Abstract
Background: Breast cancer remains a major global health burden despite therapeutic advances, necessitating further innovations to address persistent challenges like therapeutic resistance, adverse effects, and mortality in subgroups. Emerging technologies offer promising opportunities in this regard through data-driven, precise, and personalized approaches.
Main body: Omics-guided therapies leverage genome, epigenome, and other profiling to enable better characterization of complex cancer molecular alterations driving selection and monitoring of tailored treatment regimens. Gut microbiome manipulation via probiotics, prebiotics or fecal transplantation aims to counter dysbiosis linked to breast cancer progression. Nanotechnology-engineered platforms offer smart drug delivery selectively to tumors and integrated diagnostic-therapeutic functions. Artificial intelligence algorithms analyze complex datasets and images to extract detailed quantitative signatures guiding prognostic decisions and personalized care. Traditional Chinese medicine encompassing herbal remedies and acupuncture demonstrates anticancer effects in preclinical models, with early trials exploring integration with chemotherapy.
Conclusion: Substantial progress has been made in breast cancer management, yet significant unmet needs persist. Emerging modalities have shown tremendous translational potential through molecularly targeted, immunomodulatory, data-driven approaches improving upon conventional methods regarding precision, effectiveness, and quality of life. Incremental integration of these innovative technologies promises to transform breast cancer care.
Keywords
Breast cancer, Precision oncology, Omics microbiome, Nanotechnology, Artificial intelligence, Traditional Chinese medicine
Background
Breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer-related mortality in women worldwide. Incidence rates continue to rise globally. Breast cancer encompasses a heterogeneous group of tumors with varying molecular features, clinical behaviors, and responses to therapy. While survival rates have improved, challenges persist such as invasive cancers, recurrent metastatic disease, and mortality among subgroups [1-2].
The current clinical management of breast cancer involves surgery combined with systemic chemotherapy, radiation, endocrine and targeted therapies. These interventions have substantially improved prognosis, yet intrinsic and acquired resistance remains problematic. Chemotherapies and radiation are associated with adverse effects impacting quality of life. Additionally, optimal protocols for breast cancer subtypes and high-risk groups warrant further research. Substantial drug costs also limit access to cutting-edge treatments. Overall, there is impetus to develop more precise, effective, and well-tolerated approaches through emerging technologies that build upon foundational breast cancer therapies [3-6].
This review thoroughly examines latest high-potential modalities including omics profiling, immune-directed vaccines, microbiome alteration, nanotherapeutics, artificial intelligence, and traditional medicines for future breast cancer solution development. Synthesizing across these cutting-edge areas elucidates promising near-term prospects to tackle disease heterogeneity, therapeutic resistance, adverse effects, accessibility, and precision demands - bridging gaps left by current gold standards to guide next-generation treatment and monitoring.
Emerging Vaccine Approaches
Cancer antigen vaccines
Vaccines targeting tumor-associated antigens (TAAs) or neoantigens have shown promising results in early preclinical and clinical studies for breast cancer. TAAs are antigens produced by tumor cells that can elicit an immune response. Neoantigens arise from somatic tumor mutations and are unique to an individual's cancer. Animal models have supported the immunogenicity and protective capacity of TAA and neoantigen-directed vaccines. Early phase clinical trials in breast cancer patients have demonstrated safety and the ability to generate antigen-specific T-cell responses after vaccination [7-11].
Tumor-associated antigens, neoantigens: Tumor cells express antigens that are either normal self-antigens overexpressed in tumors or novel antigens created by mutations. Both categories can be leveraged as targets for cancer vaccines. Preclinical studies have identified various TAAs that could serve as vaccine targets in breast cancer. Neoantigens tailored to a tumor's mutation profile also represent highly specific targets [12,13].
