsic and acquired mechanisms contributing to resistance to docetaxel. This figure illustrates how pre-existing (intrinsic) resistance and treatment-induced (acquired) adaptations enable tumour cells to survive docetaxel exposure, leading to clonal selection and tumour relapse driven by resistant subpopulations.
Ijms 26 07119 g003
Figure 4. Schematic representation of the mechanisms underlying resistance to docetaxel. Mechanisms that enhance cell proliferation, depicted at the top of the image, include increased expression of anti-apoptotic genes and overactivation of survival pathways like PI3K/AKT. Conversely, resistance mechanisms aimed at reducing drug efficacy are shown at the bottom, including the upregulation of efflux transporters such as MDR, βIII-tubulin, metabolic enzymes, and non-coding RNAs.
Figure 4. Schematic representation of the mechanisms underlying resistance to docetaxel. Mechanisms that enhance cell proliferation, depicted at the top of the image, include increased expression of anti-apoptotic genes and overactivation of survival pathways like PI3K/AKT. Conversely, resistance mechanisms aimed at reducing drug efficacy are shown at the bottom, including the upregulation of efflux transporters such as MDR, βIII-tubulin, metabolic enzymes, and non-coding RNAs.
Ijms 26 07119 g004
Figure 5. Nanoparticle-based strategies to overcome docetaxel resistance in breast cancer cells. This schematic illustrates how modern, nanoparticle-based delivery systems enhance docetaxel efficacy by circumventing key resistance mechanisms in breast cancer. Nanocarriers such as albumin-bound nanoparticles (e.g., nab-paclitaxel), polymeric micelles, and lipid–polymer hybrids improve drug uptake through passive or active targeting and avoid P-gp-mediated efflux. Some formulations (e.g., RNA-based nanoassemblies or folate-targeted combinations) modulate resistance pathways, suppress anti-apoptotic proteins (e.g., Bcl-2, survivin), or restore apoptotic signalling. Others sustain intracellular drug retention, reduce microtubule alterations, or inhibit survival pathways (e.g., PI3K/Akt, NF-κB). Collectively, these strategies contribute to overcoming multidrug resistance, enhancing tumour cytotoxicity, and potentially delaying treatment failure.
Figure 5. Nanoparticle-based strategies to overcome docetaxel resistance in breast cancer cells. This schematic illustrates how modern, nanoparticle-based delivery systems enhance docetaxel efficacy by circumventing key resistance mechanisms in breast cancer. Nanocarriers such as albumin-bound nanoparticles (e.g., nab-paclitaxel), polymeric micelles, and lipid–polymer hybrids improve drug uptake through passive or active targeting and avoid P-gp-mediated efflux. Some formulations (e.g., RNA-based nanoassemblies or folate-targeted combinations) modulate resistance pathways, suppress anti-apoptotic proteins (e.g., Bcl-2, survivin), or restore apoptotic signalling. Others sustain intracellular drug retention, reduce microtubule alterations, or inhibit survival pathways (e.g., PI3K/Akt, NF-κB). Collectively, these strategies contribute to overcoming multidrug resistance, enhancing tumour cytotoxicity, and potentially delaying treatment failure.
Ijms 26 07119 g005
Table 1. Predictive biomarkers of docetaxel resistance in breast cancer. This table summarises molecular and clinicopathological biomarkers associated with docetaxel resistance in breast cancer, as reported in peer-reviewed, recent PubMed-indexed studies. It includes both preclinical and clinical evidence.
Table 1. Predictive biomarkers of docetaxel resistance in breast cancer. This table summarises molecular and clinicopathological biomarkers associated with docetaxel resistance in breast cancer, as reported in peer-reviewed, recent PubMed-indexed studies. It includes both preclinical and clinical evidence.
Biomarker (Type)Mechanism/RationaleEvidence TypeKey Findings (Study Details)References
ABCB1 (P-glycoprotein efflux pump)Drug efflux transporter; overexpression reduces intracellular docetaxel response.PreclinicalOverexpressed in resistant cells; inhibition restores sensitivity and drug accumulation; validated in mouse models.[28,29,30]
