Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us Membership Collaborators Partners
Publications > Journals > Oncology Advances> Article Full Text
OPEN ACCESS

Mitochondrial Dynamics in Breast Cancer Metastasis: From Metabolic Drivers to Therapeutic Targets

  • Bhuban Ruidas1,2,* 
Oncology Advances   2025;3(1):39-49

doi: 10.14218/OnA.2025.00001

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Ruidas B. Mitochondrial Dynamics in Breast Cancer Metastasis: From Metabolic Drivers to Therapeutic Targets. Oncol Adv. 2025;3(1):39-49. doi: 10.14218/OnA.2025.00001.

Abstract

Mitochondria are highly dynamic organelles that adapt to cellular stress and metabolic demands through processes such as fission, fusion, mitophagy, and transport, all of which are vital for maintaining cellular signaling and metabolic homeostasis. Fission facilitates mitochondrial division and biogenesis, while fusion enhances mitochondrial fitness and metabolic flexibility by mitigating damage. Together, these processes play a critical role in regulating cellular stress responses and apoptosis. Dysregulation of mitochondrial dynamics has been linked to impaired development and cancer progression, including breast cancer metastasis. A comprehensive understanding of mitochondrial dynamics in breast cancer progression is essential for advancing precision medicine. This review delves into the intricate molecular mechanisms governing mitochondrial biogenesis, fission, fusion, and mitophagy, with a particular focus on the role of mitophagy in maintaining mitochondrial homeostasis and its connection to metastasis progression. Furthermore, it discusses potential therapeutic strategies targeting mitochondrial dynamics and highlights the critical steps necessary to translate these approaches into clinical trials.

Keywords

Mitochondrial dynamics, Metabolic demands, Mitophagy, Breast cancer metastasis, Metabolic flexibility, Precision medicine, Therapies

Introduction

Metabolic reprogramming is a well-established hallmark of cancer, enabling cancer cells to adapt, survive, and proliferate under environmental constraints.1–3 This process is highly dynamic, resulting in distinct metabolic characteristics across different tumors, where cancer and stromal cells exhibit metabolic diversity.4,5 Originally described by Otto Warburg, tumor cells were thought to rely predominantly on aerobic glycolysis—known as the Warburg effect—for rapid growth and proliferation, attributed to presumed mitochondrial dysfunction.6 However, subsequent research has revealed that mitochondrial oxidation also plays a crucial role in supporting the metabolic demands of rapidly progressing cancers.6–8 Unlike normal cells, cancer cells frequently disrupt tightly regulated metabolic pathways, leading to the deregulation of signaling cascades. These alterations enable cancer cells to meet their elevated energy demands and support anabolic processes essential for tumor growth and survival.9,10 An emerging concept is that the tumor microenvironment imposes selective pressures that drive cancer cell evolution, ultimately promoting metastasis and drug resistance. This is particularly evident in triple-negative breast cancer (TNBC), which requires a high adenosine triphosphate (ATP) supply to sustain its rapid growth and proliferation.11–13 Metabolic rewiring is increasingly recognized as a key driver of breast cancer progression, as cancer cells adapt to fluctuations in energy demand and supply—a concept referred to as bioenergetic efficiency (BE).14,15

Breast cancer remains one of the leading causes of cancer-related mortality among women worldwide. TNBC, which accounts for approximately 15–20% of all breast cancer cases, is associated with a higher recurrence rate and poorer survival outcomes compared to non-TNBC.16,17 This aggressive subtype is characterized by the absence of three key receptors: estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. The lack of these therapeutic targets makes metastatic TNBC particularly challenging to treat, resulting in high fatality rates.18,19 Metabolic adaptations play a critical role in TNBC progression, with elevated lipolysis and fatty acid oxidation (FAO) compensating for high energy demands during rapid proliferation, even under hypoxic conditions.20,21 Metastatic cancer cells undergo extensive metabolic reprogramming, including increased fatty acid transport, lipid droplet accumulation, de novo lipogenesis, and β-oxidation, to meet their heightened ATP requirements. While glycolysis was once considered the predominant pathway driving cancer progression, subsequent findings have highlighted the direct involvement of mitochondrial metabolism in facilitating rapid tumor growth.22,23 Interestingly, mitochondrial bioenergetics and FAO have been shown to play pivotal roles in maintaining cancer stemness, supporting survival and proliferation, and driving chemoresistance in TNBC.24,25 This growing understanding underscores the importance of targeting metabolic pathways as a potential therapeutic strategy for aggressive breast cancer subtypes.26–28 Maintenance of BE is critical for metastatic cancer growth. BE is defined as the amount of ATP produced per molecule of nutrient metabolized in the mitochondria, while mitochondrial ATP synthesis capacity refers to the rate of ATP generation per unit of time.29,30 However, mitochondrial adaptation extends beyond BE, involving changes in mitochondrial architecture and adjustments to nutrient availability.31 In this context, TNBC preferentially utilizes readily available fatty acids as a primary energy source to meet its extensive energy demands required for rapid growth and survival.32 This is achieved through the strategic hijacking of mitochondrial bioenergetics machinery, enabling TNBC cells to sustain their high metabolic requirements.33,34 Mitochondria, widely regarded as central hubs of cellular metabolism, play a pivotal dual role in supporting the energetic and biosynthetic demands of oncogenesis. Their functions extend to facilitating malignant transformation, driving tumor progression, and influencing anticancer immunosurveillance mechanisms.35,36 Moreover, mitochondria support uncontrolled proliferation, drive tumor advancement, and enable anaplerotic processes that are vital for sustaining cancer cell metabolism under diverse and challenging environmental conditions.37,38

Mitochondrial dynamics encompass the processes of movement, tethering, fusion, and fission that collectively shape mitochondrial architecture.39 Increasing evidence highlights the critical role of mitochondrial dynamics in maintaining cell viability, preventing senescence, ensuring mitochondrial health, supporting bioenergetic function, regulating quality control, and mediating intracellular signaling.40,41 These dynamics are intricately linked to the balance between energy demand and ATP synthesis capacity, with mitochondrial architecture adapting in response to shifting metabolic needs. Transient shifts between interconnected and fragmented states reorganize mitochondrial components and remove damaged material through coordinated mitophagy, fission, and fusion, maintaining a healthy mitochondrial population.42,43 However, the dual demands of bioenergetic adaptation and quality control may conflict under specific nutrient conditions. Nutrient availability and metabolites can drive structural changes in mitochondria as an adaptive response to fluctuating ATP demands.44,45 This dynamic remodeling represents a potential therapeutic target, particularly in aggressive cancers, where disrupting the interplay between mitochondrial bioenergetics and architecture could impair the metabolic flexibility essential for tumor survival and proliferation.46,47

This review provides a comprehensive analysis of the critical role of mitochondrial dynamics and bioenergetics in driving the rapid progression of cancer cells, with a particular focus on breast cancer. It explores how these processes contribute to breast cancer metastasis and highlights their potential as therapeutic targets. The discussion spans the full spectrum of mitochondrial bioenergetics and biosynthesis, including biogenesis, turnover, fission, fusion, mitophagy, oxidative stress regulation, metabolism, and ATP synthesis. Furthermore, the review delves into the regulatory mechanisms linking mitochondrial dynamics to cancer progression, emphasizing their significance in identifying therapeutic opportunities. By consolidating current knowledge, it offers valuable insights into potential drug targets and novel therapeutic strategies to improve breast cancer treatment outcomes.

Mitochondrial metabolism and breast cancer plasticity

Metabolic variations across cancer types provide critical insights into cancer progression.48,49 Cellular metabolism differs between low- and high-grade tumors and between primary and metastatic sites.50 Recent studies have demonstrated the strong inhibitory effects of elesclomol on diverse cell types, including cancer stem cells, drug-resistant cells, and cells with low glycolytic activity, primarily by targeting and enhancing mitochondrial metabolism.51 Otto Warburg first identified aerobic glycolysis as a key driver of tumor growth, a concept later expanded in the 1990s with findings linking mitochondrial respiration to metastasis.52,53 Despite decades of research, progress in understanding energy metabolism reprogramming—one of cancer’s hallmarks—has been limited. While molecular therapies and immunotherapies have taken center stage, recent discoveries linking metabolic reprogramming to immune evasion and drug resistance have renewed interest in targeting cancer metabolism.54–56

