Beyond Conventional Approaches: The Revolutionary Role of Nanoparticles in Breast Cancer
Mohan Liu
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Investigation (equal), Writing - original draft (lead), Writing - review & editing (equal)
Search for more papers by this authorYusi Wang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Formal analysis (lead), Writing - original draft (supporting)
Search for more papers by this authorYan Li
SiChuan Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Vaccines and Biological Products, Chengdu, Sichuan, China
Contribution: Writing - review & editing (equal)
Search for more papers by this authorYibing Zhang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Investigation (equal), Validation (equal)
Search for more papers by this authorBailing Zhou
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Data curation (equal), Investigation (equal)
Search for more papers by this authorCorresponding Author
Lei Yang
Sichuan Institute for Drug Control, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Validation (equal), Writing - review & editing (equal)
Search for more papers by this authorCorresponding Author
Xi Yan
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Investigation (equal), Supervision (equal), Writing - review & editing (equal)
Search for more papers by this authorCorresponding Author
Li Yang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Conceptualization (lead), Supervision (lead)
Search for more papers by this authorMohan Liu
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Investigation (equal), Writing - original draft (lead), Writing - review & editing (equal)
Search for more papers by this authorYusi Wang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Formal analysis (lead), Writing - original draft (supporting)
Search for more papers by this authorYan Li
SiChuan Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Vaccines and Biological Products, Chengdu, Sichuan, China
Contribution: Writing - review & editing (equal)
Search for more papers by this authorYibing Zhang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Investigation (equal), Validation (equal)
Search for more papers by this authorBailing Zhou
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Contribution: Data curation (equal), Investigation (equal)
Search for more papers by this authorCorresponding Author
Lei Yang
Sichuan Institute for Drug Control, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Validation (equal), Writing - review & editing (equal)
Search for more papers by this authorCorresponding Author
Xi Yan
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Investigation (equal), Supervision (equal), Writing - review & editing (equal)
Search for more papers by this authorCorresponding Author
Li Yang
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Correspondence: Lei Yang ([email protected])
Xi Yan ([email protected])
Li Yang ([email protected])
Contribution: Conceptualization (lead), Supervision (lead)
Search for more papers by this authorThese authors contributed equally: Mohan Liu, Yusi Wang and Yan Li.
ABSTRACT
Breast cancer (BCa) remains a significant health challenge worldwide, with a high propensity for early metastasis and poor prognosis. While surgery, chemotherapy, and radiotherapy are fundamental for managing BCa, severe side effects, such as low patient adherence and suboptimal survival outcomes, cause concern. Therefore, there is a critical need to innovate new approaches that facilitate early detection, accurate diagnosis, and more effective treatment strategies for BCa. Nanotechnological approaches have been introduced for the diagnosis and treatment of various cancers, especially BCa. The current review aims to emphasize and highlight possible applications of nanomedicine in early detection, accurate diagnosis and efficient treatment strategies for BCa. Nanocarriers can deliver chemotherapeutic agents, enhancing cytotoxicity against BCa cells and preventing the development of drug resistance. Nanoparticles also boost the efficacy of gene therapy which promotes their potential for regulating gene expression. The co-delivery of drugs and genes by nanoparticles can have a synergistic effect on BCa and remodel the tumor microenvironment. In this review, we discussed the latest advances in the application of nanomedicines for diagnosing and treating BCa. Current research highlights the potential benefits of nanomedicine over traditional approaches and further efforts to translate these research findings into clinical practice for BCa.
Conflicts of Interest
The authors declare no conflicts of interest.
Open Research
Data Availability Statement
All data generated during the study appear in the submitted article.
References
- 1F. Derakhshan and J. S. Reis-Filho, “Pathogenesis of Triple-Negative Breast Cancer,” Annual Review of Pathology: Mechanisms of Disease 17 (2022): 181–204.
- 2R. A. Leon-Ferre and M. P. Goetz, “Advances In Systemic Therapies for Triple Negative Breast Cancer,” BMJ 381 (2023): e071674.
- 3Y. Li, H. Zhang, Y. Merkher, et al., “Recent Advances in Therapeutic Strategies for Triple-Negative Breast Cancer,” Journal of Hematology & Oncology 15, no. 1 (2022): 121.
- 4M. Nakhjavani, R. M. Samarasinghe, and S. Shigdar, “Triple-Negative Breast Cancer Brain Metastasis: An Update on Druggable Targets, Current Clinical Trials, and Future Treatment Options,” Drug Discovery Today 27, no. 5 (2022): 1298–1314.
- 5M. Toi, T. Kinoshita, J. R. Benson, et al., “Non-Surgical Ablation for Breast Cancer: An Emerging Therapeutic Option,” Lancet Oncology 25, no. 3 (2024): e114–e125.
- 6A. C. Garrido-Castro, N. U. Lin, and K. Polyak, “Insights Into Molecular Classifications of Triple-Negative Breast Cancer: Improving Patient Selection for Treatment,” Cancer Discovery 9, no. 2 (2019): 176–198.
- 7M. Robson, K. J. Ruddy, S. A. Im, et al., “Patient-Reported Outcomes in Patients With a Germline BRCA Mutation and HER2-negative Metastatic Breast Cancer Receiving Olaparib versus Chemotherapy in the OlympiAD Trial,” European Journal of Cancer 120 (2019): 20–30.
- 8J. Ettl, R. G. W. Quek, K. H. Lee, et al., “Quality of Life With Talazoparib Versus Physician's Choice of Chemotherapy in Patients With Advanced Breast Cancer and Germline BRCA1/2 Mutation: Patient-Reported Outcomes From the EMBRACA Phase Iii Trial,” Annals of Oncology 29, no. 9 (2018): 1939–1947.
- 9P. Schmid, S. Adams, H. S. Rugo, et al., “Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer,” New England Journal of Medicine 379, no. 22 (2018): 2108–2121.
- 10X. Bai, J. Ni, J. Beretov, P. Graham, and Y. Li, “Triple-Negative Breast Cancer Therapeutic Resistance: Where Is the Achilles' Heel?,” Cancer Letters 497 (2021): 100–111.
- 11E. S. McDonald, A. S. Clark, J. Tchou, P. Zhang, and G. M. Freedman, “Clinical Diagnosis and Management of Breast Cancer,” Journal of Nuclear Medicine 57, no. Suppl 1 (2016): 9S–16S.
- 12S. H. Jafari, Z. Saadatpour, A. Salmaninejad, et al., “Breast Cancer Diagnosis: Imaging Techniques and Biochemical Markers,” Journal of Cellular Physiology 233, no. 7 (2018): 5200–5213.
- 13A. S. Tagliafico, M. Piana, D. Schenone, R. Lai, A. M. Massone, and N. Houssami, “Overview of Radiomics In Breast Cancer Diagnosis and Prognostication,” Breast 49 (2020): 74–80.
- 14C. Mannelli, “Tissue vs Liquid Biopsies for Cancer Detection: Ethical Issues,” Journal of Bioethical inquiry 16, no. 4 (2019): 551–557.
- 15R. Baskar, K. A. Lee, R. Yeo, and K. W. Yeoh, “Cancer and Radiation Therapy: Current Advances and Future Directions,” International Journal of Medical Sciences 9, no. 3 (2012): 193–199.
- 16F. A. Vicini, R. S. Cecchini, J. R. White, et al., “Long-Term Primary Results of Accelerated Partial Breast Irradiation After Breast-Conserving Surgery for Early-Stage Breast Cancer: A Randomised, Phase 3, Equivalence Trial,” Lancet 394, no. 10215 (2019): 2155–2164.
- 17F. Poggio, E. Blondeaux, M. Tagliamento, et al., “Efficacy of Adjuvant Chemotherapy Schedules for Breast Cancer According to Body Mass Index: Results From the Phase III GIM2 Trial,” ESMO Open 9, no. 8 (2024): 103650.
- 18T. Ria, R. Roy, U. S. Mandal, and U. H. Sk, “Prospects of Nano-Theranostic Approaches Against Breast and Cervical Cancer,” Biochimica et Biophysica Acta (BBA)—Reviews on Cancer 1879, no. 6 (2024): 189227.
- 19J. Liu, T. Zheng, and Y. Tian, “Functionalized h-BN Nanosheets as a Theranostic Platform for SERS Real-Time Monitoring of MicroRNA and Photodynamic Therapy,” Angewandte Chemie International Edition 58, no. 23 (2019): 7757–7761.
- 20L. Ren, S. Chen, H. Li, et al., “MRI-Guided Liposomes for Targeted Tandem Chemotherapy and Therapeutic Response Prediction,” Acta Biomaterialia 35 (2016): 260–268.
- 21G. Mikhaylov, U. Mikac, A. A. Magaeva, et al., “Ferri-Liposomes as an MRI-Visible Drug-Delivery System for Targeting Tumours and Their Microenvironment,” Nature Nanotechnology 6, no. 9 (2011): 594–602.
- 22L. H. Law, J. Huang, P. Xiao, et al., “Multiple CEST Contrast Imaging of Nose-to-Brain Drug Delivery Using Iohexol Liposomes at 3T MRI,” Journal of Controlled Release 354 (2023): 208–220.
- 23N. El-Sayed, V. Trouillet, A. Clasen, G. Jung, K. Hollemeyer, and M. Schneider, “NIR-Emitting Gold Nanoclusters-Modified Gelatin Nanoparticles as a Bioimaging Agent in Tissue,” Advanced Healthcare Materials 8, no. 24 (2019): e1900993.
- 24S. Bonacchi, A. Cantelli, G. Battistelli, et al., “Photoswitchable NIR-Emitting Gold Nanoparticles,” Angewandte Chemie International Edition 55, no. 37 (2016): 11064–11068.
- 25A. Zhang, S. Pan, Y. Zhang, et al., “Carbon-Gold Hybrid Nanoprobes for Real-Time Imaging, Photothermal/Photodynamic and Nanozyme Oxidative Therapy,” Theranostics 9, no. 12 (2019): 3443–3458.
- 26M. Aioub and M. A. El-Sayed, “A Real-Time Surface Enhanced Raman Spectroscopy Study of Plasmonic Photothermal Cell Death Using Targeted Gold Nanoparticles,” Journal of the American Chemical Society 138, no. 4 (2016): 1258–1264.
- 27Y. L. Xie, V. R. de Jager, R. Y. Chen, et al., “Fourteen-Day PET/CT Imaging to Monitor Drug Combination Activity in Treated Individuals With Tuberculosis,” Science Translational Medicine 13, no. 579 (2021): eabd7618.
- 28H. Lee, A. F. Shields, B. A. Siegel, et al., “(64)Cu-MM-302 Positron Emission Tomography Quantifies Variability of Enhanced Permeability and Retention of Nanoparticles in Relation to Treatment Response in Patients With Metastatic Breast Cancer,” Clinical Cancer Research 23, no. 15 (2017): 4190–4202.
- 29P. Joyce, C. J. Allen, M. J. Alonso, et al., “A Translational Framework to DELIVER Nanomedicines to the Clinic,” Nature Nanotechnology 19, no. 11 (2024): 1597–1611.
- 30M. Ashrafizadeh, A. Zarrabi, A. Bigham, et al., “(Nano)Platforms in Breast Cancer Therapy: Drug/Gene Delivery, Advanced Nanocarriers and Immunotherapy,” Medicinal Research Reviews 43, no. 6 (2023): 2115–2176.
- 31S. Torres Quintas, A. Canha-Borges, M. J. Oliveira, B. Sarmento, and F. Castro, “Special Issue: Nanotherapeutics in Women's Health Emerging Nanotechnologies for Triple-Negative Breast Cancer Treatment,” Small 20, no. 41 (2024): e2300666.
- 32T. Liu, X. Si, L. Liu, et al., “Injectable Nano-In-Gel Vaccine for Spatial and Temporal Control of Vaccine Kinetics and Breast Cancer Postsurgical Therapy,” ACS Nano 18, no. 4 (2024): 3087–3100.
- 33S. K. Misra, X. Wang, I. Srivastava, et al., “Combinatorial Therapy for Triple Negative Breast Cancer Using Hyperstar Polymer-Based Nanoparticles,” Chemical Communications 51, no. 93 (2015): 16710–16713.