EPB41L4A-AS2 (lncRNA)Loss of tumour-suppressor lncRNA increases ABCB1 expression.PreclinicalAbsent in resistant cells; low expression associated with poor response to docetaxel.[31]
LINC00667 (exosomal lncRNA)Sponges miR-200b-3p to upregulate BCL2, reducing apoptosis.PreclinicalFound in resistant TNBC exosomes; downregulation sensitises cells to docetaxel.[32]
circABCB1 (circular RNA)CircABCB1 contributed to the docetaxel resistance of breast cancer, possibly via miR-153-3p sponging.PreclinicalThe overexpression of circABCB1 contributed to cell viability, docetaxel-resistance, and migration/invasion.[33]
circUBR5 (circular RNA)circUBR5 sponges miR-340-5p, releasing suppression of CMTM6 and promoting c-MYC-driven ribosome biogenesis, contributing to docetaxel resistance.PreclinicalKnocking down circUBR5 increased miR-340-5p activity, decreased CMTM6 levels, suppressed c-MYC activity, and sensitized cells to docetaxel—inducing apoptosis and reducing colony formation.[34]
HORMAD1 (protein)Promotes DNA damage tolerance via enhanced homologous recombination repair and protective autophagy, reducing docetaxel-induced apoptosis.PreclinicalHORMAD1 overexpression protects TNBC cells from docetaxel-induced DNA damage and apoptosis; its knockdown restores chemosensitivity via impaired DNA repair and enhanced apoptosis.[35]
TUBB3 (βIII-tubulin)Alters microtubule dynamics, reducing docetaxel binding.Preclinical + ClinicalHigh expression correlates with poor docetaxel response; inversely related to sensitivity.[22,36]
METTL3 (RNA methyltransferase), LINC00662 (lncRNA)Promotes docetaxel resistance by m6A-dependent stabilisation of LINC00662, forming a feedback loop with miR-186-5p that sustains METTL3 expression.PreclinicalHigh METTL3 and LINC00662 levels were observed in docetaxel-resistant TNBC cells and patient samples; disrupting the METTL3/LINC00662/miR-186-5p axis restored chemosensitivity and increased apoptosis.[37,38]
BAD (pro-apoptotic protein)Facilitates necroptosis during prolonged mitotic arrest induced by docetaxel, preventing mitotic slippage and survival of resistant cells.PreclinicalBAD expression promotes mitotic arrest and necroptotic death upon docetaxel treatment; its loss enables mitotic slippage and survival of chemoresistant polyploid cells. Tumours with high BAD show better response to taxanes.[39]
ER/PR-positive statusHormone-driven, low-proliferation tumours less responsive to taxanes.ClinicalAssociated with lower pCR rates compared to ER-negative tumours.[40] and others
HER2-positive statusHigh proliferation and HER2-targetability improve taxane response.ClinicalHER2+ tumours respond well to docetaxel-based regimens with HER2 inhibition.[40] and others
Triple-negative subtypeHigh initial sensitivity; prone to relapse if pCR not achieved.ClinicalHigher pCR rates but vulnerable to resistance upon incomplete response.[40] and others
Table 7. Comparison of delivery systems in breast cancer (2020–present).
Table 7. Comparison of delivery systems in breast cancer (2020–present).
Delivery SystemFormulation TypeMechanismProsConsReference
LiposomesLipid bilayer vesicles (~50–200 nm) encapsulating drugs (e.g., PEGylated liposomal doxorubicin).Passive tumour targeting via EPR; PEGylation (“stealth”) extends circulation; can be functionalized with ligands for active targeting.Biodegradable, biocompatible; carry both hydrophilic and hydrophobic drugs; protect drug, improve pharmacokinetics (prolonged half-life, stability) and reduce systemic toxicity.Rapid clearance by mononuclear phagocyte system without PEGylation; potential premature drug leakage and short circulation half-life; high manufacturing cost.[175]