Glucose metabolism begins with hexokinase converting glucose into glucose-6-phosphate, ultimately yielding ATP and pyruvate for mitochondrial respiration. Pyruvate enters the citric acid (tricarboxylic acid, TCA) cycle as acetyl-CoA, which fuels oxidative phosphorylation or is converted by ATP citrate lyase into cytoplasmic acetyl-CoA. This acetyl-CoA is essential for FAO and synthesis, with ACCα regulating FAO inhibition via malonyl-CoA production. Citrate serves as a key carbon source for fatty acid and cholesterol synthesis, facilitated by fatty acid synthase (FASN) and supported by NADPH (nicotinamide adenine dinucleotide phosphate) from the pentose phosphate pathway.57 Enzymes such as elongases and desaturases further process long-chain fatty acids into complex lipids crucial for membrane biogenesis, protein modification, and cellular signaling in metastasis.58,59 Elevated levels of FASN, citrate synthase, and ATP citrate lyase are commonly observed in malignancies, highlighting their role in tumor growth and survival.60 This interplay between glycolysis and lipid metabolism reveals promising therapeutic targets in cancer treatment.61,62 Cancer metabolic plasticity is believed to drive fatty acid (FA), phospholipid, and lipid synthesis through de novo lipogenesis, promoting tumor growth and survival.63 The tumor-specific antigen OA-519 highlights FASN activity, directly linking FA synthesis to cancer progression.64 Enhanced de novo FA synthesis in neoplastic tissues plays a critical role in tumor development, making enzyme targeting a promising therapeutic strategy.65,66 Genome-scale metabolic models of breast cancer gene expression underscore the importance of FA biosynthesis in early tumor progression, where anabolic processes dominate, whereas advanced stages rely more on reactive oxygen species (ROS) detoxification.67,68 This study offers fresh insights into tumor metabolism, demonstrating that ATP synthesis is lower in primary solid tumors than in normal tissues but increases in metastases.69 By quantifying tumor energy generation rates for the first time, these findings challenge the long-standing view of uniformly high tumor metabolism. Significant metabolic shifts occur throughout tumor progression, with significantly reduced TCA cycle flux in five primary tumors compared to their tissues of origin, yet elevated TCA flux in two breast cancer lung metastasis models relative to both advanced primary lesions and healthy lung tissue.70,71 Despite increased glycolytic flux, oxidative phosphorylation remains the dominant ATP source, suggesting reduced ATP production in primary tumors and increased ATP synthesis in metastases.72 Recent findings link triple-negative breast cancer’s invasiveness to its dominance over normal and immune cell mitochondrial metabolism, a mechanism also associated with therapy resistance. This may explain the elevated TCA flux observed in metastases.73,74

During breast cancer brain metastasis, breast cancer cells undergoing epithelial–mesenchymal transition show increased glycolysis and lipid metabolism, providing the energy and biosynthetic resources needed for local invasion.75 Moreover, cancer cells precisely regulate mitochondrial bioenergetics and dynamics to enhance fatty acid utilization, meeting their elevated energy demands and driving tumor growth. Beyond fatty acid metabolism, this mitochondrial adaptability highlights mitochondrial dynamics as a promising target for cancer therapy, warranting further exploration to fully understand its therapeutic potential.76,77

Mitochondrial fission, fusion, and mitophagy in breast cancer progression

Mitochondria are dynamic, double-membrane-bound organelles vital for cellular functions such as energy production via oxidative phosphorylation.78 Their morphology varies by cell type, reflecting specialized roles—fibroblast mitochondria form elongated filaments (1–10 µm long, ∼700 nm in diameter), while hepatocyte mitochondria are mostly spherical or ovoid.79 Constantly remodeled through fission, fusion, and motility, mitochondria adapt to cellular demands. In cultured fibroblasts, rapid fission and fusion enable the complete redistribution of mitochondrial green fluorescent protein (GFP) within an hour. The balance between fission and fusion governs mitochondrial morphology across different cell types and conditions.80,81 Recent evidence highlights a strong link between cancer progression and disrupted mitochondrial dynamics, which are regulated by the interplay of fission, fusion, and mitophagy processes.82–84 Mitochondrial fission, driven by proteins such as dynamin-related protein 1 (Drp1) and mitochondrial fission factor, supports cellular division and growth. In contrast, fusion, mediated by mitofusins (MFN1/2) and optic atrophy 1 (OPA1), maintains mitochondrial integrity and metabolic flexibility.85 In metastatic breast cancer, altered phosphorylation and expression of Drp1 play a pivotal role in modulating these dynamics, underscoring their significance in oncogenesis and their potential as therapeutic targets.86–88

Mitochondrial fission is essential for growing and dividing cells, ensuring an adequate mitochondrial population. The GTPase Drp1, critical for mitochondrial fission, is activated via phosphorylation by MAP kinases ERK1/2.89 During fission, Drp1 translocates to the outer mitochondrial membrane, binding to receptors such as Fis1, Mff, Mid49, and Mid51 to mediate membrane constriction and division.90,91 Mutations impairing Drp1 disrupt fission, causing excessive fusion and elongated mitochondrial tubules.92 Recent studies suggest that targeting mitochondrial fission factors can suppress breast cancer proliferation and invasion, offering therapeutic potential.93–95 Elevated levels of mitochondrial fission factors, including Drp1, also serve as prognostic markers. Higher Drp1 expression in metastatic cells suggests its upregulation may signal early breast cancer metastasis.96,97 Moreover, high Drp1 expression and mitochondrial fragmentation are vital for brain tumor-initiating cells, while Drp1 inhibition via genetic ablation or mDIVI1 reduces their tumorigenicity in vitro and in vivo.98 Drp1-dependent fission supports stem cell maintenance in mammary epithelial stem-like cells, enabling the preferential inheritance of newly generated mitochondria during asymmetric division. This enhances stem-like properties, whereas cells with older mitochondria exhibit reduced growth and differentiation potential.99–101 Drp1 inhibition disrupts mitochondrial asymmetry, diminishing stemness and impairing pluripotency reprogramming, emphasizing its role in stem cell biology. Targeting Drp1 to eliminate cancer stem cells offers a promising strategy for achieving lasting cancer cures by addressing tumor recurrence and resistance at its root.102–104 Moreover, the Notch pathway plays a pivotal role in breast cancer stem cell metastasis, with high Notch1 messenger RNA expression strongly linked to poor prognosis. Activation of NICD1, the functional domain of the Notch receptor, upregulates the mitochondrial fission factor Drp1, promoting metastasis and presenting therapeutic opportunities.105 Mitochondrial fission also impacts breast cancer progression through apoptosis and metabolic reprogramming. Since apoptosis is essential for tumor suppression, disrupted fission impairs apoptotic signaling, enabling tumor growth and metastasis. Targeting mitochondrial fission presents a promising strategy for breast cancer therapy.106,107

Mitochondrial fusion, in contrast, involves the merging of the outer and inner membranes of two distinct mitochondria, ensuring the maintenance of a healthy mitochondrial pool through quality control mechanisms. In contrast, mitochondrial fission involves the splitting of a single mitochondrion into two. The dynamic balance of fusion and fission generates interconnected mitochondrial networks that support cellular energy needs and function.108 Mitochondrial fusion and fission are regulated by key genes, including (1) mitofusins (MFN1, MFN2) for outer membrane fusion, (2) OPA1/mitochondrial genome maintenance 1 for inner membrane fusion, and (3) Drp1/Dnm1 for membrane division. These GTP-hydrolyzing GTPases from the dynamin superfamily are regulated through interactions with binding partners, playing a critical role in both normal and dysfunctional cellular processes.109,110 During mitochondrial fusion, MFN1 and MFN2 facilitate outer mitochondrial membrane fusion, while OPA1 mediates inner mitochondrial membrane fusion.111 Evidence shows that the M2 subtype of pyruvate kinase (PKM2) binds to MFN2, promoting mitochondrial fusion and oxidative phosphorylation while reducing glycolysis in cancer cells. This metabolic shift inhibits cancer cell proliferation and metastasis.112 mTOR, a serine/threonine kinase in the PIKK family, regulates mitochondrial fusion and division by modulating fusion protein expression. Through the mTOR-MFN2-PKM2 pathway, mTOR promotes the interaction between MFN2 and PKM2, which is essential for glycolysis and oxidative phosphorylation in breast cancer cells.113 High OPA1 expression correlates with poor prognosis in breast cancer, whereas OPA1 inhibition reduces tumor growth, aggressiveness, and neovascularization, thereby inhibiting metastasis.114

Additionally, mitochondrial fusion exhibits anti-apoptotic activity, with fusion stimulation significantly delaying apoptosis.115 This process plays a critical role in breast cancer progression and presents promising therapeutic opportunities.116,117