- 34J. Chen, Y. Zhu, C. Wu, and J. Shi, “Nanoplatform-Based Cascade Engineering for Cancer Therapy,” Chemical Society Reviews 49, no. 24 (2020): 9057–9094.
- 35J. Halder, D. Pradhan, P. Biswasroy, et al, “Trends in Iron Oxide Nanoparticles: A Nano-Platform for Theranostic Application in Breast Cancer,” Journal of Drug Targeting 30, no. 10 (2022): 1055–1075.
- 36X. Chen, M. Hou, X. Zhang, H. Liu, W. Li, and W. Hong, “Active Targeted Janus Theranostic Nanoplatforms Enable Chemo-Photothermal Therapy to Inhibit the Growth of Breast Cancer,” Molecular Pharmaceutics 20, no. 11 (2023): 5800–5810.
- 37P. Aggarwal, J. B. Hall, C. B. McLeland, M. A. Dobrovolskaia, and S. E. McNeil, “Nanoparticle Interaction With Plasma Proteins as It Relates to Particle Biodistribution, Biocompatibility and Therapeutic Efficacy,” Advanced Drug Delivery Reviews 61, no. 6 (2009): 428–437.
- 38R. Zhao, C. Fu, Z. Wang, et al., “A pH-Responsive Nanoparticle Library With Precise pH Tunability by Co-Polymerization With Non-Ionizable Monomers,” Angewandte Chemie International Edition 61, no. 19 (2022): e202200152.
- 39G. T. Tietjen, L. G. Bracaglia, W. M. Saltzman, and J. S. Pober, “Focus on Fundamentals: Achieving Effective Nanoparticle Targeting,” Trends in Molecular Medicine 24, no. 7 (2018): 598–606.
- 40D. Barba, A. León-Sosa, P. Lugo, et al., “Breast Cancer, Screening and Diagnostic Tools: All You Need to Know,” Critical Reviews in Oncology/Hematology 157 (2021): 103174.
- 41Q. Xie, S. Li, X. Feng, et al., “All-In-One Approaches for Triple-Negative Breast Cancer Therapy: Metal-Phenolic Nanoplatform for MR Imaging-Guided Combinational Therapy,” Journal of Nanobiotechnology 20, no. 1 (2022): 226.
- 42Y. Du, C. Yang, F. Li, et al., “Core-Shell-Satellite Nanomaces as Remotely Controlled Self-Fueling Fenton Reagents for Imaging-Guided Triple-Negative Breast Cancer-Specific Therapy,” Small 16, no. 31 (2020): e2002537.
- 43J. C. Hsu, P. C. Naha, K. C. Lau, et al., “An All-In-One Nanoparticle (AION) Contrast Agent for Breast Cancer Screening With DEM-CT-MRI-NIRF Imaging,” Nanoscale 10, no. 36 (2018): 17236–17248.
- 44D. Yao, Y. Wang, R. Zou, et al., “Molecular Engineered Squaraine Nanoprobe for NIR-II/Photoacoustic Imaging and Photothermal Therapy of Metastatic Breast Cancer,” ACS Applied Materials & Interfaces 12, no. 4 (2020): 4276–4284.
- 45C. Zhu, Q. Ma, L. Gong, et al., “Manganese-Based Multifunctional Nanoplatform for Dual-Modal Imaging and Synergistic Therapy of Breast Cancer,” Acta Biomaterialia 141 (2022): 429–439.
- 46Y. Wang, L. Jiang, Y. Zhang, et al., “Fibronectin-Targeting and Cathepsin B-Activatable Theranostic Nanoprobe for MR/Fluorescence Imaging and Enhanced Photodynamic Therapy for Triple Negative Breast Cancer,” ACS Applied Materials & Interfaces 12, no. 30 (2020): 33564–33574.
- 47Z. Cheng, Y. Jin, J. Li, et al., “Fibronectin-Targeting and Metalloproteinase-Activatable Smart Imaging Probe for Fluorescence Imaging and Image-Guided Surgery of Breast Cancer,” Journal of Nanobiotechnology 21, no. 1 (2023): 112.
- 48K. Welsher, Z. Liu, D. Daranciang, and H. Dai, “Selective Probing and Imaging of Cells With Single Walled Carbon Nanotubes as Near-Infrared Fluorescent Molecules,” Nano Letters 8, no. 2 (2008): 586–590.
- 49Q. Jiang, L. Liu, Q. Li, et al., “NIR-Laser-Triggered Gadolinium-Doped Carbon Dots for Magnetic Resonance Imaging, Drug Delivery and Combined Photothermal Chemotherapy for Triple Negative Breast Cancer,” Journal of Nanobiotechnology 19, no. 1 (2021): 64.
- 50Y. Lee, J. Ni, J. Beretov, V. C. Wasinger, P. Graham, and Y. Li, “Recent Advances of Small Extracellular Vesicle Biomarkersin Breast Cancer Diagnosis and Prognosis,” Molecular cancer 22, no. 1 (2023): 33.
- 51B. E. Dogan and L. W. Turnbull, “Imaging of Triple-Negative Breast Cancer,” Annals of Oncology 23, no. Suppl 6 (2012): vi23–vi29.
- 52M. J. Duffy, D. Evoy, and E. W. McDermott, “CA 15-3: Uses and Limitation as a Biomarker for Breast Cancer,” Clinica Chimica Acta 411, no. 23–24 (2010): 1869–1874.
- 53R. Arshad, M. H. Kiani, A. Rahdar, et al., “Nano-Based Theranostic Platforms for Breast Cancer: A Review of Latest Advancements,” Bioengineering 9, no. 7 (2022): 320.
- 54N. Ferreira, A. Marques, H. Águas, et al., “Label-Free Nanosensing Platform for Breast Cancer Exosome Profiling,” ACS Sensors 4, no. 8 (2019): 2073–2083.
- 55H. Lee, E. Lee, D. K. Kim, N. K. Jang, Y. Y. Jeong, and S. Jon, “Antibiofouling Polymer-Coated Superparamagnetic Iron Oxide Nanoparticles as Potential Magnetic Resonance Contrast Agents for In Vivo Cancer Imaging,” Journal of the American Chemical Society 128, no. 22 (2006): 7383–7389.
- 56R. John, R. Rezaeipoor, S. G. Adie, et al., “In Vivo Magnetomotive Optical Molecular Imaging Using Targeted Magnetic Nanoprobes,” Proceedings of the National Academy of Sciences 107, no. 18 (2010): 8085–8090.
- 57P. V. Ostroverkhov, A. S. Semkina, V. A. Naumenko, et al., “Synthesis and Characterization of Bacteriochlorin Loaded Magnetic Nanoparticles (MNP) for Personalized MRI Guided Photosensitizers Delivery to Tumor,” Journal of Colloid and Interface Science 537 (2019): 132–141.
- 58S. Y. Ong, C. Zhang, X. Dong, and S. Q. Yao, “Recent Advances in Polymeric Nanoparticles for Enhanced Fluorescence and Photoacoustic Imaging,” Angewandte Chemie International Edition 60, no. 33 (2021): 17797–17809.
- 59L. Li, J. Li, R. Hu, et al., “Tumor Cell Targeting and Responsive Nanoplatform for Multimodal-Imaging Guided Chemodynamic/Photodynamic/Photothermal Therapy Toward Triple Negative Breast Cancer,” ACS Applied Materials & Interfaces 15, no. 23 (2023): 27706–27718.
- 60R. Wang, L. Cheng, L. He, et al., “Nitric Oxide Nano-Reactor DNMF/PLGA Enables Tumor Vascular Microenvironment and Chemo-Hyperthermia Synergetic Therapy,” Journal of Nanobiotechnology 22, no. 1 (2024): 110.
- 61S. Yue, P. Zhang, M. Qin, et al., “An Enzyme-Like Activity Nanoprobe Based on Fe(III)–Rutin Hydrate Biomineral for MR Imaging and Therapy of Triple Negative Breast Cancer,” Advanced Functional Materials 32, no. 31 (2022): 2202848.
- 62J. Ackermann, J. T. Metternich, S. Herbertz, and S. Kruss, “Biosensing With Fluorescent Carbon Nanotubes,” Angewandte Chemie International Edition 61, no. 18 (2022): e202112372.
- 63L. Sistemich, P. Galonska, J. Stegemann, J. Ackermann, and S. Kruss, “Near-Infrared Fluorescence Lifetime Imaging of Biomolecules With Carbon Nanotubes,” Angewandte Chemie International Edition 62, no. 24 (2023): e202300682.
- 64F. Mouffouk, S. Aouabdi, E. Al-Hetlani, H. Serrai, T. Alrefae, and L. Leo Chen, “New Generation of Electrochemical Immunoassay Based on Polymeric Nanoparticles for Early Detection of Breast Cancer,” International Journal of Nanomedicine 12 (2017): 3037–3047.
- 65H. Shi, Z. Huang, T. Xu, A. Sun, and J. Ge, “New Diagnostic and Therapeutic Strategies for Myocardial Infarction via Nanomaterials,” EBioMedicine 78 (2022): 103968.
- 66S. M. Patil, V. C. Karade, J. H. Kim, A. D. Chougale, and P. B. Patil, “Electrochemical Detection of a Breast Cancer Biomarker With an Amine-Functionalized Nanocomposite Pt-Fe(3)O(4)-MWCNTs-NH(2) as a Signal-Amplifying Label,” ACS Applied Materials & Interfaces 16, no. 20 (2024): 25601–25609.
- 67K. Kuntamung, J. Jakmunee, and K. Ounnunkad, “A Label-Free Multiplex Electrochemical Biosensor for the Detection of Three Breast Cancer Biomarker Proteins Employing Dye/Metal Ion-Loaded and Antibody-Conjugated Polyethyleneimine-Gold Nanoparticles,” Journal of Materials Chemistry B 9, no. 33 (2021): 6576–6585.
- 68Q. Huang, X. Zhu, X. Sun, et al., “Synergetic-Effect-Enhanced Electrochemiluminescence of Zein-Protected Au-Ag Bimetallic Nanoclusters for CA15-3 Detection,” Analytica Chimica Acta 1278 (2023): 341760.
- 69A. M. Shawky and M. El-Tohamy, “Signal Amplification Strategy of Label-Free Ultrasenstive Electrochemical Immunosensor Based Ternary Ag/TiO2/rGO Nanocomposites for Detecting Breast Cancer Biomarker Ca 15-3,” Materials Chemistry and Physics 272 (2021): 124983.
- 70M. L. Yola, “Sensitive Sandwich-Type Voltammetric Immunosensor for Breast Cancer Biomarker HER2 Detection Based on Gold Nanoparticles Decorated Cu-MOF and Cu(2)ZnSnS(4) NPs/Pt/g-C(3)N(4) Composite,” Microchimica Acta 188, no. 3 (2021): 78.
- 71A. A. Saeed, J. L. A. Sánchez, C. K. O'Sullivan, and M. N. Abbas, “DNA Biosensors Based on Gold Nanoparticles-Modified Graphene Oxide for the Detection of Breast Cancer Biomarkers for Early Diagnosis,” Bioelectrochemistry 118 (2017): 91–99.
- 72A. A. Lahcen, S. Rauf, A. Aljedaibi, et al., “Laser-Scribed Graphene Sensor Based on Gold Nanostructures and Molecularly Imprinted Polymers: Application for Her-2 Cancer Biomarker Detection,” Sensors and Actuators B: Chemical 347 (2021): 130556.
- 73Y. W. Hartati, L. K. Letelay, S. Gaffar, S. Wyantuti, and H. H. Bahti, “Cerium Oxide-Monoclonal Antibody Bioconjugate for Electrochemical Immunosensing of HER2 as a Breast Cancer Biomarker,” Sensing and Bio-Sensing Research 27 (2020): 100316.
- 74H. Tang, H. Wang, C. Yang, D. Zhao, Y. Qian, and Y. Li, “Nanopore-Based Strategy for Selective Detection of Single Carcinoembryonic Antigen (CEA) Molecules,” Analytical Chemistry 92, no. 4 (2020): 3042–3049.