Polymeric NPsBiodegradable polymer nanoparticles (e.g., PLA/PLGA nanospheres or nanocapsules loaded with paclitaxel—PICN® is a polymeric PTX NP approved in India for metastatic breast cancer).Polymer matrix entraps drug and releases it via controlled degradation; passive EPR targeting (with possible ligand-mediated active targeting).Highly versatile (wide choice of polymers); stable during storage and scalable manufacturing; tunable surface properties and drug release; high drug payload capacity; improve bioavailability and circulation time.Possible stability issues (tendency to aggregate); require precise manufacturing conditions; some formulations need PEGylation for prolonged circulation; potential toxicity of residual monomers or solvents.[176]
Polymeric MicellesSelf-assembled amphiphilic copolymer micelles (10–100 nm) solubilizing hydrophobic drugs in a core (e.g., PEG-PLA micelle paclitaxel, Genexol-PM®).Spontaneous micelle formation above a critical micelle concentration; drugs carried in core are released upon micelle dissociation or stimulus in tumour microenvironment.Easy to prepare; improve water solubility of hydrophobic drugs; prolong circulation and enhance tumour accumulation via EPR; increase drug efficacy and reduce toxicity (no need for harsh solubilizers like Cremophor)Limited stability in bloodstream—dilution below critical micelle concentration causes disassembly and rapid drug clearance (short half-life in circulation).[177]
Albumin-Bound NPsAlbumin-based nanoparticles or albumin–drug complexes (e.g., nab-Paclitaxel, Abraxane® ~130 nm, an albumin-bound paclitaxel approved for metastatic breast cancer).Exploit albumin’s natural pathways: passive tumour accumulation via EPR and active transcytosis (gp60 receptor) and binding to SPARC in tumour stroma, enhancing drug delivery to tumour sites.Biocompatible, non-immunogenic carrier; avoids toxic solvents (Abraxane is Cremophor-free); long circulation and tumour uptake via albumin receptors; improves drug solubility and bioavailabilityRequire cross-linking for nanoparticle stability (e.g., glutaraldehyde crosslinker, which can leave toxic residues); net negative charge of albumin can limit drug loading unless chemically modified.[175]
Table 8. Nanoparticle-based strategies to overcome docetaxel resistance in breast cancer preclinical studies (2020–2025). This table summarises peer-reviewed, PubMed-indexed preclinical studies published between 2020 and 2025 that investigated nanoparticle-based strategies to overcome docetaxel resistance in breast cancer. It includes information on nanoparticle type, therapeutic cargo, targeting strategies, resistance mechanisms addressed, key findings, development stage, and references.
Table 8. Nanoparticle-based strategies to overcome docetaxel resistance in breast cancer preclinical studies (2020–2025). This table summarises peer-reviewed, PubMed-indexed preclinical studies published between 2020 and 2025 that investigated nanoparticle-based strategies to overcome docetaxel resistance in breast cancer. It includes information on nanoparticle type, therapeutic cargo, targeting strategies, resistance mechanisms addressed, key findings, development stage, and references.
Nanoparticle TypeCargo (Co-Delivered Agents)Targeting StrategyResistance Mechanism AddressedKey FindingsStageReference
pH-sensitive PLGA nanoparticleDocetaxel + Disulfiram (DSF)pH-triggered release, TPGS-mediated P-gp inhibitionP-gp efflux, CSC survival, tumour stroma barrierRestored sensitivity in resistant cells, enhanced tumour accumulation, inhibited metastasis, superior efficacy in vivoPreclinical[187]
Liposome (CUR-DTX-L)Docetaxel + CurcuminPassive targeting (EPR)MDR via efflux transporters, survival signallingSynergistic cytotoxicity, prolonged half-life, tumour growth inhibition in xenograft modelPreclinical[188]
RGD-decorated PLGA nanoparticleDocetaxel (+ MRI/fluorescent tracers)αvβ3 integrin targetingLimited tumour uptake, systemic toxicityHigher tumour localization, reduced cardiotoxicity, improved efficacy in TNBC and HER2+ modelsPreclinical[189]
PLGA–TPGS polymeric nanoparticleDocetaxelPassive targeting, TPGS-mediated P-gp inhibitionGeneral MDR, poor intracellular accumulationIncreased potency, reduced IC50, sustained release, improved anti-proliferative effectPreclinical[190]