Mitochondrial dynamics, including fission and fusion, are closely associated with mitophagy, which facilitates the selective autophagic elimination of mitochondria. Mitophagy plays a vital role in cellular homeostasis by encapsulating and degrading damaged or dysfunctional mitochondria, preventing the accumulation of harmful protein aggregates and excessive ROS.118 This process maintains a healthy mitochondrial network, maintaining bioenergetic capacity and supporting overall cellular function. Mitophagy relies on mitochondrial fission, as evidenced by its inhibition through a dominant-negative Drp1 mutant.119 The selective removal of photodamaged mitochondria highlights fission’s role as a quality control mechanism, isolating damaged segments for autophagic degradation while preserving a functional mitochondrial network.120 Recent studies on breast cancer have highlighted the role of the PINK1 and Parkin genes in mitochondrial quality control through the targeted elimination of damaged mitochondria.121,122 PINK1 recruits the E3 ubiquitin ligase Parkin from the cytosol to damaged mitochondria, where Parkin ubiquitinates outer mitochondrial membrane proteins, marking the mitochondrion for autophagic degradation. In tumor cells, mitochondrial F1F0 ATPase can use glycolytic ATP to restore membrane potential, and mitochondrial fusion can compensate for missing components, rescuing impaired organelles.123 However, mitochondrial fission can overwhelm these compensatory mechanisms, leading to complete depolarization and initiating PINK1-mediated mitophagy. This intricate process highlights the balance between mitochondrial dynamics, damage sensing, and quality control.124

Additionally, accumulating evidence links intracellular ROS to tumor progression. Mitophagy plays a tumor-suppressive role by removing dysfunctional mitochondria and reducing ROS production in tumor cells.125 For instance, ULK1 depletion in breast cancer cells impairs mitophagy, leading to excessive ROS accumulation. This aberrant ROS production activates the NLRP3 inflammasome, promoting breast cancer bone metastasis.126 Recent studies have highlighted the role of BRCA1 as a tumor suppressor in regulating mitophagy. BRCA1 deficiency disrupts mitophagy by inhibiting mitochondrial fission, resulting in damaged mitochondria and elevated ROS levels, which activate the NLRP3 inflammasome, thereby promoting breast cancer metastasis.127 Similarly, BNIP3 loss impairs mitophagy, leading to ROS accumulation and enhanced growth and metastasis of TNBC.128 These findings suggest that restoring mitophagy to reduce ROS could be a promising therapeutic strategy. Interestingly, certain drugs and compounds activate mitophagy to exert antitumor effects, further underscoring its role in cancer suppression.129,130 However, under metabolic stress or nutrient deprivation, mitophagy may promote tumor survival by enabling cancer cells to adapt. For instance, MRPL52 upregulation activates protective mitophagy via the PINK1/Parkin pathway, helping breast cancer cells survive hypoxia.131 Similarly, MUC1, located on the mitochondrial outer membrane, interacts with ATAD3A to promote PINK1-dependent mitophagy. Inhibiting either MUC1 or mitophagy alone reduces tumor proliferation, but combined inhibition demonstrates enhanced suppression of breast tumor growth both in vitro and in vivo.132 These findings reveal the dual role of mitophagy in breast cancer, acting as a tumor suppressor by reducing ROS and eliminating damaged mitochondria and as a survival mechanism under adverse conditions.133 Combining mitophagy modulation with therapies targeting mitochondrial fission and fusion offers a promising avenue for breast cancer treatment.

Mitochondrial dynamics as a therapeutic target in breast cancer metastasis

Mitochondrial dynamics, including fusion, fission, and mitophagy, are integral to tumor signaling pathways and mitochondrial metabolism, playing a critical role in cellular adaptability.134 In normal cells, a balance between mitochondrial fusion and fission maintains homeostasis. However, in cancer cells, this balance is disrupted, driving proliferation, metastasis, and cancer stem cell maintenance (Fig. 1).135 In TNBC, studies have investigated alterations in mitochondrial dynamics, revealing significant variability in their expression.105,136 However, the connection between mitochondrial dynamics and metabolic phenotypes remains inconsistent across different breast cancer models and studies.137,138 Mitophagy, the selective autophagy of mitochondria, is essential for maintaining intracellular stability by removing damaged or dysfunctional mitochondria, thereby preserving cellular homeostasis.139 Targeted mitochondrial therapy shows promise, as mitochondrial fission is linked to poor breast cancer prognosis. Inhibiting fission has demonstrated the potential to reduce metastasis, presenting a novel therapeutic strategy.140 The small molecule Mdivi-1 binds to Drp1, suppressing mitochondrial fission, lowering ATP production, and inducing apoptosis in breast cancer cells.141 Additionally, the peptide inhibitor P110 blocks the Drp1-Fis1 interaction, effectively halting fission. With fewer side effects than Mdivi-1, P110 holds greater potential for clinical application.142 Promoting mitochondrial fusion has been shown to enhance TNBC sensitivity to chemotherapy. In mice with MDA-MB-231 breast cancer cells, injections of P-Mito or Mdivi-1 increased sensitivity to doxorubicin to a similar extent.143 The poor prognosis of TNBC is linked to its high aggressiveness, partly driven by elevated ROS levels. Studies indicate that reducing ROS content in cancer cells can attenuate metastasis.144 Mitochondrial peroxide-reducing proteins, particularly Prdx3 and Prdx5, are often upregulated in various cancers, contributing to drug resistance.145,146 Mitochondrial transplantation also represents a novel approach to breast cancer treatment, potentially mitigating the toxic side effects of conventional therapies.147 However, the complexity of this method currently limits its clinical application. Table 1 provides an overview of the key regulators and emerging therapeutic strategies targeting mitochondrial fission, fusion, and mitophagy in breast cancer.86,116,122,127,129,130,148–166

Mitochondrial dynamics in normal and cancer cells.
Fig. 1  Mitochondrial dynamics in normal and cancer cells.

Mitochondrial dynamics, governed by the interplay of fission and fusion proteins, are essential for maintaining cellular function and homeostasis. Fission facilitates glycolysis, mitophagy, apoptosis, and cell division, while fusion supports ATP production and reactive oxygen species (ROS) generation through oxidative phosphorylation (OXPHOS). In normal cells, the equilibrium between mitochondrial fusion and fission ensures proper cellular health and function. However, in cancer cells, this balance is disrupted, with a shift toward increased fission. This imbalance fuels uncontrolled proliferation, metastasis, and the preservation of cancer stem cell phenotypes, underscoring the pivotal role of altered mitochondrial dynamics in cancer progression. ATP, adenosine triphosphate; mROS, mitochondrial reactive oxygen species. (The figure is adopted from Gundamaraju et al.,135 2021.)

Table 1

Current therapies or regulators that actively modulate mitochondrial fission, fusion, and mitophagy in breast cancer

Targeted therapy/drug candidateFunctional targetCancer targetReferences
Gene knockdownFissionBreast cancer148
HypoxiaFissionMDA-MB-231149
CisplatinFissionTNBC150
Mdivi-1 (mito division inhibitor 1)FissionBreast and lung151
PaclitaxelFissionBreast cancer152
IsotoosendaninFissionTNBC153
P110 (peptide inhibitor)FissionBreast cancer154
DynsoreFissionTNBC155
BRCA1 (breast cancer associated gene 1)FusionMDA-MB-231127
RACGAP1/LncRNA (long non-coding RNAs)Fission and fusionMCF-7 and MDA-MB-23186
MiR-195 (microRNA 195)Fission and fusionMCF-7 and MDA-MB-231116
MiDs (mito dynamics)Fission and fusionMCF-7156
PKM2 (pyruvate kinase M 2)FusionMCF-7157
SilibininFusion and mitophagyMCF-7 and MDA-MB-231158
Polyphyllin IMitophagyMCF-7 and MDA-MB-231159
WarangaloneMitophagyMCF-7 and MDA-MB-231122
FlubendazoleMitophagyMCF-7 and MDA-MB-231160
TaloxifeneMitophagyMCF-7161
CepharanthineMitophagyMDA-MB-231162
KaempferolMitophagyMCF-10AT130
CostunolideMitophagyMCF-7163
Copper complex CPT8MitophagyMDA-MB-231164
Ru-TPE-PPh complexMitophagyMDA-MB-231165
BaicaleinMitophagyMDA-MB-231166
Acid ground nano-realgar processed productMitophagyMDA-MB-435S129

Clinical significance and prospects

Recent research underscores the pivotal role of mitochondrial dynamics and bioenergetics in a wide range of diseases, extending far beyond cancer progression.167–170 Cancer cells rely on altered mitochondrial function and oxidative metabolism to fuel proliferation and tumorigenesis.171 They demonstrate elevated oxidative phosphorylation and disrupted mitochondrial dynamics, including increased fission, decreased fusion, and enhanced mitophagy, making these processes promising therapeutic targets. While interventions targeting oxidative phosphorylation, the TCA cycle, and mitochondrial dynamics have been investigated, their clinical efficacy is often undermined by drug resistance.172 Understanding mitochondrial dynamics and bioenergetics in breast cancer is key to advancing personalized treatments. Future research should focus on unraveling the roles of mitochondria in cellular metabolism, cancer progression, and therapy resistance. Insights into metabolic flexibility and the regulation of mitochondrial function are crucial for enhancing therapeutic efficacy. Additionally, exploring the physiological factors influencing mitochondrial fitness will be vital for optimizing targeted therapies and improving outcomes for breast cancer patients.