- 75J. Li, L. Liu, Y. Ai, Y. Liu, H. Sun, and Q. Liang, “Self-Polymerized Dopamine-Decorated Au NPs and Coordinated With Fe-MOF as a Dual Binding Sites and Dual Signal-Amplifying Electrochemical Aptasensor for the Detection of CEA,” ACS Applied Materials & Interfaces 12, no. 5 (2020): 5500–5510.
- 76T. Yang, Y. Gao, Z. Liu, J. Xu, L. Lu, and Y. Yu, “Three-Dimensional Gold Nanoparticles/Prussian Blue-poly(3,4-ethylenedioxythiophene) Nanocomposite as Novel Redox Matrix for Label-Free Electrochemical Immunoassay of Carcinoembryonic Antigen,” Sensors and Actuators B: Chemical 239 (2017): 76–84.
- 77S. Cotchim, P. Thavarungkul, P. Kanatharana, and W. Limbut, “Multiplexed Label-Free Electrochemical Immunosensor for Breast Cancer Precision Medicine,” Analytica Chimica Acta 1130 (2020): 60–71.
- 78W. Wen, J. Y. Huang, T. Bao, et al., “Increased Electrocatalyzed Performance Through Hairpin Oligonucleotide Aptamer-Functionalized Gold Nanorods Labels and Graphene-Streptavidin Nanomatrix: Highly Selective and Sensitive Electrochemical Biosensor of Carcinoembryonic Antigen,” Biosensors and Bioelectronics 83 (2016): 142–148.
- 79P. A. Rasheed and N. Sandhyarani, “Femtomolar Level Detection of BRCA1 Gene Using a Gold Nanoparticle Labeled Sandwich Type DNA Sensor,” Colloids and Surfaces B: Biointerfaces 117 (2014): 7–13.
- 80P. Abdul Rasheed and N. Sandhyarani, “Attomolar Detection of BRCA1 Gene Based on Gold Nanoparticle Assisted Signal Amplification,” Biosensors and Bioelectronics 65 (2015): 333–340.
- 81P. A. Rasheed, T. Radhakrishnan, P. K. Shihabudeen, and N. Sandhyarani, “Reduced Graphene Oxide-Yttria Nanocomposite Modified Electrode for Enhancing the Sensitivity of Electrochemical Genosensor,” Biosensors and Bioelectronics 83 (2016): 361–367.
- 82M. Pourmadadi, A. Ghaemi, A. Khanizadeh, et al., “Breast Cancer Detection Based on Cancer Antigen 15-3; Emphasis on Optical and Electrochemical Methods: A Review,” Biosensors and Bioelectronics 260 (2024): 116425.
- 83P. Tarantino, G. Viale, M. F. Press, et al., “ESMO Expert Consensus Statements (ECS) on the Definition, Diagnosis, and Management of HER2-low Breast Cancer,” Annals of Oncology 34, no. 8 (2023): 645–659.
- 84Z. Turk, A. Armani, D. Jafari-Gharabaghlou, S. Madakbas, E. Bonabi, and N. Zarghami, “A New Insight Into the Early Detection of HER2 Protein in Breast Cancer Patients With a Focus on Electrochemical Biosensors Approaches: A Review,” International Journal of Biological Macromolecules 272, no. Pt 1 (2024): 132710.
- 85M. J. Duffy, “Serum Tumor Markers in Breast Cancer: Are They of Clinical Value?,” Clinical Chemistry 52, no. 3 (2006): 345–351.
- 86S. Tang, F. Zhou, Y. Sun, et al., “CEA in Breast Ductal Secretions as a Promising Biomarker for the Diagnosis of Breast Cancer: A Systematic Review and Meta-Analysis,” Breast Cancer 23, no. 6 (2016): 813–819.
- 87Y. Fu and H. Li, “Assessing Clinical Significance of Serum CA15-3 and Carcinoembryonic Antigen (CEA) Levels in Breast Cancer Patients: A Meta-Analysis,” Medical Science Monitor 22 (2016): 3154–3162.
- 88J. Kotsopoulos, J. Gronwald, T. Huzarski, et al., “Bilateral Oophorectomy and All-Cause Mortality in Women With BRCA1 and BRCA2 Sequence Variations,” JAMA Oncology 10, no. 4 (2024): 484–492.
- 89F. Ye, S. Dewanjee, Y. Li, et al., “Advancements in Clinical Aspects of Targeted Therapy and Immunotherapy in Breast Cancer,” Molecular Cancer 22, no. 1 (2023): 105.
- 90S. Mayor, “Tamoxifen Reduces Breast Cancer by a Third in High Risk Women,” BMJ 325, no. 7365 (2002): 613a.
- 91C. C. O'Sullivan, R. Clarke, M. P. Goetz, and J. Robertson, “Cyclin-Dependent Kinase 4/6 Inhibitors for Treatment of Hormone Receptor-Positive, ERBB2-Negative Breast Cancer: A Review,” JAMA Oncology 9, no. 9 (2023): 1273–1282.
- 92M. Gnant, A. C. Dueck, S. Frantal, et al., “Adjuvant Palbociclib for Early Breast Cancer: The PALLAS Trial Results (ABCSG-42/AFT-05/BIG-14-03),” Journal of Clinical Oncology 40, no. 3 (2022): 282–293.
- 93G. N. Hortobagyi, S. M. Stemmer, H. A. Burris, et al., “Overall Survival With Ribociclib Plus Letrozole in Advanced Breast Cancer,” New England Journal of Medicine 386, no. 10 (2022): 942–950.
- 94M. P. Goetz, M. Toi, J. Huober, et al., “Abemaciclib Plus a Nonsteroidal Aromatase Inhibitor as Initial Therapy for HR+, HER2− Advanced Breast Cancer: Final Overall Survival Results of MONARCH 3,” Annals of Oncology 35, no. 8 (2024): 718–727.
- 95O. The Editors of The Lancet Oncology, “Expression of Concern-Alpelisib Plus Fulvestrant In PIK3CA-mutated, Hormone Receptor-Positive Advanced Breast Cancer After a CDK4/6 Inhibitor (BYlieve): One Cohort of a Phase 2, Multicentre, Open-Label, Non-Comparative Study,” Lancet Oncology 25, no. 2 (2024): e42.
- 96J. Gómez Tejeda Zañudo, R. Barroso-Sousa, E. Jain, et al., “Exemestane Plus Everolimus and Palbociclib in Metastatic Breast Cancer: Clinical Response and Genomic/Transcriptomic Determinants of Resistance in a Phase I/II Trial,” Nature Communications 15, no. 1 (2024): 2446.
- 97A. C. Wolff, A. A. Lazar, I. Bondarenko, et al., “Randomized Phase IIi Placebo-Controlled Trial of Letrozole Plus Oral Temsirolimus as First-Line Endocrine Therapy in Postmenopausal Women With Locally Advanced or Metastatic Breast Cancer,” Journal of Clinical Oncology 31, no. 2 (2013): 195–202.
- 98T. A. Traina, K. Miller, D. A. Yardley, et al., “Enzalutamide for the Treatment of Androgen Receptor-Expressing Triple-Negative Breast Cancer,” Journal of Clinical Oncology 36, no. 9 (2018): 884–890.
- 99Q. Lu, W. Xia, K. Lee, et al., “Bicalutamide Plus Aromatase Inhibitor in Patients With Estrogen Receptor-Positive/Androgen Receptor-Positive Advanced Breast Cancer,” Oncologist 25, no. 1 (2020): 21–e15.
- 100J. Gong, A. C. Mita, Z. Wei, et al., “Phase II Study of Erdafitinib in Patients With Tumors With FGFR Amplifications: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol K1,” JCO Precision Oncology 8 (2024): e2300406.
- 101H. Fohlin, A. Nordenskjöld, J. Rosell, et al., “Breast Cancer Hormone Receptor Levels and Benefit From Adjuvant Tamoxifen in a Randomized Trial With Long-Term Follow-Up,” Acta Oncologica 63 (2024): 535–541.
- 102S. A. Hurvitz, R. Hegg, W. P. Chung, et al., “Trastuzumab Deruxtecan versus Trastuzumab Emtansine in Patients With HER2-Positive Metastatic Breast Cancer: Updated Results From DESTINY-Breast03, a Randomised, Open-Label, Phase 3 Trial,” Lancet 401, no. 10371 (2023): 105–117.
- 103F. Lynce, C. Mainor, R. N. Donahue, et al., “Adjuvant Nivolumab, Capecitabine or the Combination in Patients With Residual Triple-Negative Breast Cancer: The OXEL Randomized Phase II Study,” Nature Communications 15, no. 1 (2024): 2691.
- 104E. A. Mittendorf, H. Zhang, C. H. Barrios, et al., “Neoadjuvant Atezolizumab in Combination With Sequential Nab-Paclitaxel and Anthracycline-Based Chemotherapy Versus Placebo and Chemotherapy in Patients With Early-Stage Triple-Negative Breast Cancer (IMpassion031): A Randomised, Double-Blind, Phase 3 Trial,” Lancet 396, no. 10257 (2020): 1090–1100.
- 105L. Pusztai, C. Denkert, J. O'Shaughnessy, et al., “Event-Free Survival by Residual Cancer Burden With Pembrolizumab in Early-Stage TNBC: Exploratory Analysis From KEYNOTE-522,” Annals of Oncology 35, no. 5 (2024): 429–436.
- 106C. P. Prasad, S. K. Chaurasiya, W. Guilmain, and T. Andersson, “WNT5A Signaling Impairs Breast Cancer Cell Migration and Invasion via Mechanisms Independent of the Epithelial-Mesenchymal Transition,” Journal of Experimental & Clinical Cancer Research 35, no. 1 (2016): 144.
- 107Y. Zhao, N. Liu, P. Liu, et al., “Robust Boron Nanoplatform Provokes Potent Tumoricidal Activities via Inhibiting Heat Shock Protein,” Asian Journal of Pharmaceutical Sciences 17, no. 5 (2022): 728–740.
- 108J. Morry, W. Ngamcherdtrakul, S. Gu, et al., “Targeted Treatment of Metastatic Breast Cancer by PLK1 siRNA Delivered by an Antioxidant Nanoparticle Platform,” Molecular Cancer Therapeutics 16, no. 4 (2017): 763–772.
- 109B. Wang, X. Huang, H. Liang, et al., “PLK1 Inhibition Sensitizes Breast Cancer Cells to Radiation via Suppressing Autophagy,” International Journal of Radiation Oncology*Biology*Physics 110, no. 4 (2021): 1234–1247.
- 110B. C. Taylor, X. Sun, P. I. Gonzalez-Ericsson, et al., “NKG2A Is a Therapeutic Vulnerability in Immunotherapy Resistant MHC-I Heterogeneous Triple-Negative Breast Cancer,” Cancer Discovery 14, no. 2 (2024): 290–307.
- 111M. M. Agwa, M. M. Abu-Serie, D. A. Abdelmonsif, et al., “Vitamin D3/phospholipid Complex Decorated Caseinate Nanomicelles for Targeted Delivery of Synergistic Combination Therapy in Breast Cancer,” International Journal of Pharmaceutics 607 (2021): 120965.
- 112R. Rattan, R. Ali Fehmi, and A. Munkarah, “Metformin: An Emerging New Therapeutic Option for Targeting Cancer Stem Cells and Metastasis,” Journal of Oncology 2012 (2012): 928127.
- 113L. M. Spring, S. A. Wander, F. Andre, B. Moy, N. C. Turner, and A. Bardia, “Cyclin-Dependent Kinase 4 and 6 Inhibitors for Hormone Receptor-Positive Breast Cancer: Past, Present, and Future,” Lancet 395, no. 10226 (2020): 817–827.
- 114S. Goel, J. S. Bergholz, and J. J. Zhao, “Targeting CDK4 and CDK6 in Cancer,” Nature Reviews Cancer 22, no. 6 (2022): 356–372.
- 115S. J. Weintraub, C. A. Prater, and D. C. Dean, “Retinoblastoma Protein Switches the E2F Site From Positive to Negative Element,” Nature 358, no. 6383 (1992): 259–261.