Animal models, early clinical trials: Animal models including mice and non-human primates have been utilized to establish proof-of-concept for vaccines targeting TAAs such as HER2. Findings have indicated vaccines can induce antigen-specific immunity that impacts tumor growth. Early clinical evaluation has progressed to phase I and II trials exploring safety and immunogenicity. Small randomized trials have suggested clinically meaningful benefits in patients with breast cancer [14-18]. As depicted in Figure 1, stem cells have emerged as a promising source for engineering 3D organoid models that recapitulate the complex structure and function of organs with high fidelity. Studies demonstrate that stem cells are more stable than differentiated cells, enabling the formation of heterogeneous populations containing stem, differentiated, and functional cell types that accurately model tissue physiology and pathology [19].
Figure 1: personalized drug screening and precision oncology applications [19].
DNA vaccines
DNA vaccines involve direct injection of genetically engineered DNA to produce target antigens. This approach allows in vivo production of antigens to activate the immune system. DNA vaccines are relatively stable, cost-efficient, and safer compared to other vaccine platforms. Evidence for DNA vaccines comes from animal models and human clinical trials are ongoing [20,21].
Vector-based anti-cancer vaccines
Viral vectors including adenovirus, vaccinia, and others have been engineered as anti-cancer vaccines. They can induce robust T cell responses due to natural adjuvanticity. Vector-based methods demonstrate protection against tumor progression in preclinical breast cancer models. Several phase I/II trials indicate safety and immunogenicity, with evidence of clinical responses [22-24].
Omics-Guided Therapies
The development of advanced "omics" technologies that permit high-throughput analysis of the genome, epigenome, transcriptome, proteome, and metabolome have enabled better characterization of the complex molecular alterations driving breast cancer. Leveraging omics approaches has facilitated more precise therapy selection, monitoring of treatment response, and identification of new therapeutic targets and agents.
Genomics: mutation profiling, microarrays
Genomic analysis examining mutations, polymorphisms and expression changes associated with breast cancer has uncovered molecular subtypes, prognostic gene signatures for personalized treatment, and actionable alterations matching targeted drugs. Genome sequencing and DNA microarrays now allow comprehensive examination of the genetic basis of a patient's cancer [25-27].
Epigenomics: miRNA, DNA methylation, histone modifications
The study of epigenomic modifications regulating gene expression has revealed new biomarkers and therapeutic opportunities in breast cancer. Aberrant miRNA levels, altered DNA methylation, and histone modification patterns are linked to tumor progression and response to certain therapies. Epigenetic signatures are emerging to guide management and counter drug resistance [28,29].
Proteomics/metabolomics: Biomarkers, insights into pathogenesis
Advances in proteomic and metabolomic profiling have enabled detection of protein and metabolic signatures associated with breast cancer initiation, diagnosis, staging, and treatment outcomes. Novel proteomic approaches can identify circulating tumor cells and tumor-educated blood platelets revealing protein biomarkers. Metabolomics examines metabolic reprogramming in cancer cells. Together, these strategies provide insights about pathological changes as well as biomarkers detectable in liquid biopsies [30-35].
Applications for therapy selection, monitoring, novel targets.
Omics information can assist in matching patients with tailored treatments, monitoring therapeutic response, and revealing new drug targets. For example, genomic tests identify candidates for HER2-targeted therapy. Signatures are emerging to predict immunotherapy efficacy. Metabolomics reveals stem-cell dependent metabolic pathways against which drugs could be directed. Integrative analysis of multi-omics data may have the greatest utility for precision oncology applications [36.37].
Microbiome Manipulation
The human microbiome has emerged as a modulator of anti-cancer immune responses. There is growing evidence that the gut microbiota influences breast cancer risk and progression, representing a new target for prevention and therapy. Modifying the microbiome through probiotics, prebiotics, bacteriotherapy or fecal transplantation may confer anti-tumor effects.
Gut microbiota dysbiosis in breast cancer
Compared to healthy women, breast cancer patients exhibit gut microbiota imbalance or dysbiosis characterized by decreased bacterial diversity and alterations in the abundance of certain bacteria. Dysbiosis disrupts microbiome interactions with host immunity. Animal models support direct pro-tumorigenic microbial effects promoting breast cancer growth and metastasis [38-39].