In metastatic breast cancer, an imbalance in mitochondrial dynamics plays a pivotal role.78,173 Experimental studies reveal that metastatic breast cancer cells exhibit heightened mitochondrial fission, reduced fusion, and increased fragmentation. These alterations underscore the potential of targeting mitochondrial processes in breast cancer treatment.46,174 Despite significant progress in understanding mitochondrial dynamics and mitophagy in tumor progression, key questions remain unresolved. Emerging evidence highlights the complex and context-dependent roles of these processes, which vary by tumor type, stage, and microenvironment. Further research is essential to elucidate how mitochondrial dynamics drive metastatic cancer progression and how these mechanisms can be targeted with greater precision. Advancing knowledge in this area holds the potential to develop more effective, targeted therapies, addressing current treatment challenges and improving outcomes for breast cancer patients.

Conclusions

This review highlights the pivotal role of mitochondrial dynamics and bioenergetics in driving breast cancer progression and metastasis. By elucidating the intricate interplay between mitochondrial functions—biogenesis, turnover, fission, fusion, mitophagy, oxidative stress regulation, metabolism, and ATP synthesis—it reveals the critical regulatory mechanisms that fuel cancer cell adaptation. Targeting these processes presents promising therapeutic opportunities, paving the way for mitochondria-focused strategies to enhance drug efficacy and improve treatment outcomes. Ultimately, this comprehensive analysis underscores the potential of mitochondrial-directed interventions as a transformative approach in breast cancer therapy.

Declarations

Acknowledgement

None.

Funding

This work did not receive any funding.

Conflict of interest

The author has no conflict of interest related to this publication.

Authors’ contributions

BR is the sole author of the manuscript.