- 116W. R. Sellers, J. W. Rodgers, and W. G. Kaelin, Jr., “A Potent Transrepression Domain in the Retinoblastoma Protein Induces a Cell Cycle Arrest When Bound to E2F Sites,” Proceedings of the National Academy of Sciences 92, no. 25 (1995): 11544–11548.
- 117C. J. Sherr and J. M. Roberts, “CDK Inhibitors: Positive and Negative Regulators of G1-phase Progression,” Genes & Development 13, no. 12 (1999): 1501–1512.
- 118C. L. Braal, E. M. Jongbloed, S. M. Wilting, R. H. J. Mathijssen, S. L. W. Koolen, and A. Jager, “Inhibiting CDK4/6 in Breast Cancer With Palbociclib, Ribociclib, and Abemaciclib: Similarities and Differences,” Drugs 81, no. 3 (2021): 317–331.
- 119S. E. Nunnery and I. A. Mayer, “Targeting the PI3K/AKT/mTOR Pathway in Hormone-Positive Breast Cancer,” Drugs 80, no. 16 (2020): 1685–1697.
- 120I. M. Browne, F. André, S. Chandarlapaty, L. A. Carey, and N. C. Turner, “Optimal Targeting of PI3K-AKT and mTOR in Advanced Oestrogen Receptor-Positive Breast Cancer,” Lancet Oncology 25, no. 4 (2024): e139–e151.
- 121K. Ganesan, C. Xu, J. Wu, et al., “Ononin Inhibits Triple-Negative Breast Cancer Lung Metastasis by Targeting the EGFR-Mediated PI3K/Akt/mTOR Pathway,” Science China Life Sciences 67, no. 9 (2024): 1849–1866.
- 122C. M. Venema, R. D. Bense, T. G. Steenbruggen, et al., “Consideration of Breast Cancer Subtype in Targeting the Androgen Receptor,” Pharmacology & Therapeutics 200 (2019): 135–147.
- 123E. Choupani, M. Mahmoudi Gomari, S. Zanganeh, et al., “Newly Developed Targeted Therapies Against the Androgen Receptor in Triple-Negative Breast Cancer: A Review,” Pharmacological Reviews 75, no. 2 (2023): 309–327.
- 124J. Feng, L. Li, N. Zhang, et al., “Androgen and AR Contribute to Breast Cancer Development and Metastasis: An Insight of Mechanisms,” Oncogene 36, no. 20 (2017): 2775–2790.
- 125B. D. Lehmann, J. A. Bauer, X. Chen, et al., “Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies,” Journal of Clinical Investigation 121, no. 7 (2011): 2750–2767.
- 126M. D. Burstein, A. Tsimelzon, G. M. Poage, et al., “Comprehensive Genomic Analysis Identifies Novel Subtypes and Targets of Triple-Negative Breast Cancer,” Clinical Cancer Research 21, no. 7 (2015): 1688–1698.
- 127J. Boers, C. M. Venema, E. F. J. de Vries, et al., “Serial [(18)F]-FDHT-PET to Predict Bicalutamide Efficacy in Patients With Androgen Receptor Positive Metastatic Breast Cancer,” European Journal of Cancer 144 (2021): 151–161.
- 128A. Servetto, L. Formisano, and C. L. Arteaga, “FGFR Signaling and Endocrine Resistance In Breast Cancer: Challenges for the Clinical Development of FGFR Inhibitors,” Biochimica et Biophysica Acta (BBA)—Reviews on Cancer 1876, no. 2 (2021): 188595.
- 129L. Formisano, Y. Lu, A. Servetto, et al., “Aberrant FGFR Signaling Mediates Resistance to CDK4/6 Inhibitors In ER+ Breast Cancer,” Nature Communications 10, no. 1 (2019): 1373.
- 130J. Z. Drago, L. Formisano, D. Juric, et al., “FGFR1 Amplification Mediates Endocrine Resistance But Retains TORC Sensitivity in Metastatic Hormone Receptor-Positive (HR(+)) Breast Cancer,” Clinical Cancer Research 25, no. 21 (2019): 6443–6451.
- 131T. Helsten, S. Elkin, E. Arthur, B. N. Tomson, J. Carter, and R. Kurzrock, “The FGFR Landscape in Cancer: Analysis of 4,853 Tumors by Next-Generation Sequencing,” Clinical Cancer Research 22, no. 1 (2016): 259–267.
- 132H. T. Mohamed, R. Gadalla, N. El-Husseiny, et al., “Inflammatory Breast Cancer: Activation of the Aryl Hydrocarbon Receptor and Its Target CYP1B1 Correlates Closely With Wnt5a/b-β-catenin Signalling, the Stem Cell Phenotype and Disease Progression,” Journal of Advanced Research 16 (2019): 75–86.
- 133D. Feng, J. Wang, W. Yang, et al., “Regulation of Wnt/PCP Signaling Through p97/VCP-KBTBD7-mediated Vangl Ubiquitination and Endoplasmic Reticulum-Associated Degradation,” Science Advances 7, no. 20 (2021): eabg2099.
- 134K. VanderVorst, C. A. Dreyer, J. Hatakeyama, et al., “Vangl-Dependent Wnt/Planar Cell Polarity Signaling Mediates Collective Breast Carcinoma Motility and Distant Metastasis,” Breast Cancer Research 25, no. 1 (2023): 52.
- 135Z. Chen, C. Tang, Y. Zhu, et al., “TrpC5 Regulates Differentiation Through the Ca2+/Wnt5a Signalling Pathway in Colorectal Cancer,” Clinical Science 131, no. 3 (2017): 227–237.
- 136Q. Liu, H. Zhu, K. Tiruthani, et al., “Nanoparticle-Mediated Trapping of Wnt Family Member 5A in Tumor Microenvironments Enhances Immunotherapy for B-Raf Proto-Oncogene Mutant Melanoma,” ACS Nano 12, no. 2 (2018): 1250–1261.
- 137L. Wang, Q. Zhang, and Q. You, “Targeting the HSP90-CDC37-kinase Chaperone Cycle: A Promising Therapeutic Strategy for Cancer,” Medicinal Research Reviews 42, no. 1 (2022): 156–182.
- 138J. M. Park, Y. J. Kim, S. Park, et al., “A Novel HSP90 Inhibitor Targeting the C-Terminal Domain Attenuates Trastuzumab Resistance In HER2-positive Breast Cancer,” Molecular cancer 19, no. 1 (2020): 161.
- 139Z. Yu, P. Deng, Y. Chen, et al., “Inhibition of the PLK1-Coupled Cell Cycle Machinery Overcomes Resistance to Oxaliplatin in Colorectal Cancer,” Advanced Science 8, no. 23 (2021): e2100759.
- 140C. Gelot, M. T. Kovacs, S. Miron, et al., “Polθ Is Phosphorylated by PLK1 to Repair Double-Strand Breaks in Mitosis,” Nature 621, no. 7978 (2023): 415–422.
- 141S. Cheng, X. Wan, L. Yang, et al., “RGCC-Mediated PLK1 Activity Drives Breast Cancer Lung Metastasis by Phosphorylating AMPKα2 to Activate Oxidative Phosphorylation and Fatty Acid Oxidation,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 342.
- 142S. Kandala, M. Ramos, L. Voith von Voithenberg, et al., “Chronic Chromosome Instability Induced by Plk1 Results in Immune Suppression in Breast Cancer,” Cell Reports 42, no. 12 (2023): 113266.
- 143Z. Zhang, L. Cheng, J. Li, et al., “Targeting Plk1 Sensitizes Pancreatic Cancer to Immune Checkpoint Therapy,” Cancer Research 82, no. 19 (2022): 3532–3548.
- 144K. Hu, J. H. Law, A. Fotovati, and S. E. Dunn, “Small Interfering RNA Library Screen Identified Polo-Like kinase-1 (PLK1) as a Potential Therapeutic Target for Breast Cancer That Uniquely Eliminates Tumor-Initiating Cells,” Breast Cancer Research 14, no. 1 (2012): R22.
- 145X. Liu, J. Song, H. Zhang, et al., “Immune Checkpoint HLA-E:CD94-NKG2A Mediates Evasion of Circulating Tumor Cells From NK Cell Surveillance,” Cancer Cell 41, no. 2 (2023): 272–287.e9.
- 146A. Frazao, M. Messaoudene, N. Nunez, et al., “CD16(+)NKG2A(high) Natural Killer Cells Infiltrate Breast Cancer-Draining Lymph Nodes,” Cancer Immunology Research 7, no. 2 (2019): 208–218.
- 147B. Salomé, J. P. Sfakianos, D. Ranti, et al., “NKG2A and HLA-E Define an Alternative Immune Checkpoint Axis in Bladder Cancer,” Cancer Cell 40, no. 9 (2022): 1027–1043.e9.
- 148D. Feldman, A. V. Krishnan, S. Swami, E. Giovannucci, and B. J. Feldman, “The Role of Vitamin D in Reducing Cancer Risk and Progression,” Nature Reviews Cancer 14, no. 5 (2014): 342–357.
- 149C. Carlberg and A. Muñoz, “An Update on Vitamin D Signaling and Cancer,” Seminars in Cancer Biology 79 (2022): 217–230.
- 150K. Visvanathan, A. M. Mondul, A. Zeleniuch-Jacquotte, et al., “Circulating Vitamin D and Breast Cancer Risk: An International Pooling Project of 17 Cohorts,” European Journal of Epidemiology 38, no. 1 (2023): 11–29.
- 151D. Guo, X. Ji, H. Xie, et al., “Targeted Reprogramming of Vitamin B(3) Metabolism as a Nanotherapeutic Strategy Towards Chemoresistant Cancers,” Advanced Materials 35, no. 36 (2023): e2301257.
- 152D. Nassar and C. Blanpain, “Cancer Stem Cells: Basic Concepts and Therapeutic Implications,” Annual Review of Pathology: Mechanisms of Disease 11 (2016): 47–76.
- 153L. Yang, P. Shi, G. Zhao, et al., “Targeting Cancer Stem Cell Pathways for Cancer Therapy,” Signal Transduction and Targeted Therapy 5, no. 1 (2020): 8.
- 154E. Batlle and H. Clevers, “Cancer Stem Cells Revisited,” Nature Medicine 23, no. 10 (2017): 1124–1134.
- 155A. Pavlopoulou, Y. Oktay, K. Vougas, M. Louka, C. E. Vorgias, and A. G. Georgakilas, “Determinants of Resistance to Chemotherapy and Ionizing Radiation in Breast Cancer Stem Cells,” Cancer Letters 380, no. 2 (2016): 485–493.
- 156J. Dittmer, “Breast Cancer Stem Cells: Features, Key Drivers and Treatment Options,” Seminars in Cancer Biology 53 (2018): 59–74.
- 157C. M. Perou, T. Sørlie, M. B. Eisen, et al., “Molecular Portraits of Human Breast Tumours,” Nature 406, no. 6797 (2000): 747–752.
- 158A. Marusyk, V. Almendro, and K. Polyak, “Intra-Tumour Heterogeneity: A Looking Glass for Cancer?,” Nature Reviews Cancer 12, no. 5 (2012): 323–334.
- 159S. K. Yeo and J. L. Guan, “Breast Cancer: Multiple Subtypes Within a Tumor?,” Trends in Cancer 3, no. 11 (2017): 753–760.
- 160P. A. Francis, M. M. Regan, G. F. Fleming, et al., “Adjuvant Ovarian Suppression in Premenopausal Breast Cancer,” New England Journal of Medicine 372, no. 5 (2015): 436–446.
- 161S. Sharma, M. Rajani, S. Aggarwal, S. Puri, and V. N. Baijal, “Spontaneous Pneumothorax and Pneumomediastinum In Metastatic Lung Disease,” The Indian Journal of Chest Diseases & Allied Sciences 30, no. 2 (1988): 125–132.
- 162S. S. Onkar, N. M. Carleton, P. C. Lucas, et al., “The Great Immune Escape: Understanding the Divergent Immune Response in Breast Cancer Subtypes,” Cancer Discovery 13, no. 1 (2023): 23–40.