Solid lipid nanoparticle (SLN)DocetaxelPassive targeting, controlled releaseEMT, IL-6/BCL-2 survival signallingHigh cytotoxicity, G2/M arrest, prevented metastasis, suppressed IL-6 and BCL-2Preclinical[191]
Folate-targeted pH/ROS-dual responsive nanoparticleDocetaxel + CinnamaldehydeFolate receptor targeting, stimuli-responsive releaseTNBC metastasis, immune evasionImmunogenic cell death, blocked invasion, halted metastasis, enhanced anti-PD-1 responsePreclinical[192]
Exosome-coated polyamine nanocomplexDocetaxel + miR-34aBiomimetic targeting (exosomal membrane)miR-34a loss, anti-apoptotic signallingHigh cytotoxicity, BCL-2 downregulation, potent apoptosis inductionPreclinical[193]
Lipid-coated mesoporous silica nanoparticle (LP-MSN)Docetaxel + TamoxifenSequential release (Tamoxifen then DTX)CYP3A4-mediated metabolic resistanceEnhanced cytotoxicity via CYP3A4 inhibition, selective toxicity to TNBC cellsPreclinical[194]
Table 9. Nanoparticle-based docetaxel strategies in breast cancer clinical trials. This table summarizes Phase I–III clinical trials registered between 2020 and 2025 on nanoparticle-based docetaxel strategies aimed at overcoming resistance in breast cancer. Data are sourced from ClinicalTrials.gov and EudraCT. Only human studies are included.
Table 9. Nanoparticle-based docetaxel strategies in breast cancer clinical trials. This table summarizes Phase I–III clinical trials registered between 2020 and 2025 on nanoparticle-based docetaxel strategies aimed at overcoming resistance in breast cancer. Data are sourced from ClinicalTrials.gov and EudraCT. Only human studies are included.
Trial IdentifierNanoparticle FormulationCombination TherapyTargeting Strategy/Delivery TypePhaseStatusObjective Summary
NCT03671044Nanosomal Docetaxel Lipid Suspension (NDLS)None (monotherapy)Lipid-based, polysorbate-free formulation to improve solubility and tumour deliveryPhase 3RecruitingCompare efficacy and safety of NDLS vs. conventional docetaxel in TNBC patients resistant to prior chemotherapy.
NCT04931823CPO-100 (Albumin-bound Docetaxel)NoneAlbumin nanoparticle, solvent-free to enhance safety and tumour targetingPhase 1ActiveEvaluate MTD, safety, PK, and preliminary efficacy in advanced solid tumours refractory to standard treatment, including breast cancer.
NCT05114915Albumin-bound Docetaxel (HB1801)NoneAlbumin-stabilized nanoparticle, solvent-free for safer deliveryPhase 1RecruitingAssess safety, tolerability, PK, and preliminary efficacy in advanced solid tumours unresponsive to standard therapies (includes breast cancer).
NCT05254665Polymeric Micellar DocetaxelNonePolymeric micelle nanoparticle for improved tumour-specific deliveryPhase 2Not yet recruitingConfirm dose, assess safety and anti-tumour efficacy in taxane-resistant advanced solid tumours, including breast cancer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Postigo-Corrales, F.; Beltrán-Videla, A.; Lázaro-Sánchez, A.D.; Hurtado, A.M.; Conesa-Zamora, P.; Arroyo, A.B.; Luengo-Gil, G. Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions. Int. J. Mol. Sci. 2025, 26, 7119. https://doi.org/10.3390/ijms26157119

AMA Style

Postigo-Corrales F, Beltrán-Videla A, Lázaro-Sánchez AD, Hurtado AM, Conesa-Zamora P, Arroyo AB, Luengo-Gil G. Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions. International Journal of Molecular Sciences. 2025; 26(15):7119. https://doi.org/10.3390/ijms26157119

Chicago/Turabian Style

Postigo-Corrales, Fátima, Asunción Beltrán-Videla, Antonio David Lázaro-Sánchez, Ana María Hurtado, Pablo Conesa-Zamora, Ana Belén Arroyo, and Ginés Luengo-Gil. 2025. "Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions" International Journal of Molecular Sciences 26, no. 15: 7119. https://doi.org/10.3390/ijms26157119

APA Style

Postigo-Corrales, F., Beltrán-Videla, A., Lázaro-Sánchez, A. D., Hurtado, A. M., Conesa-Zamora, P., Arroyo, A. B., & Luengo-Gil, G. (2025). Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions. International Journal of Molecular Sciences, 26(15), 7119. https://doi.org/10.3390/ijms26157119

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

Article Metrics

Back to TopTop