References

  1. Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: Still emerging. Cell Metab 2022;34(3):355-377 View Article PubMed/NCBI
  2. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv 2016;2(5):e1600200 View Article PubMed/NCBI
  3. McGuirk S, Audet-Delage Y, St-Pierre J. Metabolic Fitness and Plasticity in Cancer Progression. Trends Cancer 2020;6(1):49-61 View Article PubMed/NCBI
  4. Park JH, Pyun WY, Park HW. Cancer Metabolism: Phenotype, Signaling and Therapeutic Targets. Cells 2020;9(10):2308 View Article PubMed/NCBI
  5. Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, et al. Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (2020) 2023;4(2):e218 View Article PubMed/NCBI
  6. WARBURG O. On the origin of cancer cells. Science 1956;123(3191):309-314 View Article PubMed/NCBI
  7. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009;324(5930):1029-1033 View Article PubMed/NCBI
  8. Zamzami N, Marchetti P, Castedo M, Decaudin D, Macho A, Hirsch T, et al. Sequential reduction of mitochondrial transmembrane potential and generation of reactive oxygen species in early programmed cell death. J Exp Med 1995;182(2):367-377 View Article PubMed/NCBI
  9. Hoy AJ, Nagarajan SR, Butler LM. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat Rev Cancer 2021;21(12):753-766 View Article PubMed/NCBI
  10. Schiliro C, Firestein BL. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021;10(5):1056 View Article PubMed/NCBI
  11. Wang L, Zhang S, Wang X. The Metabolic Mechanisms of Breast Cancer Metastasis. Front Oncol 2020;10:602416 View Article PubMed/NCBI
  12. Ruidas B, Sur TK, Das Mukhopadhyay C, Sinha K, Som Chaudhury S, Sharma P, et al. Quercetin: a silent retarder of fatty acid oxidation in breast cancer metastasis through steering of mitochondrial CPT1. Breast Cancer 2022;29(4):748-760 View Article PubMed/NCBI
  13. Bartlome S, Berry CC. Recent insights into the effects of metabolism on breast cancer cell dormancy. Br J Cancer 2022;127(8):1385-1393 View Article PubMed/NCBI
  14. Sankofi BM, Valencia-Rincón E, Sekhri M, Ponton-Almodovar AL, Bernard JJ, Wellberg EA. The impact of poor metabolic health on aggressive breast cancer: adipose tissue and tumor metabolism. Front Endocrinol (Lausanne) 2023;14:1217875 View Article PubMed/NCBI
  15. Young CM, Beziaud L, Dessen P, Madurga Alonso A, Santamaria-Martínez A, Huelsken J. Metabolic dependencies of metastasis-initiating cells in female breast cancer. Nat Commun 2023;14(1):7076 View Article PubMed/NCBI
  16. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012;490(7418):61-70 View Article PubMed/NCBI
  17. Anders CK, Abramson V, Tan T, Dent R. The Evolution of Triple-Negative Breast Cancer: From Biology to Novel Therapeutics. Am Soc Clin Oncol Educ Book 2016;35:34-42 View Article PubMed/NCBI
  18. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer 2007;109(9):1721-1728 View Article PubMed/NCBI
  19. Cai SL, Liu JJ, Liu YX, Yu SH, Liu X, Lin XQ, et al. Characteristics of recurrence, predictors for relapse and prognosis of rapid relapse triple-negative breast cancer. Front Oncol 2023;13:1119611 View Article PubMed/NCBI
  20. Baenke F, Peck B, Miess H, Schulze A. Hooked on fat: the role of lipid synthesis in cancer metabolism and tumour development. Dis Model Mech 2013;6(6):1353-1363 View Article PubMed/NCBI
  21. Monaco ME. Fatty acid metabolism in breast cancer subtypes. Oncotarget 2017;8(17):29487-29500 View Article PubMed/NCBI
  22. Talari NK, Mattam U, Meher NK, Paripati AK, Mahadev K, Krishnamoorthy T, et al. Lipid-droplet associated mitochondria promote fatty-acid oxidation through a distinct bioenergetic pattern in male Wistar rats. Nat Commun 2023;14(1):766 View Article PubMed/NCBI
  23. Grasso D, Zampieri LX, Capelôa T, Van de Velde JA, Sonveaux P. Mitochondria in cancer. Cell Stress 2020;4(6):114-146 View Article PubMed/NCBI
  24. Chen M, Huang J. The expanded role of fatty acid metabolism in cancer: new aspects and targets. Precis Clin Med 2019;2(3):183-191 View Article PubMed/NCBI
  25. Xu S, Chen T, Dong L, Li T, Xue H, Gao B, et al. Fatty acid synthase promotes breast cancer metastasis by mediating changes in fatty acid metabolism. Oncol Lett 2021;21(1):27 View Article PubMed/NCBI
  26. Ruidas B, Choudhury N, Chaudhury SS, Sur TK, Bhowmick S, Saha A, et al. Precision targeting of fat metabolism in triple negative breast cancer with a biotinylated copolymer. J Mater Chem B 2025;13(4):1363-1371 View Article PubMed/NCBI
  27. Azam A, Sounni NE. Lipid Metabolism Heterogeneity and Crosstalk with Mitochondria Functions Drive Breast Cancer Progression and Drug Resistance. Cancers (Basel) 2022;14(24):6267 View Article PubMed/NCBI
  28. Mallick R, Bhowmik P, Duttaroy AK. Targeting fatty acid uptake and metabolism in cancer cells: A promising strategy for cancer treatment. Biomed Pharmacother 2023;167:115591 View Article PubMed/NCBI
  29. Liesa M, Shirihai OS. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab 2013;17(4):491-506 View Article PubMed/NCBI
  30. Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol 2011;13(5):589-598 View Article PubMed/NCBI
  31. Mollica MP, Lionetti L, Crescenzo R, D’Andrea E, Ferraro M, Liverini G, et al. Heterogeneous bioenergetic behaviour of subsarcolemmal and intermyofibrillar mitochondria in fed and fasted rats. Cell Mol Life Sci 2006;63(3):358-366 View Article PubMed/NCBI
  32. Wang Z, Jiang Q, Dong C. Metabolic reprogramming in triple-negative breast cancer. Cancer Biol Med 2020;17(1):44-59 View Article PubMed/NCBI
  33. Avagliano A, Ruocco MR, Aliotta F, Belviso I, Accurso A, Masone S, et al. Mitochondrial Flexibility of Breast Cancers: A Growth Advantage and a Therapeutic Opportunity. Cells 2019;8(5):401 View Article PubMed/NCBI
  34. Lunetti P, Di Giacomo M, Vergara D, De Domenico S, Maffia M, Zara V, et al. Metabolic reprogramming in breast cancer results in distinct mitochondrial bioenergetics between luminal and basal subtypes. FEBS J 2019;286(4):688-709 View Article PubMed/NCBI
  35. Porporato PE, Filigheddu N, Pedro JMB, Kroemer G, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res 2018;28(3):265-280 View Article PubMed/NCBI
  36. Bao X, Zhang J, Huang G, Yan J, Xu C, Dou Z, et al. The crosstalk between HIFs and mitochondrial dysfunctions in cancer development. Cell Death Dis 2021;12(2):215 View Article PubMed/NCBI
  37. Ruidas B, Sur TK, Pal K, Som Chaudhury S, Prasad P, Sinha K, et al. Herbometallic nano-drug inducing metastatic growth inhibition in breast cancer through intracellular energy depletion. Mol Biol Rep 2020;47(5):3745-3763 View Article PubMed/NCBI
  38. Wang SF, Tseng LM, Lee HC. Role of mitochondrial alterations in human cancer progression and cancer immunity. J Biomed Sci 2023;30(1):61 View Article PubMed/NCBI
  39. Spinelli JB, Haigis MC. The multifaceted contributions of mitochondria to cellular metabolism. Nat Cell Biol 2018;20(7):745-754 View Article PubMed/NCBI
  40. Chan DC. Mitochondrial Dynamics and Its Involvement in Disease. Annu Rev Pathol 2020;15:235-259 View Article PubMed/NCBI
  41. Grandemange S, Herzig S, Martinou JC. Mitochondrial dynamics and cancer. Semin Cancer Biol 2009;19(1):50-56 View Article PubMed/NCBI
  42. Ghosh P, Vidal C, Dey S, Zhang L. Mitochondria Targeting as an Effective Strategy for Cancer Therapy. Int J Mol Sci 2020;21(9):3363 View Article PubMed/NCBI
  43. Sastre-Serra J, Nadal-Serrano M, Pons DG, Roca P, Oliver J. Mitochondrial dynamics is affected by 17β-estradiol in the MCF-7 breast cancer cell line. Effects on fusion and fission related genes. Int J Biochem Cell Biol 2012;44(11):1901-1905 View Article PubMed/NCBI
  44. Humphries BA, Cutter AC, Buschhaus JM, Chen YC, Qyli T, Palagama DSW, et al. Enhanced mitochondrial fission suppresses signaling and metastasis in triple-negative breast cancer. Breast Cancer Res 2020;22(1):60 View Article PubMed/NCBI
  45. Chang JC, Chang HS, Wu YC, Cheng WL, Lin TT, Chang HJ, et al. Mitochondrial transplantation regulates antitumour activity, chemoresistance and mitochondrial dynamics in breast cancer. J Exp Clin Cancer Res 2019;38(1):30 View Article PubMed/NCBI
  46. Weiner-Gorzel K, Murphy M. Mitochondrial dynamics, a new therapeutic target for Triple Negative Breast Cancer. Biochim Biophys Acta Rev Cancer 2021;1875(2):188518 View Article PubMed/NCBI
  47. Zhao J, Zhang J, Yu M, Xie Y, Huang Y, Wolff DW, et al. Mitochondrial dynamics regulates migration and invasion of breast cancer cells. Oncogene 2013;32(40):4814-4824 View Article PubMed/NCBI
  48. Li H, Ning S, Ghandi M, Kryukov G, Gopal S, Deik A, et al. The landscape of cancer cell line metabolism. Nat Med 2019;25(5):850-860 View Article PubMed/NCBI