Probiotics, prebiotics, bacteriotherapy
Strategies that increase levels of anti-inflammatory and anti-cancer phytochemical-producing bacteria may counter breast cancer-associated microbiota changes. Probiotic supplements can modulate gut dysbiosis and inhibit tumor progression in preclinical breast cancer models. Prebiotics promote the expansion of protective resident microbes. Bacteriotherapy involves oral consumption or injection of microbial strains with anti-tumor capabilities [40-45].
Fecal microbiota transplantation
Introducing fecal material from healthy donors into breast tumor–bearing mice delays tumor growth and enhances immune surveillance through normalization of gut microbiota. Early clinical research on fecal microbiota transplantation in cancer patients shows potential to attenuate common treatment side effects as well as modify anti-tumor immunity [46,47].
Nanotechnology-Based Precision Approaches
Nanotechnology-engineered devices and systems have shown tremendous potential to improve the precision of breast cancer diagnosis and therapy. Nanoparticle delivery platforms can transport therapeutics selectively to tumors, overcome drug resistance mechanisms, and integrate both imaging and therapeutic functions into unified “theranostic” systems.
Smart nanoformulations for chemotherapeutics
Conventional chemotherapies have limitations in water insolubility, lack of tumor specificity and dose-limiting systemic toxicities. "Smart" nanoformulations such as liposomes, dendrimers, nanogels and polymeric nanoparticles can encapsulate chemotherapeutics to improve pharmacokinetics and biodistribution, target tumors through surface modification, and enable controlled drug release at tumor sites through engineered biomaterials and external triggers. This increases intratumoral drug delivery and antitumor efficacy while reducing off-target effects [48-50].
Theranostic nanoparticles
"Theranostic" nanoparticles with both imaging and drug delivery capabilities permit spatiotemporal monitoring and individualized optimization of therapy. By integrating modalities like fluorescence dyes, radioisotope labeling, Raman imaging and MRI contrast, nanoparticles can provide real-time insight into in vivo behavior. An early example is the FDA approval of Abextram, a liposomal nanoformulation harboring an imaging agent and doxorubicin for metastatic breast cancer treatment [51,52].
Applications in combination therapy, drug resistance
Nanotechnology platforms have potential to improve combinatorial treatments and counter breast cancer drug resistance. Co-delivery of synergistic chemotherapeutic combinations using a single nanoparticle carrier may maximize efficacy. Strategies are also being developed to target nanoparticles to breast cancer stem cells responsible for tumor regeneration and therapeutic recalcitrance. Overall, multifunctional nanoparticle technologies offer opportunities to overcome existing challenges in breast cancer management [53-56].
Artificial Intelligence and Breast Cancer Management
Artificial intelligence (AI) encompasses advanced analytical techniques enabling machines to perform tasks mimicking human cognition and decision making as depicted in Table 1. The application of AI is transforming all aspects of breast cancer care from risk assessment to treatment.
|
AI Technology |
Application |
Purpose |
Development Stage |
|
Deep learning image analysis |
Automated screening mammography |
Early breast cancer detection |
FDA-approved algorithms, improved cancer detection over human review in studies |
|
Convolutional neural networks |
Histopathology image classification |
Diagnosis and subtype categorization |
Algorithms approaching pathologist-level accuracy, integration into hospital workflows |
|
Multi-layer neural networks |
Risk modeling with clinical datasets |
Assessment of prognosis, recurrence risk |
Validation ongoing in retrospective patient cohorts |
|
Random forest models |
Gene expression signature analysis |
Prediction of relapse, treatment response |
Commercially available prognostic tests guided by AI-derived biomarkers |
|
Deep reinforcement learning |
Treatment recommendation systems |
Evidence-based guidelines for precision therapy |
Pilot studies demonstrating feasibility to optimize regimens accounting for patient factors |
Medical imaging analysis
AI algorithms can rapidly analyze complex medical images and extract detailed quantitative data surpassing human capability. Machine learning has enabled automated breast cancer screening and improved diagnostic accuracy from mammograms and histopathology. AI applied to medical imaging data identifies subtle signatures correlated with genomics and clinical outcomes [57,58].