- 163A. E. Teschendorff, A. Miremadi, S. E. Pinder, I. O. Ellis, and C. Caldas, “An Immune Response Gene Expression Module Identifies a Good Prognosis Subtype in Estrogen Receptor Negative Breast Cancer,” Genome Biology 8, no. 8 (2007): R157.
- 164K. G. K. Deepak, R. Vempati, G. P. Nagaraju, et al., “Tumor Microenvironment: Challenges and Opportunities in Targeting Metastasis of Triple Negative Breast Cancer,” Pharmacological Research 153 (2020): 104683.
- 165P. Bhat-Nakshatri, B. Kumar, E. Simpson, et al., “Breast Cancer Cell Detection and Characterization From Breast Milk-Derived Cells,” Cancer Research 80, no. 21 (2020): 4828–4839.
- 166P. Li, M. Lu, J. Shi, et al., “Lung Mesenchymal Cells Elicit Lipid Storage In Neutrophils That Fuel Breast Cancer Lung Metastasis,” Nature Immunology 21, no. 11 (2020): 1444–1455.
- 167B. S. Wiseman and Z. Werb, “Stromal Effects on Mammary Gland Development and Breast Cancer,” Science 296, no. 5570 (2002): 1046–1049.
- 168R. Kalluri and M. Zeisberg, “Fibroblasts in Cancer,” Nature Reviews Cancer 6, no. 5 (2006): 392–401.
- 169Z. Yang, X. Yang, S. Xu, et al., “Reprogramming of Stromal Fibroblasts by SNAI2 Contributes to Tumor Desmoplasia and Ovarian Cancer Progression,” Molecular Cancer 16, no. 1 (2017): 163.
- 170S. Ding, N. Qiao, Q. Zhu, et al., “Single-Cell Atlas Reveals a Distinct Immune Profile Fostered by T Cell-B Cell Crosstalk in Triple Negative Breast Cancer,” Cancer Communications 43, no. 6 (2023): 661–684.
- 171L. Mosca, A. Ilari, F. Fazi, Y. G. Assaraf, and G. Colotti, “Taxanes in Cancer Treatment: Activity, Chemoresistance and Its Overcoming,” Drug Resistance Updates 54 (2021): 100742.
- 172L. Garcia-Martinez, Y. Zhang, Y. Nakata, H. L. Chan, and L. Morey, “Epigenetic Mechanisms in Breast Cancer Therapy and Resistance,” Nature Communications 12, no. 1 (2021): 1786.
- 173A. Glaviano, S. A. Wander, R. D. Baird, et al., “Mechanisms of Sensitivity and Resistance to CDK4/CDK6 Inhibitors In Hormone Receptor-Positive Breast Cancer Treatment,” Drug Resistance Updates 76 (2024): 101103.
- 174H. Jia, C. I. Truica, B. Wang, et al., “Immunotherapy for Triple-Negative Breast Cancer: Existing Challenges and Exciting Prospects,” Drug Resistance Updates 32 (2017): 1–15.
- 175R. Wang, P. Kumar, M. Reda, et al., “Nanotechnology Applications in Breast Cancer Immunotherapy,” Small (Weinheim an der Bergstrasse, Germany) 20, no. 41 (2024): e2308639.
- 176X. Yang, M. Zhao, Z. Wu, et al., “Nano-Ultrasonic Contrast Agent for Chemoimmunotherapy of Breast Cancer by Immune Metabolism Reprogramming and Tumor Autophagy,” ACS Nano 16, no. 2 (2022): 3417–3431.
- 177A. M. Khalifa, M. A. Elsheikh, A. M. Khalifa, and Y. S. R. Elnaggar, “Current Strategies for Different Paclitaxel-Loaded Nano-Delivery Systems Towards Therapeutic Applications for Ovarian Carcinoma: A Review Article,” Journal of Controlled Release 311–312 (2019): 125–137.
- 178P. Tiwari, K. Yadav, R. P. Shukla, et al., “Surface Modification Strategies in Translocating Nano-Vesicles Across Different Barriers and the Role of Bio-Vesicles in Improving Anticancer Therapy,” Journal of Controlled Release 363 (2023): 290–348.
- 179Y. Li, R. Fu, Z. Duan, C. Zhu, and D. Fan, “Artificial Nonenzymatic Antioxidant MXene Nanosheet-Anchored Injectable Hydrogel as a Mild Photothermal-Controlled Oxygen Release Platform for Diabetic Wound Healing,” ACS Nano 16, no. 5 (2022): 7486–7502.
- 180Y. Luo, J. Li, Y. Hu, et al., “Injectable Thermo-Responsive Nano-Hydrogel Loading Triptolide for the Anti-Breast Cancer Enhancement via Localized Treatment Based on ‘Two Strikes’ Effects,” Acta Pharmaceutica Sinica B 10, no. 11 (2020): 2227–2245.
- 181M. Jin, Y. Hou, X. Quan, L. Chen, Z. Gao, and W. Huang, “Smart Polymeric Nanoparticles With pH-Responsive and PEG-Detachable Properties (II): Co-Delivery of Paclitaxel and VEGF siRNA for Synergistic Breast Cancer Therapy in Mice,” International Journal of Nanomedicine 16 (2021): 5479–5494.
- 182D. Li, J. Li, S. Wang, W. Teng, and Q. Wang, “Combined Self-Assembled iRGD Polymersomes for Effective Targeted siRNA Anti-Tumor Therapy,” International Journal of Nanomedicine 17 (2022): 5679–5696.
- 183S. Liu, F. Zhang, Y. Liang, et al., “Nanoparticle (NP)-Mediated APOC1 Silencing to Inhibit MAPK/ERK and NF-κB Pathway and Suppress Breast Cancer Growth and Metastasis,” Science China Life Sciences 66, no. 11 (2023): 2451–2465.
- 184M. Yang, C. Qin, L. Tao, et al., “Synchronous Targeted Delivery of TGF-β siRNA to Stromal and Tumor Cells Elicits Robust Antitumor Immunity Against Triple-Negative Breast Cancer by Comprehensively Remodeling the Tumor Microenvironment,” Biomaterials 301 (2023): 122253.
- 185Y. Song, C. Tang, and C. Yin, “Combination Antitumor Immunotherapy With VEGF and PIGF siRNA via Systemic Delivery of Multi-Functionalized Nanoparticles to Tumor-Associated Macrophages and Breast Cancer Cells,” Biomaterials 185 (2018): 117–132.
- 186S. Taghavi, A. HashemNia, F. Mosaffa, S. Askarian, K. Abnous, and M. Ramezani, “Preparation and Evaluation of Polyethylenimine-Functionalized Carbon Nanotubes Tagged With 5TR1 Aptamer for Targeted Delivery of Bcl-xL shRNA Into Breast Cancer Cells,” Colloids and Surfaces B: Biointerfaces 140 (2016): 28–39.
- 187J. Xiao, X. Duan, Q. Meng, et al., “Effective Delivery of p65 shRNA by Optimized Tween 85-polyethyleneimine Conjugate for Inhibition of Tumor Growth and Lymphatic Metastasis,” Acta Biomaterialia 10, no. 6 (2014): 2674–2683.
- 188C. H. Kapadia, S. A. Ioele, and E. S. Day, “Layer-by-Layer Assembled PLGA Nanoparticles Carrying miR-34a Cargo Inhibit the Proliferation and Cell Cycle Progression of Triple-Negative Breast Cancer Cells,” Journal of Biomedical Materials Research. Part A 108, no. 3 (2020): 601–613.
- 189Y. Kim, J. Choi, E. H. Kim, et al, “Design of PD-L1-Targeted Lipid Nanoparticles to Turn on PTEN for Efficient Cancer Therapy,” Advanced Science 11, no. 22 (2024): e2309917.
- 190X. Ma, S. Yang, T. Zhang, et al., “Bioresponsive Immune-Booster-Based Prodrug Nanogel for Cancer Immunotherapy,” Acta Pharmaceutica Sinica B 12, no. 1 (2022): 451–466.
- 191F. Hu, R. Zhang, W. Guo, et al., “PEGylated-PLGA Nanoparticles Coated With pH-Responsive Tannic Acid-Fe(Iii) Complexes for Reduced Premature Doxorubicin Release and Enhanced Targeting in Breast Cancer,” Molecular Pharmaceutics 18, no. 6 (2021): 2161–2173.
- 192M. Qian, G. Jiang, W. Guo, and R. Huang, “A Biodegradable Nanosuspension Locally Used for Inhibiting Postoperative Recurrence and Brain Metastasis of Breast Cancer,” Nano Letters 24, no. 10 (2024): 3165–3175.
- 193Y. Dai, H. Zhao, K. He, et al., “NIR-II Excitation Phototheranostic Nanomedicine for Fluorescence/Photoacoustic Tumor Imaging and Targeted Photothermal-Photonic Thermodynamic Therapy,” Small 17, no. 42 (2021): e2102527.
- 194X. Lu, L. Miao, W. Gao, et al., “Engineered PLGA Microparticles for Long-Term, Pulsatile Release of STING Agonist for Cancer Immunotherapy,” Science Translational Medicine 12, no. 556 (2020): eaaz6606.
- 195Y. P. Chen, L. Xu, T. W. Tang, et al., “STING Activator c-di-GMP-Loaded Mesoporous Silica Nanoparticles Enhance Immunotherapy Against Breast Cancer,” ACS Applied Materials & Interfaces 12, no. 51 (2020): 56741–56752.
- 196S. Zuo, Z. Wang, X. Jiang, et al., “Regulating Tumor Innervation by Nanodrugs Potentiates Cancer Immunochemotherapy and Relieve Chemotherapy-Induced Neuropathic Pain,” Biomaterials 309 (2024): 122603.
- 197S. Wang, X. Liu, S. Chen, et al., “Regulation of Ca(2+) Signaling for Drug-Resistant Breast Cancer Therapy With Mesoporous Silica Nanocapsule Encapsulated Doxorubicin/sIRNA Cocktail,” ACS Nano 13, no. 1 (2019): 274–283.
- 198L. Li, Y. Gao, Y. Zhang, et al., “A Biomimetic Nanogel System Restores Macrophage Phagocytosis for Magnetic Resonance Imaging-Guided Synergistic Chemoimmunotherapy of Breast Cancer,” Advanced Healthcare Materials 12, no. 26 (2023): e2300967.
- 199Y. Chen, Z. Yao, P. Liu, et al., “A Self-Assembly Nano-Prodrug for Triple-Negative Breast Cancer Combined Treatment by Ferroptosis Therapy and Chemotherapy,” Acta Biomaterialia 159 (2023): 275–288.
- 200X. Kong, Y. Qi, X. Wang, et al., “Nanoparticle Drug Delivery Systems and Their Applications as Targeted Therapies for Triple Negative Breast Cancer,” Progress in Materials Science 134 (2023): 101070.
- 201R. Vivek, R. Thangam, V. NipunBabu, et al., “Multifunctional HER2-antibody Conjugated Polymeric Nanocarrier-Based Drug Delivery System for Multi-Drug-Resistant Breast Cancer Therapy,” ACS Applied Materials & Interfaces 6, no. 9 (2014): 6469–6480.
- 202A. Da Silva-Candal, T. Brown, V. Krishnan, et al., “Shape Effect in Active Targeting of Nanoparticles to Inflamed Cerebral Endothelium Under Static and Flow Conditions,” Journal of Controlled Release 309 (2019): 94–105.
- 203A. Ahmad, F. Khan, R. K. Mishra, and R. Khan, “Precision Cancer Nanotherapy: Evolving Role of Multifunctional Nanoparticles for Cancer Active Targeting,” Journal of Medicinal Chemistry 62, no. 23 (2019): 10475–10496.
- 204C. Caro, A. Avasthi, J. M. Paez-Muñoz, M. Pernia Leal, and M. L. García-Martín, “Passive Targeting of High-Grade Gliomas via the EPR Effect: A Closed Path for Metallic Nanoparticles?,” Biomaterials Science 9, no. 23 (2021): 7984–7995.