  49. Ortmayr K, Dubuis S, Zampieri M. Metabolic profiling of cancer cells reveals genome-wide crosstalk between transcriptional regulators and metabolism. Nat Commun 2019;10(1):1841 View Article PubMed/NCBI
  50. Elia I, Doglioni G, Fendt SM. Metabolic Hallmarks of Metastasis Formation. Trends Cell Biol 2018;28(8):673-684 View Article PubMed/NCBI
  51. Zheng P, Zhou C, Lu L, Liu B, Ding Y. Elesclomol: a copper ionophore targeting mitochondrial metabolism for cancer therapy. J Exp Clin Cancer Res 2022;41(1):271 View Article PubMed/NCBI
  52. Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol 1927;8(6):519-530 View Article PubMed/NCBI
  53. Zamzami N, Susin SA, Marchetti P, Hirsch T, Gómez-Monterrey I, Castedo M, et al. Mitochondrial control of nuclear apoptosis. J Exp Med 1996;183(4):1533-1544 View Article PubMed/NCBI
  54. Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov 2022;12(1):31-46 View Article PubMed/NCBI
  55. Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, et al. Impact of cancer metabolism on therapy resistance - Clinical implications. Drug Resist Updat 2021;59:100797 View Article PubMed/NCBI
  56. Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov 2022;21(2):141-162 View Article PubMed/NCBI
  57. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 2009;324(5930):1076-1080 View Article PubMed/NCBI
  58. Morad SA, Cabot MC. Ceramide-orchestrated signalling in cancer cells. Nat Rev Cancer 2013;13(1):51-65 View Article PubMed/NCBI
  59. Wymann MP, Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol 2008;9(2):162-176 View Article PubMed/NCBI
  60. Snaebjornsson MT, Janaki-Raman S, Schulze A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab 2020;31(1):62-76 View Article PubMed/NCBI
  61. Li H, Feng Z, He ML. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics 2020;10(16):7053-7069 View Article PubMed/NCBI
  62. Kuhajda FP, Jenner K, Wood FD, Hennigar RA, Jacobs LB, Dick JD, et al. Fatty acid synthesis: a potential selective target for antineoplastic therapy. Proc Natl Acad Sci U S A 1994;91(14):6379-6383 View Article PubMed/NCBI
  63. Abramson HN. The lipogenesis pathway as a cancer target. J Med Chem 2011;54(16):5615-5638 View Article PubMed/NCBI
  64. Santos CR, Schulze A. Lipid metabolism in cancer. FEBS J 2012;279(15):2610-2623 View Article PubMed/NCBI
  65. Koundouros N, Poulogiannis G. Reprogramming of fatty acid metabolism in cancer. Br J Cancer 2020;122(1):4-22 View Article PubMed/NCBI
  66. Knobloch M, Braun SM, Zurkirchen L, von Schoultz C, Zamboni N, Araúzo-Bravo MJ, et al. Metabolic control of adult neural stem cell activity by Fasn-dependent lipogenesis. Nature 2013;493(7431):226-230 View Article PubMed/NCBI
  67. Jerby L, Wolf L, Denkert C, Stein GY, Hilvo M, Oresic M, et al. Metabolic associations of reduced proliferation and oxidative stress in advanced breast cancer. Cancer Res 2012;72(22):5712-5720 View Article PubMed/NCBI
  68. Ruidas B, Som Chaudhury S, Pal K, Sarkar PK, Das Mukhopadhyay C. A novel herbometallic nanodrug has the potential for antibacterial and anticancer activity through oxidative damage. Nanomedicine (Lond) 2019;14(9):1173-1189 View Article PubMed/NCBI
  69. Bartman CR, Weilandt DR, Shen Y, Lee WD, Han Y, TeSlaa T, et al. Slow TCA flux and ATP production in primary solid tumours but not metastases. Nature 2023;614(7947):349-357 View Article PubMed/NCBI
  70. Liu A, Curran MA. Tumor hypermetabolism confers resistance to immunotherapy. Semin Cancer Biol 2020;65:155-163 View Article PubMed/NCBI
  71. Zheng P, Lin Z, Ding Y, Duan S. Targeting the dynamics of cancer metabolism in the era of precision oncology. Metabolism 2023;145:155615 View Article PubMed/NCBI
  72. Uslu C, Kapan E, Lyakhovich A. Cancer resistance and metastasis are maintained through oxidative phosphorylation. Cancer Lett 2024;587:216705 View Article PubMed/NCBI
  73. Behnam B, Taghizadeh-Hesary F. Mitochondrial Metabolism: A New Dimension of Personalized Oncology. Cancers (Basel) 2023;15(16):4058 View Article PubMed/NCBI
  74. Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, et al. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024;23(1):261 View Article PubMed/NCBI
  75. Liu B, Zhang X. Metabolic Reprogramming Underlying Brain Metastasis of Breast Cancer. Front Mol Biosci 2021;8:791927 View Article PubMed/NCBI
  76. Ruidas B. Mitochondrial Lipid metabolism in metastatic breast cancer. Mitochondrial Communications 2024;2(1):58-66 View Article PubMed/NCBI
  77. Wang Y, Harada-Shoji N, Kitamura N, Yamazaki Y, Ebata A, Amari M, et al. Mitochondrial dynamics as a novel treatment strategy for triple-negative breast cancer. Cancer Med 2024;13(2):e6987 View Article PubMed/NCBI
  78. Minarrieta L, Annis MG, Audet-Delage Y, Kuasne H, Pacis A, St-Louis C, et al. Mitochondrial elongation impairs breast cancer metastasis. Sci Adv 2024;10(45):eadm8212 View Article PubMed/NCBI
  79. Liu Y, Sun Y, Guo Y, Shi X, Chen X, Feng W, et al. An Overview: The Diversified Role of Mitochondria in Cancer Metabolism. Int J Biol Sci 2023;19(3):897-915 View Article PubMed/NCBI
  80. Chen W, Zhao H, Li Y. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct Target Ther 2023;8(1):333 View Article PubMed/NCBI
  81. Chen H, Chan DC. Mitochondrial Dynamics in Regulating the Unique Phenotypes of Cancer and Stem Cells. Cell Metab 2017;26(1):39-48 View Article PubMed/NCBI
  82. Xing J, Qi L, Liu X, Shi G, Sun X, Yang Y. Roles of mitochondrial fusion and fission in breast cancer progression: a systematic review. World J Surg Oncol 2022;20(1):331 View Article PubMed/NCBI
  83. Mostafavi S, Eskandari N. Mitochondrion: Main organelle in orchestrating cancer escape from chemotherapy. Cancer Rep (Hoboken) 2024;7(2):e1942 View Article PubMed/NCBI
  84. Scheid AD, Beadnell TC, Welch DR. Roles of mitochondria in the hallmarks of metastasis. Br J Cancer 2021;124(1):124-135 View Article PubMed/NCBI
  85. Döhla J, Kuuluvainen E, Gebert N, Amaral A, Englund JI, Gopalakrishnan S, et al. Metabolic determination of cell fate through selective inheritance of mitochondria. Nat Cell Biol 2022;24(2):148-154 View Article PubMed/NCBI
  86. Ren K, Zhou D, Wang M, Li E, Hou C, Su Y, et al. RACGAP1 modulates ECT2-Dependent mitochondrial quality control to drive breast cancer metastasis. Exp Cell Res 2021;400(1):112493 View Article PubMed/NCBI
  87. Boulton DP, Caino MC. Mitochondrial Fission and Fusion in Tumor Progression to Metastasis. Front Cell Dev Biol 2022;10:849962 View Article PubMed/NCBI
  88. Manjunath M, Ravindran F, Sharma S, Siddiqua H, Raghavan SC, Choudhary B. Disarib, a Specific BCL2 Inhibitor, Induces Apoptosis in Triple-Negative Breast Cancer Cells and Impedes Tumour Progression in Xenografts by Altering Mitochondria-Associated Processes. Int J Mol Sci 2024;25(12):6485 View Article PubMed/NCBI
  89. Kamerkar SC, Kraus F, Sharpe AJ, Pucadyil TJ, Ryan MT. Dynamin-related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat Commun 2018;9(1):5239 View Article PubMed/NCBI
  90. Smirnova E, Griparic L, Shurland DL, van der Bliek AM. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol Biol Cell 2001;12(8):2245-2256 View Article PubMed/NCBI
  91. Kyriakoudi S, Drousiotou A, Petrou PP. When the Balance Tips: Dysregulation of Mitochondrial Dynamics as a Culprit in Disease. Int J Mol Sci 2021;22(9):4617 View Article PubMed/NCBI
  92. Yang Z, Wang L, Yang C, Pu S, Guo Z, Wu Q, et al. Mitochondrial Membrane Remodeling. Front Bioeng Biotechnol 2021;9:786806 View Article PubMed/NCBI
  93. Lu G, Lai Y, Wang T, Lin W, Lu J, Ma Y, et al. Mitochondrial fission regulator 2 (MTFR2) promotes growth, migration, invasion and tumour progression in breast cancer cells. Aging (Albany NY) 2019;11(22):10203-10219 View Article PubMed/NCBI
  94. Alalawy AI, Sakran M, Alzuaibr FM, Alotaibi MA, El-Hefnawy ME, Hazazi AY, et al. Inhibition of Drp1 orchestrates the responsiveness of breast cancer cells to paclitaxel but insignificantly relieves paclitaxel-related ovarian damage in mice. Sci Rep 2023;13(1):22782 View Article PubMed/NCBI
  95. Silva-Pavez E, Mendoza E, Morgado-Cáceres P, Ahumada-Castro U, Bustos G, Kangme-Encalada M, et al. Mitochondrial division inhibitor (mdivi-1) induces extracellular matrix (ECM)-detachment of viable breast cancer cells by a DRP1-independent mechanism. Sci Rep 2024;14(1):14178 View Article PubMed/NCBI
  96. You L, Wang M, Liu X, Song M, Zhou J, Feng J, et al. DRP1: shedding light on the complex nexus of mitochondrial fission and breast cancer. Future Oncol 2025;21(5):593-603 View Article PubMed/NCBI
  97. Peiris-Pagès M, Bonuccelli G, Sotgia F, Lisanti MP. Mitochondrial fission as a driver of stemness in tumor cells: mDIVI1 inhibits mitochondrial function, cell migration and cancer stem cell (CSC) signalling. Oncotarget 2018;9(17):13254-13275 View Article PubMed/NCBI