Predictive biomarkers and multiomics integration
Machine learning tools integrating complex molecular and clinical variables have defined prognostic signatures guiding precision treatment strategies. AI modeling of tumor genomic and transcriptomic profiles predicts response to targeted therapies and immunotherapies. Hybrid AI techniques show promise for reconciling multi-dimensional omics datasets to uncover biologically relevant patterns in breast cancer [59-62].
AI-based clinical decision support systems
AI-powered clinical decision support systems integrate patient health records with real-time data analytics to assist physicians with personalized treatment planning. Pilot studies indicate AI recommendation systems can promote evidence-based breast cancer therapy selection and comorbidity management. AI-based methods may enhance risk modeling for adequate patient screening and enable optimized medication dosing [63,64].
Traditional Chinese Medicine
Traditional Chinese medicine (TCM) encompasses a range of long-used therapeutic practices including herbal remedies, acupuncture and dietary measures that have shown promise as adjuvant treatments for breast cancer as depicted in Table 2. Commonly used Chinese herbs and formulations demonstrate antitumor effects in preclinical models. Ongoing studies are exploring the integration of TCM with conventional chemotherapy regimens.
|
TCM Type |
Herb/Formula Examples |
Preclinical Anti-Cancer Effects |
Clinical Trial Stage with Chemotherapy |
|
Single Herbs |
Astragalus, Tinospora cordifolia, Scutellaria barbata |
Cytotoxic, pro-apoptotic, anti-angiogenic, immunomodulatory |
- |
|
Composite Formulas |
Ren Shen Yang Rong Tang, Yindan Jiedu granules, Huachansu injection |
Inhibit tumor growth, modulate signaling pathways |
Early phase trials of Huachansu plus chemotherapeutics: improved response, survival |
|
Acupuncture |
- |
Anti-inflammatory effects in breast cancer models |
Phase II trials show reduced side effects of aromatase inhibitors |
|
Dietary Therapy |
Lingzhi mushroom |
Stimulate immune cytokines, induce cancer cell apoptosis |
- |
Commonly used herbs and formulations
Chinese medicinal herbs and mixtures with historical applications against malignancies are being investigated for breast cancer management. Notable single herbs include Astragalus mongholicus, Tinospora cordifolia, and Scutellaria barbata. Composite formulations such as Ren Shen Yang Rong Tang, Yindan Jiedu granules and Huachansu injection also exhibit anticancer activities in breast cancer models [65,66].
Anti-cancer effects and mechanisms of action
Laboratory studies indicate commonly used Chinese medicinal plants mediate cytotoxic, pro-apoptotic, anti-proliferative, anti-angiogenic, anti-inflammatory, and immunostimulatory responses in breast cancer cells and animal models. Extracts and isolated compounds modulate gene expression and signaling pathways associated with tumor progression and treatment sensitivity. Findings support both direct antitumor effects and immunomodulatory mechanisms [67,68].
Clinical trials of herbs with chemotherapy
Early phase trials demonstrate safety, positive impacts on quality of life, and potential anticancer activity of herbal additions to conventional chemotherapy. For example, Huachansu injection combined with chemotherapeutics was well-tolerated while appearing to improve tumor response and survival outcomes compared to chemotherapy alone. Larger randomized trials are still needed to conclusively evaluate combinations of Chinese herbal components with standard breast cancer regimens [69,70].
Future Directions and Limitations
Despite recent advances, significant challenges remain in breast cancer management warranting further research. More definitive phase III trials with larger patient cohorts are required to validate preliminary findings and conclusively determine clinical efficacy of emerging approaches like microbiome, nanotechnology, and AI-based interventions. Their real-world feasibility and health economics should also be established.