- 205M. Abotaleb, P. Kubatka, M. Caprnda, et al., “Chemotherapeutic Agents for the Treatment of Metastatic Breast Cancer: An Update,” Biomedicine & Pharmacotherapy 101 (2018): 458–477.
- 206H. L. Chang, B. Schwettmann, H. L. McArthur, and I. S. Chan, “Antibody-Drug Conjugates in Breast Cancer: Overcoming Resistance and Boosting Immune Response,” Journal of Clinical Investigation 133, no. 18 (2023): e172156.
- 207D. Gao, S. Asghar, J. Ye, et al., “Dual-Targeted Enzyme-Sensitive Hyaluronic Acid Nanogels Loading Paclitaxel for the Therapy of Breast Cancer,” Carbohydrate Polymers 294 (2022): 119785.
- 208H. Ding, P. Tan, S. Fu, et al., “Preparation and Application of pH-Responsive Drug Delivery Systems,” Journal of Controlled Release 348 (2022): 206–238.
- 209Z. Zhang, R. Ma, and L. Shi, “Cooperative Macromolecular Self-Assembly Toward Polymeric Assemblies With Multiple and Bioactive Functions,” Accounts of Chemical Research 47, no. 4 (2014): 1426–1437.
- 210J. Lin, H. Yang, Y. Zhang, et al., “Ferrocene-Based Polymeric Nanoparticles Carrying Doxorubicin for Oncotherapeutic Combination of Chemotherapy and Ferroptosis,” Small 19, no. 2 (2023): e2205024.
- 211S. Zhang, H. Gao, and G. Bao, “Physical Principles of Nanoparticle Cellular Endocytosis,” ACS Nano 9, no. 9 (2015): 8655–8671.
- 212V. Jain, H. Kumar, H. V. Anod, et al., “A Review of Nanotechnology-Based Approaches for Breast Cancer and Triple-Negative Breast Cancer,” Journal of Controlled Release 326 (2020): 628–647.
- 213W. Ngamcherdtrakul, D. S. Bejan, W. Cruz-Muñoz, et al., “Targeted Nanoparticle for Co-Delivery of HER2 siRNA and a Taxane to Mirror the Standard Treatment of HER2+ Breast Cancer: Efficacy in Breast Tumor and Brain Metastasis,” Small 18, no. 11 (2022): e2107550.
- 214C. Hu, X. Yang, R. Liu, et al., “Coadministration of iRGD With Multistage Responsive Nanoparticles Enhanced Tumor Targeting and Penetration Abilities for Breast Cancer Therapy,” ACS Applied Materials & Interfaces 10, no. 26 (2018): 22571–22579.
- 215M. A. Amini, T. Ahmed, F. C. F. Liu, et al., “Exploring the Transformability of Polymer-Lipid Hybrid Nanoparticles and Nanomaterial-Biology Interplay to Facilitate Tumor Penetration, Cellular Uptake and Intracellular Targeting of Anticancer Drugs,” Expert Opinion on Drug Delivery 18, no. 7 (2021): 991–1004.
- 216C. Marques, M. J. Hajipour, C. Marets, et al., “Identification of the Proteins Determining the Blood Circulation Time of Nanoparticles,” ACS Nano 17, no. 13 (2023): 12458–12470.
- 217R. K. Jain, R. T. Tong, and L. L. Munn, “Effect of Vascular Normalization by Antiangiogenic Therapy on Interstitial Hypertension, Peritumor Edema, and Lymphatic Metastasis: Insights From a Mathematical Model,” Cancer Research 67, no. 6 (2007): 2729–2735.
- 218Q. Cheng, X. Shi, Y. Chen, et al., “Tumor Microenvironment-Activatable Nanosystem Capable of Overcoming Multiple Therapeutic Obstacles for Augmenting Immuno/Metal-Ion Therapy,” ACS Nano 18, no. 12 (2024): 8996–9010.
- 219M. Zhou, C. Zhou, H. Geng, et al., “EGCG-Enabled Deep Tumor Penetration of Phosphatase and Acidity Dual-Responsive Nanotherapeutics for Combinatory Therapy of Breast Cancer,” Small (2024): e2406245.
- 220M. Zhou, Y. Yao, S. Ma, et al., “Dual-Targeted and Dual-Sensitive Self-Assembled Protein Nanocarrier Delivering hVEGI-192 for Triple-Negative Breast Cancer,” International Journal of Biological Macromolecules 245 (2023): 125475.
- 221H. Sun, Y. Zhang, and Z. Zhong, “Reduction-Sensitive Polymeric Nanomedicines: An Emerging Multifunctional Platform for Targeted Cancer Therapy,” Advanced Drug Delivery Reviews 132 (2018): 16–32.
- 222H. Gao, Z. Cao, H. Liu, et al., “Multifunctional Nanomedicines-Enabled Chemodynamic-Synergized Multimodal Tumor Therapy via Fenton and Fenton-Like Reactions,” Theranostics 13, no. 6 (2023): 1974–2014.
- 223S. Shen, J. Qiu, D. Huo, and Y. Xia, “Nanomaterial-Enabled Photothermal Heating and Its Use for Cancer Therapy via Localized Hyperthermia,” Small 20, no. 7 (2024): e2305426.
- 224K. K. Lee, K. W. Park, S. C. Lee, and C. S. Lee, “Perfluorocarbon-Polyepinephrine Core-Shell Nanoparticles as a Near-Infrared Light Activatable Theranostic Platform for Bimodal Imaging-Guided Photothermal/Chemodynamic Synergistic Cancer Therapy,” Theranostics 15, no. 3 (2025): 1077–1093.
- 225S. Sarkar and N. Levi-Polyachenko, “Conjugated Polymer Nano-Systems for Hyperthermia, Imaging and Drug Delivery,” Advanced Drug Delivery Reviews 163–164 (2020): 40–64.
- 226J. Peng, Y. Xiao, W. Li, et al., “Photosensitizer Micelles Together With IDO Inhibitor Enhance Cancer Photothermal Therapy and Immunotherapy,” Advanced Science 5, no. 5 (2018): 1700891.
- 227Y. Qiu, Z. Wu, Y. Chen, et al., “Nano Ultrasound Contrast Agent for Synergistic Chemo-Photothermal Therapy and Enhanced Immunotherapy Against Liver Cancer and Metastasis,” Advanced Science 10, no. 21 (2023): e2300878.
- 228L. Zhao, F. Chang, Y. Tong, et al., “A Multifunctional Bimetallic Nanoplatform for Synergic Local Hyperthermia and Chemotherapy Targeting HER2-Positive Breast Cancer,” Advanced Science 11, no. 16 (2024): e2308316.
- 229H. Zhu, T. Li, X. Peng, et al, “Tumor Microenvironment-Driven Structural Transformation of Vanadium-Based MXenzymes to Amplify Oxidative Stress for Multimodal Tumor Therapy,” Advanced Science 12, no. 11 (2025): e2408998.
- 230S. G. Alamdari, M. Amini, N. Jalilzadeh, et al., “Recent Advances in Nanoparticle-Based Photothermal Therapy for Breast Cancer,” Journal of Controlled Release 349 (2022): 269–303.
- 231J. Kadkhoda, A. Tarighatnia, M. R. Tohidkia, N. D. Nader, and A. Aghanejad, “Photothermal Therapy-Mediated Autophagy in Breast Cancer Treatment: Progress and Trends,” Life Sciences 298 (2022): 120499.
- 232R. Han, Y. Xiao, Q. Yang, et al., “Ag(2)S Nanoparticle-Mediated Multiple Ablations Reinvigorates the Immune Response for Enhanced Cancer Photo-Immunotherapy,” Biomaterials 264 (2021): 120451.
- 233C. Xiao, X. Wang, S. Li, et al., “A Cuproptosis-Based Nanomedicine Suppresses Triple Negative Breast Cancers by Regulating Tumor Microenvironment and Eliminating Cancer Stem Cells,” Biomaterials 313 (2025): 122763.
- 234H. Yuan, C. Qiu, X. Wang, et al., “Engineering Semiconducting Polymeric Nanoagonists Potentiate cGAS-STING Pathway Activation and Elicit Long Term Memory Against Recurrence In Breast Cancer,” Advanced Materials 37, no. 4 (2025): e2406662.
- 235E. Ostańska, D. Aebisher, and D. Bartusik-Aebisher, “The Potential of Photodynamic Therapy in Current Breast Cancer Treatment Methodologies,” Biomedicine & Pharmacotherapy 137 (2021): 111302.
- 236S. Kwiatkowski, B. Knap, D. Przystupski, et al., “Photodynamic Therapy—Mechanisms, Photosensitizers and Combinations,” Biomedicine & Pharmacotherapy 106 (2018): 1098–1107.
- 237X. Huang, I. H. El-Sayed, W. Qian, and M. A. El-Sayed, “Cancer Cell Imaging and Photothermal Therapy in the Near-Infrared Region by Using Gold Nanorods,” Journal of the American Chemical Society 128, no. 6 (2006): 2115–2120.
- 238Y. Hao, Y. Chen, X. He, et al., “Polymeric Nanoparticles With ROS-Responsive Prodrug and Platinum Nanozyme for Enhanced Chemophotodynamic Therapy of Colon Cancer,” Advanced Science 7, no. 20 (2020): 2001853.
- 239D. Bechet, P. Couleaud, C. Frochot, M. L. Viriot, F. Guillemin, and M. Barberi-Heyob, “Nanoparticles as Vehicles for Delivery of Photodynamic Therapy Agents,” Trends in Biotechnology 26, no. 11 (2008): 612–621.
- 240Y. Li, D. Hu, M. Pan, et al., “Near-Infrared Light and Redox Dual-Activatable Nanosystems for Synergistically Cascaded Cancer Phototherapy With Reduced Skin Photosensitization,” Biomaterials 288 (2022): 121700.
- 241A. Roshanzadeh, H. C. D. Medeiros, C. K. Herrera, et al, “Next-Generation Photosensitizers: Cyanine-Carborane Salts for Superior Photodynamic Therapy of Metastatic Cancer,” Angewandte Chemie—International Edition in English 64, no.19 (2025): e202419759.
- 242X. Feng, Y. Zhang, W. Lin, et al., “A Self-Amplifying Photodynamic Biomedicine for Cascade Immune Activation Against Triple-Negative Breast Cancer,” Small 21, no.9 (2025): e2410214.
- 243Z. Zhang, Q. Zhao, Q. Xu, et al., “A Mitochondria-Interfering Nanocomplex Cooperates With Photodynamic Therapy to Boost Antitumor Immunity,” Biomaterials 317 (2025): 123094.
- 244H. Zhang, M. Cui, D. Tang, et al., “Localization of Cancer Cells for Subsequent Robust Photodynamic Therapy by ROS Responsive Polymeric Nanoparticles With Anti-Metastasis Complexes NAMI-A,” Advanced Materials 36, no. 14 (2024): e2310298.
- 245J. Zhang, X. Li, and L. Huang, “Non-Viral Nanocarriers for siRNA Delivery in Breast Cancer,” Journal of Controlled Release 190 (2014): 440–450.
- 246T. Kim, H. S. Han, K. Yang, et al., “Nanoengineered Polymeric RNA Nanoparticles for Controlled Biodistribution and Efficient Targeted Cancer Therapy,” ACS Nano 18, no. 11 (2024): 7972–7988.
- 247F. Wu, J. Yang, J. Liu, et al., “Signaling Pathways in Cancer-Associated Fibroblasts and Targeted Therapy for Cancer,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 218.
- 248H. Wang, Y. Li, G. Shi, et al., “A Novel Antitumor Strategy: Simultaneously Inhibiting Angiogenesis and Complement by Targeting VEGFA/PIGF and C3b/C4b,” Molecular Therapy—Oncolytics 16 (2020): 20–29.
- 249A. D. Wagner, C. Thomssen, J. Haerting, and S. Unverzagt, “Vascular-Endothelial-Growth-Factor (VEGF) Targeting Therapies for Endocrine Refractory or Resistant Metastatic Breast Cancer,” Cochrane Database of Systematic Reviews no. 7 (2012): Cd008941.