  98. Xie Q, Wu Q, Horbinski CM, Flavahan WA, Yang K, Zhou W, et al. Mitochondrial control by DRP1 in brain tumor initiating cells. Nat Neurosci 2015;18(4):501-510 View Article PubMed/NCBI
  99. Prieto J, León M, Ponsoda X, Sendra R, Bort R, Ferrer-Lorente R, et al. Early ERK1/2 activation promotes DRP1-dependent mitochondrial fission necessary for cell reprogramming. Nat Commun 2016;7:11124 View Article PubMed/NCBI
  100. Katajisto P, Döhla J, Chaffer CL, Pentinmikko N, Marjanovic N, Iqbal S, et al. Stem cells. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 2015;348(6232):340-343 View Article PubMed/NCBI
  101. Lunova M, Jirsa M, Dejneka A, Sullivan GJ, Lunov O. Mechanical regulation of mitochondrial morphodynamics in cancer cells by extracellular microenvironment. Biomater Biosyst 2024;14:100093 View Article PubMed/NCBI
  102. Humphries BA, Zhang A, Buschhaus JM, Bevoor A, Farfel A, Rajendran S, et al. Enhanced mitochondrial fission inhibits triple-negative breast cancer cell migration through an ROS-dependent mechanism. iScience 2023;26(6):106788 View Article PubMed/NCBI
  103. Rosdah AA, K Holien J, Delbridge LM, Dusting GJ, Lim SY. Mitochondrial fission - a drug target for cytoprotection or cytodestruction?. Pharmacol Res Perspect 2016;4(3):e00235 View Article PubMed/NCBI
  104. Lee KM, Giltnane JM, Balko JM, Schwarz LJ, Guerrero-Zotano AL, Hutchinson KE, et al. MYC and MCL1 Cooperatively Promote Chemotherapy-Resistant Breast Cancer Stem Cells via Regulation of Mitochondrial Oxidative Phosphorylation. Cell Metab 2017;26(4):633-647.e7 View Article PubMed/NCBI
  105. Chen L, Zhang J, Lyu Z, Chen Y, Ji X, Cao H, et al. Positive feedback loop between mitochondrial fission and Notch signaling promotes survivin-mediated survival of TNBC cells. Cell Death Dis 2018;9(11):1050 View Article PubMed/NCBI
  106. Yi L, Shang XJ, Lv L, Wang Y, Zhang J, Quan C, et al. Cadmium-induced apoptosis of Leydig cells is mediated by excessive mitochondrial fission and inhibition of mitophagy. Cell Death Dis 2022;13(11):928 View Article PubMed/NCBI
  107. Yu S, Cao Z, Cai F, Yao Y, Chang X, Wang X, et al. ADT-OH exhibits anti-metastatic activity on triple-negative breast cancer by combinatorial targeting of autophagy and mitochondrial fission. Cell Death Dis 2024;15(6):463 View Article PubMed/NCBI
  108. Adebayo M, Singh S, Singh AP, Dasgupta S. Mitochondrial fusion and fission: The fine-tune balance for cellular homeostasis. FASEB J 2021;35(6):e21620 View Article PubMed/NCBI
  109. Ramaiah P, Patra I, Abbas A, Fadhil AA, Abohassan M, Al-Qaim ZH, et al. Mitofusin-2 in cancer: Friend or foe?. Arch Biochem Biophys 2022;730:109395 View Article PubMed/NCBI
  110. Jasra IT, Cuesta-Gomez N, Verhoeff K, Marfil-Garza BA, Dadheech N, Shapiro AMJ. Mitochondrial regulation in human pluripotent stem cells during reprogramming and β cell differentiation. Front Endocrinol (Lausanne) 2023;14:1236472 View Article PubMed/NCBI
  111. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol 2010;11(12):872-884 View Article PubMed/NCBI
  112. Li T, Han J, Jia L, Hu X, Chen L, Wang Y. PKM2 coordinates glycolysis with mitochondrial fusion and oxidative phosphorylation. Protein Cell 2019;10(8):583-594 View Article PubMed/NCBI
  113. de la Cruz López KG, Toledo Guzmán ME, Sánchez EO, García Carrancá A. mTORC1 as a Regulator of Mitochondrial Functions and a Therapeutic Target in Cancer. Front Oncol 2019;9:1373 View Article PubMed/NCBI
  114. Zamberlan M, Boeckx A, Muller F, Vinelli F, Ek O, Vianello C, et al. Inhibition of the mitochondrial protein Opa1 curtails breast cancer growth. J Exp Clin Cancer Res 2022;41(1):95 View Article PubMed/NCBI
  115. Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell 2011;21(1):92-101 View Article PubMed/NCBI
  116. Purohit PK, Edwards R, Tokatlidis K, Saini N. MiR-195 regulates mitochondrial function by targeting mitofusin-2 in breast cancer cells. RNA Biol 2019;16(7):918-929 View Article PubMed/NCBI
  117. Fu Y, Dong W, Xu Y, Li L, Yu X, Pang Y, et al. Targeting mitochondrial dynamics by AZD5363 in triple-negative breast cancer MDA-MB-231 cell-derived spheres. Naunyn Schmiedebergs Arch Pharmacol 2023;396(10):2545-2553 View Article PubMed/NCBI
  118. Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J 2008;27(2):433-446 View Article PubMed/NCBI
  119. Fan RZ, Sportelli C, Lai Y, Salehe SS, Pinnell JR, Brown HJ, et al. A partial Drp1 knockout improves autophagy flux independent of mitochondrial function. Mol Neurodegener 2024;19(1):26 View Article PubMed/NCBI
  120. Kim I, Lemasters JJ. Mitophagy selectively degrades individual damaged mitochondria after photoirradiation. Antioxid Redox Signal 2011;14(10):1919-1928 View Article PubMed/NCBI
  121. Li J, Xu X, Huang H, Li L, Chen J, Ding Y, et al. Pink1 promotes cell proliferation and affects glycolysis in breast cancer. Exp Biol Med (Maywood) 2022;247(12):985-995 View Article PubMed/NCBI
  122. Mao L, Liu H, Zhang R, Deng Y, Hao Y, Liao W, et al. PINK1/Parkin-mediated mitophagy inhibits warangalone-induced mitochondrial apoptosis in breast cancer cells. Aging (Albany NY) 2021;13(9):12955-12972 View Article PubMed/NCBI
  123. Lhuissier C, Desquiret-Dumas V, Girona A, Alban J, Faure J, Cassereau J, et al. Mitochondrial F0F1-ATP synthase governs the induction of mitochondrial fission. iScience 2024;27(5):109808 View Article PubMed/NCBI
  124. Huang Q, Zhan L, Cao H, Li J, Lyu Y, Guo X, et al. Increased mitochondrial fission promotes autophagy and hepatocellular carcinoma cell survival through the ROS-modulated coordinated regulation of the NFKB and TP53 pathways. Autophagy 2016;12(6):999-1014 View Article PubMed/NCBI
  125. Ježek J, Cooper KF, Strich R. Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants (Basel) 2018;7(1):13 View Article PubMed/NCBI
  126. Deng R, Zhang HL, Huang JH, Cai RZ, Wang Y, Chen YH, et al. MAPK1/3 kinase-dependent ULK1 degradation attenuates mitophagy and promotes breast cancer bone metastasis. Autophagy 2021;17(10):3011-3029 View Article PubMed/NCBI
  127. Chen Q, Lei JH, Bao J, Wang H, Hao W, Li L, et al. BRCA1 Deficiency Impairs Mitophagy and Promotes Inflammasome Activation and Mammary Tumor Metastasis. Adv Sci (Weinh) 2020;7(6):1903616 View Article PubMed/NCBI
  128. Chourasia AH, Tracy K, Frankenberger C, Boland ML, Sharifi MN, Drake LE, et al. Mitophagy defects arising from BNip3 loss promote mammary tumor progression to metastasis. EMBO Rep 2015;16(9):1145-1163 View Article PubMed/NCBI
  129. Fang J, Zou X, Gong L, Xi J, Liu Y, Yang X, et al. Acid ground nano-realgar processed product inhibits breast cancer by inducing mitophagy via the p53/BNIP3/NIX pathway. J Cell Mol Med 2023;27(22):3478-3490 View Article PubMed/NCBI
  130. Cao J, Ma X, Yan X, Zhang G, Hong S, Ma R, et al. Kaempferol induces mitochondrial dysfunction and mitophagy by activating the LKB1/AMPK/MFF pathway in breast precancerous lesions. Phytother Res 2023;37(8):3602-3616 View Article PubMed/NCBI
  131. Li X, Wang M, Li S, Chen Y, Wang M, Wu Z, et al. HIF-1-induced mitochondrial ribosome protein L52: a mechanism for breast cancer cellular adaptation and metastatic initiation in response to hypoxia. Theranostics 2021;11(15):7337-7359 View Article PubMed/NCBI
  132. Li Q, Chu Y, Li S, Yu L, Deng H, Liao C, et al. The oncoprotein MUC1 facilitates breast cancer progression by promoting Pink1-dependent mitophagy via ATAD3A destabilization. Cell Death Dis 2022;13(10):899 View Article PubMed/NCBI
  133. Chen C, Xiang A, Lin X, Guo J, Liu J, Hu S, et al. Mitophagy: insights into its signaling molecules, biological functions, and therapeutic potential in breast cancer. Cell Death Discov 2024;10(1):457 View Article PubMed/NCBI
  134. Ni HM, Williams JA, Ding WX. Mitochondrial dynamics and mitochondrial quality control. Redox Biol 2015;4:6-13 View Article PubMed/NCBI
  135. Gundamaraju R, Lu W, Manikam R. Revisiting Mitochondria Scored Cancer Progression and Metastasis. Cancers (Basel) 2021;13(3):432 View Article PubMed/NCBI
  136. Zhao M, Li J, Chen F, Han Y, Chen D, Hu H. Engineering nanoparticles boost TNBC therapy by CD24 blockade and mitochondrial dynamics regulation. J Control Release 2023;355:211-227 View Article PubMed/NCBI
  137. Baek ML, Lee J, Pendleton KE, Berner MJ, Goff EB, Tan L, et al. Mitochondrial structure and function adaptation in residual triple negative breast cancer cells surviving chemotherapy treatment. Oncogene 2023;42(14):1117-1131 View Article PubMed/NCBI
  138. Saito K, Zhang Q, Yang H, Yamatani K, Ai T, Ruvolo V, et al. Exogenous mitochondrial transfer and endogenous mitochondrial fission facilitate AML resistance to OxPhos inhibition. Blood Adv 2021;5(20):4233-4255 View Article PubMed/NCBI