Predictive biomarker development and advanced bioinformatics analytics will be pivotal to realize precision oncology applications of multi-omics profiling. Integrative analysis of heterogeneous datasets remains difficult, and predictive signatures still require prospective clinical confirmation. Novel machine learning techniques show promise to distill meaningful biological patterns from noisy high-dimensional omics and imaging data [71,73].
Cancer antigen vaccines and immunotherapies face obstacles such as suboptimal immunogenicity, immunosuppressive mechanisms limiting efficacy, and risk of autoimmunity which should be key areas of focus. Both preclinical models and clinical trials must better recapitulate native immune microenvironments to enable translational success [74,75]. More research into microbiota dysbiosis and diet in breast cancer can help guide tailored therapeutic manipulation of the gut ecosystem [76]. Effects of traditional herbs require elucidation of bioactive compounds and mechanisms of action to allow pharmacologic optimization. Legal and regulatory hurdles also constrain the integration of complementary medicine despite early signals of benefit [77].
Conclusions
While substantial progress has been made in breast cancer management with conventional therapeutic approaches, significant unmet needs persist including disease relapse, adverse effects, and mortality among high-risk subgroups. Emerging modalities such as omics-based biomarkers, cancer vaccines, microbiome/nutrition manipulation, nanotherapeutics, artificial intelligence, and traditional medicines have shown tremendous translational potential to address existing challenges through data-driven precision diagnosis/prognosis and molecularly targeted, immunomodulatory, personalized interventions with favorable safety profiles. Thoughtful integration of these innovative technologies with current breast cancer care strategies can facilitate incremental improvements in risk stratification, early detection, therapeutic selection for precision oncology, monitoring of treatment response, as well as discovery of fresh targets and agents as summarized in Table 3.
|
Emerging Modality |
Key Components |
Mechanism of Action |
Current Development Stage |
|
Cancer Antigen Vaccines |
Tumor-associated antigens (TAAs), Neoantigens |
Stimulate immune response against antigens on tumor cells |
Preclinical animal models show therapeutic potential, early phase clinical trials demonstrate safety and immunogenicity |
|
DNA Vaccines |
Engineered DNA injected to produce tumor antigens in vivo |
Antigen production activates anti-tumor immunity |
Preclinical efficacy established; phase I/II human trials ongoing |
|
Vector-based Vaccines |
Viruses engineered to express TAAs |
Robust cytotoxic T cell response due to natural viral adjuvanticity |
Protective capacity and safety evidenced in animal models, phase I/II evaluation of patient immunogenicity and clinical impact |
|
Omics-Guided Therapies |
Genomics, epigenomics, proteomics, metabolomics |
Molecular profiling guides therapy selection and reveals targets |
Commercially available tests match patients with targeted drugs based on genomic/proteomic signatures |
|
Microbiome Manipulation |
Probiotics, prebiotics, bacteriotherapy, fecal transplant |
Restore anti-inflammatory commensal bacteria depleted in cancer |
Preclinical studies demonstrate anti-tumor immune modulation, synergy with immunotherapy, early-phase patient trials |
|
Nanotechnology Platforms |
Smart nanoparticles, combination drug nanoformulations, theranostics |
Selective tumor targeting, controlled drug release, integration of therapeutic & diagnostic features |
Preclinical evidence of enhanced pharmacokinetics, biodistribution and antitumor efficacy, some nanoparticle formulations FDA-approved |
|
Artificial Intelligence |
Medical imaging analysis, multi-omics integration, clinical decision support |
Advanced machine learning analytics extract clinically meaningful patterns |
Pilot studies indicate improved diagnostic accuracy, prognostic predictions, and personalized treatment recommendations |
Recommendations
Moving forward, it is recommended that the most promising emerging breast cancer technologies proceed to definitive clinical evaluation through larger, randomized controlled trials to validate preliminary findings, conclusively establish clinical added value over standard management, and assess real-world feasibility. Development of predictive biomarkers and advanced analytics will be key to unlock the potential of high-dimensional “big data” approaches. Regulatory infrastructure and scientific consensus should be proactively developed to enable responsible translation of new modalities. Importantly, next-generation solutions must remain human-centric regarding accessibility, financial burden, and quality of life. Keeping patients at the center of future innovations will ensure breast cancer care continues to advance towards more precise, effective, and compassionate practice.