- 250Q. Ni, F. Zhang, Y. Zhang, et al., “In Situ shRNA Synthesis on DNA-Polylactide Nanoparticles to Treat Multidrug Resistant Breast Cancer,” Advanced Materials 30, no. 10 (2018).
- 251Z. Li, T. Guo, S. Zhao, and M. Lin, “The Therapeutic Effects of MUC1-C shRNA@Fe(3)O(4) Magnetic Nanoparticles in Alternating Magnetic Fields on Triple-Negative Breast Cancer,” International Journal of Nanomedicine 18 (2023): 5651–5670.
- 252M. Yang, Y. Zhang, M. Li, X. Liu, and M. Darvishi, “The Various Role of microRNAs in Breast Cancer Angiogenesis, With a Special Focus on Novel miRNA-Based Delivery Strategies,” Cancer Cell International 23, no. 1 (2023): 24.
- 253F. Abdalla, B. Singh, and H. K. Bhat, “MicroRNAs and Gene Regulation in Breast Cancer,” Journal of Biochemical and Molecular Toxicology 34, no. 11 (2020): e22567.
- 254B. Roy, S. Ghose, and S. Biswas, “Therapeutic Strategies for miRNA Delivery to Reduce Hepatocellular Carcinoma,” Seminars in Cell & Developmental Biology 124 (2022): 134–144.
- 255S. Ohno, M. Takanashi, K. Sudo, et al., “Systemically Injected Exosomes Targeted to EGFR Deliver Antitumor microRNA to Breast Cancer Cells,” Molecular Therapy 21, no. 1 (2013): 185–191.
- 256I. Pérez-Núñez, C. Rozalén, J. Á. Palomeque, et al., “LCOR Mediates Interferon-Independent Tumor Immunogenicity and Responsiveness to Immune-Checkpoint Blockade in Triple-Negative Breast Cancer,” Nature Cancer 3, no. 3 (2022): 355–370.
- 257S. Van Lint, C. Goyvaerts, S. Maenhout, et al., “Preclinical Evaluation of TriMix and Antigen mRNA-Based Antitumor Therapy,” Cancer Research 72, no. 7 (2012): 1661–1671.
- 258B. De Keersmaecker, S. Claerhout, J. Carrasco, et al., “TriMix and Tumor Antigen mRNA Electroporated Dendritic Cell Vaccination Plus Ipilimumab: Link Between T-Cell Activation and Clinical Responses In Advanced Melanoma,” Journal for Immunotherapy of Cancer 8, no. 1 (2020): e000329.
- 259Y. Barenholz, “Doxil®—The First FDA-Approved Nano-Drug: Lessons Learned,” Journal of Controlled Release 160, no. 2 (2012): 117–134.
- 260D. M. Vail, M. A. Amantea, G. T. Colbern, F. J. Martin, R. A. Hilger, and P. K. Working, “Pegylated Liposomal Doxorubicin: Proof of Principle Using Preclinical Animal Models and Pharmacokinetic Studies,” Seminars in Oncology 31, no. 6 Suppl 13 (2004): 16–35.
- 261M. E. R. O'Brien, N. Wigler, M. Inbar, et al., “Reduced Cardiotoxicity and Comparable Efficacy In a Phase III Trial of Pegylated Liposomal Doxorubicin HCl (CAELYX/Doxil) Versus Conventional Doxorubicin for First-Line Treatment of Metastatic Breast Cancer,” Annals of Oncology 15, no. 3 (2004): 440–449.
- 262H. Yuan, H. Guo, X. Luan, et al., “Albumin Nanoparticle of Paclitaxel (Abraxane) Decreases While Taxol Increases Breast Cancer Stem Cells in Treatment of Triple Negative Breast Cancer,” Molecular Pharmaceutics 17, no. 7 (2020): 2275–2286.
- 263E. Miele, G. P. Spinelli, E. Miele, F. Tomao, and S. Tomao, “Albumin-Bound Formulation of Paclitaxel (Abraxane ABI-007) in the Treatment of Breast Cancer,” International Journal of Nanomedicine 4 (2009): 99–105.
- 264Y. Yoneshima, S. Morita, M. Ando, et al., “Phase 3 Trial Comparing Nanoparticle Albumin-Bound Paclitaxel With Docetaxel for Previously Treated Advanced NSCLC,” Journal of Thoracic Oncology 16, no. 9 (2021): 1523–1532.
- 265M. Untch, C. Jackisch, A. Schneeweiss, et al., “Nab-Paclitaxel Versus Solvent-Based Paclitaxel in Neoadjuvant Chemotherapy for Early Breast Cancer (Geparsepto-Gbg 69): A Randomised, Phase 3 Trial,” Lancet Oncology 17, no. 3 (2016): 345–356.
- 266K. Tezuka, T. Takashima, S. Kashiwagi, et al., “Phase I Study of Nanoparticle Albumin-Bound Paclitaxel, Carboplatin and Trastuzumab In Women With Human Epidermal Growth Factor Receptor 2-overexpressing Breast Cancer,” Molecular and Clinical Oncology 6, no. 4 (2017): 534–538.
- 267D. Li, C. F. van Nostrum, E. Mastrobattista, T. Vermonden, and W. E. Hennink, “Nanogels for Intracellular Delivery of Biotherapeutics,” Journal of Controlled Release 259 (2017): 16–28.
- 268R. T. Chacko, J. Ventura, J. Zhuang, and S. Thayumanavan, “Polymer Nanogels: A Versatile Nanoscopic Drug Delivery Platform,” Advanced Drug Delivery Reviews 64, no. 9 (2012): 836–851.
- 269F. Dang, L. Nie, J. Zhou, et al., “Inhibition of CK1ε Potentiates the Therapeutic Efficacy of CDK4/6 Inhibitor in Breast Cancer,” Nature Communications 12, no. 1 (2021): 5386.
- 270A. Fassl, Y. Geng, and P. Sicinski, “CDK4 and CDK6 Kinases: From Basic Science to Cancer Therapy,” Science 375, no. 6577 (2022): eabc1495.
- 271J. Ma, L. Qin, and X. Li, “Role of STAT3 Signaling Pathway in Breast Cancer,” Cell Communication and Signaling 18, no. 1 (2020): 33.
- 272T. Liu, S. Song, X. Wang, and J. Hao, “Small-Molecule Inhibitors of Breast Cancer-Related Targets: Potential Therapeutic Agents for Breast Cancer,” European Journal of Medicinal Chemistry 210 (2021): 112954.
- 273K. M. Au, A. Z. Wang, and S. I. Park, “Pretargeted Delivery of PI3K/mTOR Small-Molecule Inhibitor-Loaded Nanoparticles for Treatment of Non-Hodgkin's Lymphoma,” Science Advances 6, no. 14 (2020): eaaz9798.
- 274N. Saadat, F. Liu, B. Haynes, et al., “Nano-Delivery of RAD6/Translesion Synthesis Inhibitor SMI#9 for Triple-Negative Breast Cancer Therapy,” Molecular Cancer Therapeutics 17, no. 12 (2018): 2586–2597.
- 275N. Xu, D. C. Palmer, A. C. Robeson, et al., “STING Agonist Promotes CAR T Cell Trafficking and Persistence in Breast Cancer,” Journal of Experimental Medicine 218, no. 2 (2021): e20200844.
- 276E. L. Dane, A. Belessiotis-Richards, C. Backlund, et al., “STING Agonist Delivery by Tumour-Penetrating PEG-Lipid Nanodiscs Primes Robust Anticancer Immunity,” Nature Materials 21, no. 6 (2022): 710–720.
- 277M. Farshbafnadi, A. Pastaki Khoshbin, and N. Rezaei, “Immune Checkpoint Inhibitors for Triple-Negative Breast Cancer: From Immunological Mechanisms to Clinical Evidence,” International Immunopharmacology 98 (2021): 107876.
- 278N. Gaynor, J. Crown, and D. M. Collins, “Immune Checkpoint Inhibitors: Key Trials and an Emerging Role in Breast Cancer,” Seminars in Cancer Biology 79 (2022): 44–57.
- 279X. Ren, Z. Cheng, J. He, et al., “Inhibition of Glycolysis-Driven Immunosuppression With a Nano-Assembly Enhances Response to Immune Checkpoint Blockade Therapy in Triple Negative Breast Cancer,” Nature Communications 14, no. 1 (2023): 7021.
- 280Y. Song, L. Bugada, R. Li, et al., “Albumin Nanoparticle Containing a PI3Kγ Inhibitor and Paclitaxel in Combination With α-PD1 Induces Tumor Remission of Breast Cancer in Mice,” Science Translational Medicine 14, no. 643 (2022): eabl3649.
- 281M. Shastry, S. Jacob, H. S. Rugo, and E. Hamilton, “Antibody-Drug Conjugates Targeting TROP-2: Clinical Development in Metastatic Breast Cancer,” Breast 66 (2022): 169–177.
- 282A. Dri, G. Arpino, G. Bianchini, et al., “Breaking Barriers in Triple Negative Breast Cancer (TNBC)—Unleashing the Power of Antibody-Drug Conjugates (ADCs),” Cancer Treatment Reviews 123 (2024): 102672.
- 283G. von Minckwitz, C. S. Huang, M. S. Mano, et al., “Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer,” New England Journal of Medicine 380, no. 7 (2019): 617–628.
- 284J. Cortés, S. B. Kim, W. P. Chung, et al., “Trastuzumab Deruxtecan Versus Trastuzumab Emtansine for Breast Cancer,” New England Journal of Medicine 386, no. 12 (2022): 1143–1154.
- 285S. Modi, W. Jacot, T. Yamashita, et al., “Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer,” New England Journal of Medicine 387, no. 1 (2022): 9–20.
- 286A. Bardia, S. A. Hurvitz, S. M. Tolaney, et al., “Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer,” New England Journal of Medicine 384, no. 16 (2021): 1529–1541.
- 287N. Aubrey, E. Allard-Vannier, C. Martin, et al., “Site-Specific Conjugation of Auristatins Onto Engineered scFv Using Second Generation Maleimide to Target HER2-positive Breast Cancer In Vitro,” Bioconjugate Chemistry 29, no. 11 (2018): 3516–3521.
- 288E. Douez, E. Allard-Vannier, I. A. M. Amar, et al., “Branched Pegylated Linker-Auristatin to Control Hydrophobicity for the Production of Homogeneous Minibody-Drug Conjugate against HER2-positive Breast Cancer,” Journal of Controlled Release 366 (2024): 567–584.
- 289X. Xia, X. Yang, W. Huang, X. Xia, and D. Yan, “Self-Assembled Nanomicelles of Affibody-Drug Conjugate With Excellent Therapeutic Property to Cure Ovary and Breast Cancers,” Nano-Micro Letters 14, no. 1 (2021): 33.
- 290J. Li, M. Zhao, W. Liang, S. Wu, Z. Wang, and D. Wang, “Codelivery of Shikonin and siTGF-β for Enhanced Triple Negative Breast Cancer Chemo-Immunotherapy,” Journal of Controlled Release 342 (2022): 308–320.
- 291C. M. Jogdeo, S. Panja, S. Kanvinde, E. Kapoor, K. Siddhanta, and D. Oupický, “Advances in Lipid-Based Codelivery Systems for Cancer and Inflammatory Diseases,” Advanced Healthcare Materials 12, no. 7 (2023): e2202400.
- 292Q. Y. Meng, H. L. Cong, H. Hu, and F. J. Xu, “Rational Design and Latest Advances of Codelivery Systems for Cancer Therapy,” Materials Today Bio 7 (2020): 100056.
- 293C. Song, M. Zhan, Z. Ouyang, et al., “Core-Shell Tecto Dendrimer-Mediated Cooperative Chemoimmunotherapy of Breast Cancer,” Journal of Controlled Release 358 (2023): 601–611.
- 294W. Xu, Z. Zeng, Y. Tang, et al., “Spatiotemporal-Controllable ROS-Responsive Camptothecin Nano-Bomb for Chemo/Photo/Immunotherapy In Triple-Negative Breast Cancer,” Journal of Nanobiotechnology 22, no. 1 (2024): 798.