  139. Khoramipour K, Soltany A, Khosravi P, Rezaei MH, Madadizadeh E, García-Chico C, et al. High intensity interval training as a therapy: Mitophagy restoration in breast cancer. Arch Biochem Biophys 2024;762:110213 View Article PubMed/NCBI
  140. Hu J, Zhang Y, Jiang X, Zhang H, Gao Z, Li Y, et al. ROS-mediated activation and mitochondrial translocation of CaMKII contributes to Drp1-dependent mitochondrial fission and apoptosis in triple-negative breast cancer cells by isorhamnetin and chloroquine. J Exp Clin Cancer Res 2019;38(1):225 View Article PubMed/NCBI
  141. Lucantoni F, Dussmann H, Prehn JHM. Metabolic Targeting of Breast Cancer Cells With the 2-Deoxy-D-Glucose and the Mitochondrial Bioenergetics Inhibitor MDIVI-1. Front Cell Dev Biol 2018;6:113 View Article PubMed/NCBI
  142. Chang JC, Chang HS, Yeh CY, Chang HJ, Cheng WL, Lin TT, et al. Regulation of mitochondrial fusion and mitophagy by intra-tumoral delivery of membrane-fused mitochondria or Midiv-1 enhances sensitivity to doxorubicin in triple-negative breast cancer. Biomed Pharmacother 2022;153:113484 View Article PubMed/NCBI
  143. Chang JC, Chang HS, Wu YC, Cheng WL, Lin TT, Chang HJ, et al. Antitumor Actions of Intratumoral Delivery of Membrane-Fused Mitochondria in a Mouse Model of Triple-Negative Breast Cancers. Onco Targets Ther 2020;13:5241-5255 View Article PubMed/NCBI
  144. Li L, Yu AQ. The functional role of peroxiredoxin 3 in reactive oxygen species, apoptosis, and chemoresistance of cancer cells. J Cancer Res Clin Oncol 2015;141(12):2071-2077 View Article PubMed/NCBI
  145. Chen J, Cao X, Qin X, Liu H, Chen S, Zhong S, et al. Proteomic analysis of the molecular mechanism of curcumin/β-cyclodextrin polymer inclusion complex inhibiting HepG2 cells growth. J Food Biochem 2020;44(2):e13119 View Article PubMed/NCBI
  146. Lv C, Huang Y, Wang Q, Wang C, Hu H, Zhang H, et al. Ainsliadimer A induces ROS-mediated apoptosis in colorectal cancer cells via directly targeting peroxiredoxin 1 and 2. Cell Chem Biol 2023;30(3):295-307.e5 View Article PubMed/NCBI
  147. Pasquier J, Guerrouahen BS, Al Thawadi H, Ghiabi P, Maleki M, Abu-Kaoud N, et al. Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance. J Transl Med 2013;11:94 View Article PubMed/NCBI
  148. Parida PK, Marquez-Palencia M, Ghosh S, Khandelwal N, Kim K, Nair V, et al. Limiting mitochondrial plasticity by targeting DRP1 induces metabolic reprogramming and reduces breast cancer brain metastases. Nat Cancer 2023;4(6):893-907 View Article PubMed/NCBI
  149. Sánchez-Alvarez R, De Francesco EM, Fiorillo M, Sotgia F, Lisanti MP. Mitochondrial Fission Factor (MFF) Inhibits Mitochondrial Metabolism and Reduces Breast Cancer Stem Cell (CSC) Activity. Front Oncol 2020;10:1776 View Article PubMed/NCBI
  150. Park JD, Jang HJ, Choi SH, Jo GH, Choi JH, Hwang S, et al. The ELK3-DRP1 axis determines the chemosensitivity of triple-negative breast cancer cells to CDDP by regulating mitochondrial dynamics. Cell Death Discov 2023;9(1):237 View Article PubMed/NCBI
  151. Wu D, Dasgupta A, Chen KH, Neuber-Hess M, Patel J, Hurst TE, et al. Identification of novel dynamin-related protein 1 (Drp1) GTPase inhibitors: Therapeutic potential of Drpitor1 and Drpitor1a in cancer and cardiac ischemia-reperfusion injury. FASEB J 2020;34(1):1447-1464 View Article PubMed/NCBI
  152. Kober KM, Olshen A, Conley YP, Schumacher M, Topp K, Smoot B, et al. Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain 2018;14:1744806918816462 View Article PubMed/NCBI
  153. Zhang JN, Zhang Z, Huang ZL, Guo Q, Wu ZQ, Ke C, et al. Isotoosendanin inhibits triple-negative breast cancer metastasis by reducing mitochondrial fission and lamellipodia formation regulated by the Smad2/3-GOT2-MYH9 signaling axis. Acta Pharmacol Sin 2024;45(12):2672-2683 View Article PubMed/NCBI
  154. Li Y, Shi L, Zhao F, Luo Y, Zhang M, Wu X, et al. PIM1 attenuates cisplatin-induced AKI by inhibiting Drp1 activation. Cell Signal 2024;113:110969 View Article PubMed/NCBI
  155. Alharbi BF, Al-Fahad D, Richard Dash P. Roles of Endocytic Processes and Early Endosomes on Focal Adhesion Dynamics in MDA-MB-231 Cells. Rep Biochem Mol Biol 2021;10(2):145-155 View Article PubMed/NCBI
  156. Dasgupta A, Chen KH, Wu D, Hoskin V, Mewburn J, Lima PDA, et al. An epigenetic increase in mitochondrial fission by MiD49 and MiD51 regulates the cell cycle in cancer: Diagnostic and therapeutic implications. FASEB J 2020;34(4):5106-5127 View Article PubMed/NCBI
  157. Gao T, Zhang X, Zhao J, Zhou F, Wang Y, Zhao Z, et al. SIK2 promotes reprogramming of glucose metabolism through PI3K/AKT/HIF-1α pathway and Drp1-mediated mitochondrial fission in ovarian cancer. Cancer Lett 2020;469:89-101 View Article PubMed/NCBI
  158. Si L, Fu J, Liu W, Hayashi T, Mizuno K, Hattori S, et al. Silibinin-induced mitochondria fission leads to mitophagy, which attenuates silibinin-induced apoptosis in MCF-7 and MDA-MB-231 cells. Arch Biochem Biophys 2020;685:108284 View Article PubMed/NCBI
  159. Li GB, Fu RQ, Shen HM, Zhou J, Hu XY, Liu YX, et al. Polyphyllin I induces mitophagic and apoptotic cell death in human breast cancer cells by increasing mitochondrial PINK1 levels. Oncotarget 2017;8(6):10359-10374 View Article PubMed/NCBI
  160. Zhen Y, Yuan Z, Zhang J, Chen Y, Fu Y, Liu Y, et al. Flubendazole induces mitochondrial dysfunction and DRP1-mediated mitophagy by targeting EVA1A in breast cancer. Cell Death Dis 2022;13(4):375 View Article PubMed/NCBI
  161. Tran VT, Pham DV, Choi DY, Park PH. Mitophagy Induction and Aryl Hydrocarbon Receptor-Mediated Redox Signaling Contribute to the Suppression of Breast Cancer Cell Growth by Taloxifene via Regulation of Inflammasomes Activation. Antioxid Redox Signal 2022;37(13-15):1030-1050 View Article PubMed/NCBI
  162. Shen LW, Jiang XX, Li ZQ, Li J, Wang M, Jia GF, et al. Cepharanthine sensitizes human triple negative breast cancer cells to chemotherapeutic agent epirubicin via inducing cofilin oxidation-mediated mitochondrial fission and apoptosis. Acta Pharmacol Sin 2022;43(1):177-193 View Article PubMed/NCBI
  163. Choi YJ, Choi YK, Ko SG, Cheon C, Kim TY. Investigation of Molecular Mechanisms Involved in Sensitivity to the Anti-Cancer Activity of Costunolide in Breast Cancer Cells. Int J Mol Sci 2023;24(4):4009 View Article PubMed/NCBI
  164. Zheng H, Hu C, Quan Y, Ye X, Shi X, Guo Z, et al. A copper complex that combats triple negative breast cancer by restraining angiogenesis. Dalton Trans 2023;52(22):7626-7634 View Article PubMed/NCBI
  165. Zhang GD, Wang MM, Su Y, Fang H, Xue XL, Liu HK, et al. Mitochondria-targeted ruthenium complexes can be generated in vitro and in living cells to target triple-negative breast cancer cells by autophagy inhibition. J Inorg Biochem 2024;256:112574 View Article PubMed/NCBI
  166. Hua F, Xiao YY, Qu XH, Li SS, Zhang K, Zhou C, et al. Baicalein sensitizes triple negative breast cancer MDA-MB-231 cells to doxorubicin via autophagy-mediated down-regulation of CDK1. Mol Cell Biochem 2023;478(7):1519-1531 View Article PubMed/NCBI
  167. Yapa NMB, Lisnyak V, Reljic B, Ryan MT. Mitochondrial dynamics in health and disease. FEBS Lett 2021;595(8):1184-1204 View Article PubMed/NCBI
  168. Som Chaudhury S, Nandi M, Kumar K, Ruidas B, Sur TK, Prasad P, et al. Rodent Model Preclinical Assessment of PEGylated Block Copolymer Targeting Cognition and Oxidative Stress Insults of Alzheimer’s Disease. Mol Neurobiol 2023;60(4):2036-2050 View Article PubMed/NCBI
  169. Grel H, Woznica D, Ratajczak K, Kalwarczyk E, Anchimowicz J, Switlik W, et al. Mitochondrial Dynamics in Neurodegenerative Diseases: Unraveling the Role of Fusion and Fission Processes. Int J Mol Sci 2023;24(17):13033 View Article PubMed/NCBI
  170. Sharma A, Ahmad S, Ahmad T, Ali S, Syed MA. Mitochondrial dynamics and mitophagy in lung disorders. Life Sci 2021;284:119876 View Article PubMed/NCBI
  171. Darvin P, Sasidharan Nair V. Editorial: Understanding mitochondrial dynamics and metabolic plasticity in cancer stem cells: Recent advances in cancer treatment and potential therapeutic approaches. Front Oncol 2023;13:1155774 View Article PubMed/NCBI
  172. Zhao Z, Mei Y, Wang Z, He W. The Effect of Oxidative Phosphorylation on Cancer Drug Resistance. Cancers (Basel) 2022;15(1):62 View Article PubMed/NCBI
  173. Kuang J, Liu H, Feng L, Xue Y, Tang H, Xu P. How mitochondrial dynamics imbalance affects the progression of breast cancer:a mini review. Med Oncol 2024;41(10):238 View Article PubMed/NCBI
  174. Rodrigues T, Ferraz LS. Therapeutic potential of targeting mitochondrial dynamics in cancer. Biochem Pharmacol 2020;182:114282 View Article PubMed/NCBI