- 295D. Yao, Y. Wang, K. Bian, B. Zhang, and D. Wang, “A Self-Cascaded Unimolecular Prodrug for pH-Responsive Chemotherapy and Tumor-Detained Photodynamic-Immunotherapy of Triple-Negative Breast Cancer,” Biomaterials 292 (2023): 121920.
- 296J. Chang, L. Mo, J. Song, et al., “A pH-Responsive Mesoporous Silica Nanoparticle-Based Drug Delivery System for Targeted Breast Cancer Therapy,” Journal of Materials Chemistry B 10, no. 17 (2022): 3375–3385.
- 297A. K. Grosskopf, L. Labanieh, D. D. Klysz, et al., “Delivery of CAR-T Cells in a Transient Injectable Stimulatory Hydrogel Niche Improves Treatment of Solid Tumors,” Science Advances 8, no. 14 (2022): eabn8264.
- 298Z. Zheng, X. Yang, Y. Zhang, et al., “An Injectable and pH-Responsive Hyaluronic Acid Hydrogel as Metformin Carrier for Prevention of Breast Cancer Recurrence,” Carbohydrate Polymers 304 (2023): 120493.
- 299M. Zhang, N. Ying, J. Chen, et al., “Engineering a pH-Responsive Polymeric Micelle Co-Loaded With Paclitaxel and Triptolide for Breast Cancer Therapy,” Cell Proliferation 57, no. 6 (2024): e13603.
- 300S. Khan, M. M. N. Babadaei, A. Hasan, et al., “Enzyme-Polymeric/Inorganic Metal Oxide/Hybrid Nanoparticle Bio-Conjugates in the Development of Therapeutic and Biosensing Platforms,” Journal of Advanced Research 33 (2021): 227–239.
- 301K. Li, C. Lin, M. Li, et al., “Multienzyme-Like Reactivity Cooperatively Impairs Glutathione Peroxidase 4 and Ferroptosis Suppressor Protein 1 Pathways in Triple-Negative Breast Cancer for Sensitized Ferroptosis Therapy,” ACS Nano 16, no. 2 (2022): 2381–2398.
- 302Y. Liu, R. Lu, M. Li, et al., “Dual-Enzyme Decorated Semiconducting Polymer Nanoagents for Second Near-Infrared Photoactivatable Ferroptosis-Immunotherapy,” Materials Horizons 11, no. 10 (2024): 2406–2419.
- 303C. Hu, Y. Song, Y. Zhang, et al., “Sequential Delivery of PD-1/Pd-L1 Blockade Peptide and IDO Inhibitor for Immunosuppressive Microenvironment Remodeling via an MMP-2 Responsive Dual-Targeting Liposome,” Acta Pharmaceutica Sinica B 13, no. 5 (2023): 2176–2187.
- 304D. Li, R. Zhang, G. Liu, Y. Kang, and J. Wu, “Redox-Responsive Self-Assembled Nanoparticles for Cancer Therapy,” Advanced Healthcare Materials 9, no. 20 (2020): e2000605.
- 305Y. He, Y. Xu, Y. Huang, et al., “Redox Sensitive Nano-Capsules Self-Assembled From Hyaluronic Acid-Hydroxychloroquine Conjugates for CD44-targeted Delivery of Hydroxychloroquine to Combat Breast Cancer Metastasis in Vitro and In Vivo,” Colloids and Surfaces B: Biointerfaces 210 (2022): 112249.
- 306L. Yao, X. Zhu, Y. Shan, L. Zhang, J. Yao, and H. Xiong, “Recent Progress in Anti-Tumor Nanodrugs Based on Tumor Microenvironment Redox Regulation,” Small 20, no. 25 (2024): e2310018.
- 307V. Ghalehkhondabi, M. Soleymani, and A. Fazlali, “Synthesis of Quercetin-Loaded Hyaluronic Acid-Conjugated pH/Redox Dual-Stimuli Responsive Poly(Methacrylic Acid)/Mesoporous Organosilica Nanoparticles for Breast Cancer Targeted Therapy,” International Journal of Biological Macromolecules 263, no. Pt 1 (2024): 130168.
- 308Y. Zhang, J. Zhou, X. Chen, et al., “Modulating Tumor-Stromal Crosstalk via a Redox-Responsive Nanomedicine for Combination Tumor Therapy,” Journal of Controlled Release 356 (2023): 525–541.
- 309L. Li, P. Zhang, C. Li, Y. Guo, and K. Sun, “In Vitro/Vivo Antitumor Study of Modified-Chitosan/Carboxymethyl Chitosan “Boosted” Charge-Reversal Nanoformulation,” Carbohydrate Polymers 269 (2021): 118268.
- 310X. Y. He, B. Y. Liu, J. L. Wu, S. L. Ai, R. X. Zhuo, and S. X. Cheng, “A Dual Macrophage Targeting Nanovector for Delivery of Oligodeoxynucleotides to Overcome Cancer-Associated Immunosuppression,” ACS Applied Materials & Interfaces 9, no. 49 (2017): 42566–42576.
- 311H. Peng, L. Qiao, G. Shan, et al., “Stepwise Responsive Carboxymethyl Chitosan-Based Nanoplatform for Effective Drug-Resistant Breast Cancer Suppression,” Carbohydrate Polymers 291 (2022): 119554.
- 312H. Parhiz, J. S. Brenner, P. N. Patel, et al., “Added to Pre-Existing Inflammation, mRNA-Lipid Nanoparticles Induce Inflammation Exacerbation (IE),” Journal of Controlled Release 344 (2022): 50–61.
- 313H. Soo Choi, W. Liu, P. Misra, et al., “Renal Clearance of Quantum Dots,” Nature Biotechnology 25, no. 10 (2007): 1165–1170.
- 314Z. Li, A. Ma, I. Miller, et al., “Development of Anti-PEG IgG/IgM/IgE ELISA Assays for Profiling Anti-PEG Immunoglobulin Response in PEG-Sensitized Individuals and Patients With Alpha-Gal Allergy,” Journal of Controlled Release 366 (2024): 342–348.
- 315Q. Yang, T. M. Jacobs, J. D. McCallen, et al., “Analysis of Pre-Existing IgG and IgM Antibodies Against Polyethylene Glycol (PEG) In the General Population,” Analytical Chemistry 88, no. 23 (2016): 11804–11812.
- 316T. Suzuki, Y. Suzuki, T. Hihara, et al., “PEG Shedding-Rate-Dependent Blood Clearance of PEGylated Lipid Nanoparticles in Mice: Faster PEG Shedding Attenuates Anti-PEG IgM Production,” International Journal of Pharmaceutics 588 (2020): 119792.
- 317A. Rocca, L. Cecconetto, A. Passardi, et al., “A Phase Ib Study of Lapatinib Plus Pegylated Liposomal Doxorubicin in Patients With Advanced HER2-positive Breast Cancer,” Journal of Clinical Oncology 34, no. 15_suppl (2016): 600.
10.1200/JCO.2016.34.15_suppl.600 Google Scholar
- 318A. Albanese, P. S. Tang, and W. C. W. Chan, “The Effect of Nanoparticle Size, Shape, and Surface Chemistry on Biological Systems,” Annual Review of Biomedical Engineering 14 (2012): 1–16.
- 319E. A. Sykes, J. Chen, G. Zheng, and W. C. W. Chan, “Investigating the Impact of Nanoparticle Size on Active and Passive Tumor Targeting Efficiency,” ACS Nano 8, no. 6 (2014): 5696–5706.
- 320H. F. Krug, “Nanosafety Research--Are We on the Right Track?,” Angewandte Chemie International Edition 53, no. 46 (2014): 12304–12319.
- 321J. McClements and D. J. McClements, “Standardization of Nanoparticle Characterization: Methods for Testing Properties, Stability, and Functionality of Edible Nanoparticles,” Critical Reviews in Food Science and Nutrition 56, no. 8 (2016): 1334–1362.
- 322C. Liu, X. Yan, Y. Zhang, et al., “Oral Administration of Turmeric-Derived Exosome-Like Nanovesicles With Anti-Inflammatory and Pro-Resolving Bioactions for Murine Colitis Therapy,” Journal of Nanobiotechnology 20, no. 1 (2022): 206.
- 323L. Xin, C. Wei, X. Tong, et al., “In Situ Delivery of Apoptotic Bodies Derived From Mesenchymal Stem Cells via a Hyaluronic Acid Hydrogel: A Therapy for Intrauterine Adhesions,” Bioactive Materials 12 (2022): 107–119.
- 324Q. Cheng, T. Wei, L. Farbiak, L. T. Johnson, S. A. Dilliard, and D. J. Siegwart, “Selective Organ Targeting (SORT) Nanoparticles for Tissue-Specific mRNA Delivery and CRISPR-Cas Gene Editing,” Nature Nanotechnology 15, no. 4 (2020): 313–320.
- 325P. R. Cullis and M. J. Hope, “Lipid Nanoparticle Systems for Enabling Gene Therapies,” Molecular Therapy 25, no. 7 (2017): 1467–1475.
- 326W. Wang, X. Liu, X. Zheng, H. J. Jin, and X. Li, “Biomineralization: An Opportunity and Challenge of Nanoparticle Drug Delivery Systems for Cancer Therapy,” Advanced Healthcare Materials 9, no. 22 (2020): e2001117.
- 327Y. Q. Deng, N. N. Zhang, Y. F. Zhang, et al., “Lipid Nanoparticle-Encapsulated mRNA Antibody Provides Long-Term Protection Against SARS-CoV-2 in Mice and Hamsters,” Cell Research 32, no. 4 (2022): 375–382.
- 328D. Ho, C. H. K. Wang, and E. K. H. Chow, “Nanodiamonds: The Intersection of Nanotechnology, Drug Development, and Personalized Medicine,” Science Advances 1, no. 7 (2015): e1500439.
- 329Y. Zhang, M. Leonard, Y. Shu, et al., “Overcoming Tamoxifen Resistance of Human Breast Cancer by Targeted Gene Silencing Using Multifunctional pRNA Nanoparticles,” ACS Nano 11, no. 1 (2017): 335–346.
- 330A. C. Tremain, R. P. Wallace, K. M. Lorentz, et al., “Synthetically Glycosylated Antigens for the Antigen-Specific Suppression of Established Immune Responses,” Nature Biomedical Engineering 7, no. 9 (2023): 1142–1155.
- 331J. Yin, T. Lang, D. Cun, et al., “pH-Sensitive Nano-Complexes Overcome Drug Resistance and Inhibit Metastasis of Breast Cancer by Silencing Akt Expression,” Theranostics 7, no. 17 (2017): 4204–4216.
- 332S. Peng, F. Xiao, M. Chen, and H. Gao, “Tumor-Microenvironment-Responsive Nanomedicine for Enhanced Cancer Immunotherapy,” Advanced Science 9, no. 1 (2022): e2103836.
- 333Y. He, Z. Li, C. Cong, et al., “Pyroelectric Catalysis-Based ‘Nano-Lymphatic’ Reduces Tumor Interstitial Pressure for Enhanced Penetration and Hydrodynamic Therapy,” ACS Nano 15, no. 6 (2021): 10488–10501.
- 334H. Ye, K. Wang, Q. Lu, et al., “Nanosponges of Circulating Tumor-Derived Exosomes for Breast Cancer Metastasis Inhibition,” Biomaterials 242 (2020): 119932.
- 335F. Sun, S. Ning, X. Fan, et al., “Engineered Cytomembrane Nanovesicles Trigger In Situ Storm of Engineered Extracellular Vesicles for Cascade Tumor Penetration and Immune Microenvironment Remodeling,” Nano Today 61 (2025): 102604.
- 336S. Ghosh, A. Javia, S. Shetty, et al., “Triple Negative Breast Cancer and Non-Small Cell Lung Cancer: Clinical Challenges and Nano-Formulation Approaches,” Journal of Controlled Release 337 (2021): 27–58.
- 337J. Min, H. Im, M. Allen, et al., “Computational Optics Enables Breast Cancer Profiling in Point-of-Care Settings,” ACS Nano 12, no. 9 (2018): 9081–9090.