List of Abbreviations
TAA: Tumor-Associated Antigen; HER2: Human Epidermal Growth Factor Receptor 2; miRNA: MicroRNA; MRI: Magnetic Resonance Imaging; FDA: Food and Drug Administration; AI: Artificial Intelligence; TCM: Traditional Chinese Medicine
Declarations
Ethics approval and consent to participate
Not Applicable.
Consent for publication
Not Applicable.
Availability of data and materials
All data is available, and sharing is available as well as publication.
Competing interests
The authors hereby declare that they have no competing interests.
Funding
The corresponding author supplied all study materials. There was no further funding for this study.
Authors' contributions
The authors completed the study protocol and were the primary organizers of data collection and the manuscript's draft and revision process.
Acknowledgements
The authors thank all the researchers who have made great efforts in their studies. The authors would like to thank the Deanships of all the participating Universities for supporting this work. Moreover, we are grateful to the editors, reviewers, and readers of this journal.
References
2. Contiero P, Boffi R, Borgini A, Fabiano S, Tittarelli A, Mian M, et al. Causes of death in women with breast cancer: a risks and rates study on a population-based cohort. Frontiers in Oncology. 2023;13:1270877.
3. Wang J, Wu SG. Breast cancer: an overview of current therapeutic strategies, challenge, and perspectives. Breast Cancer: Targets and Therapy. 2023 Dec 31:721-30.
4. Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, et al. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Molecular Cancer. 2023 Jul 6;22(1):105.
5. Addissouky TA, El Agroudy AE, Khalil AA. Developing a novel non-invasive serum-based diagnostic test for early detection of colorectal cancer. American Journal of Clinical Pathology. 2023 Nov 1;160(Supplement_1):S17.
6. Addissouky TA, Khalil AA. Detecting lung cancer stages earlier by appropriate markers rather than biopsy and other techniques. American Journal of Clinical Pathology. 2020 Oct;154(Supplement_1):S146-7.
7. Xie N, Shen G, Gao W, Huang Z, Huang C, Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduction and Targeted Therapy. 2023 Jan 6;8(1):9.
8. Biswas N, Chakrabarti S, Padul V, Ashili S. Designing neoantigen cancer vaccines, trials, and outcomes. Frontiers in Immunology. 2023 Feb 9;14:1105420.
9. Addissouky TA, El Sayed IE, Ali MM, Wang Y, El Baz A, Khalil AA, et al. Can vaccines stop cancer before it starts? Assessing the promise of prophylactic immunization against high-risk preneoplastic lesions. Journal of Cellular Immunology. 2023 Nov 29;5(4):127-40.
10. Addissouky TA, El Sayed IE, Ali MM, Wang Y, El Baz A, Elarabany N, et al. Oxidative stress and inflammation: elucidating mechanisms of smoking-attributable pathology for therapeutic targeting. Bulletin of the National Research Centre. 2024 Jan 22;48(1):16.
11. Addissouky TA, El Sayed IE, Ali MM, Wang Y, El Baz A, Khalil AA, et al. Molecular Pathways in Sepsis Pathogenesis: Recent Advances and Therapeutic Avenues. Journal of Cellular Immunology. 2024 Jan 20;5(6):174-83.
12. Jiang XT, Liu Q. mRNA vaccination in breast cancer: current progress and future direction. Journal of Cancer Research and Clinical Oncology. 2023 Sep;149(11):9435-50.
13. Ho SY, Chang CM, Liao HN, Chou WH, Guo CL, Yen Y, et al. Current trends in neoantigen-based cancer vaccines. Pharmaceuticals. 2023 Mar 5;16(3):392.
14. Wawruszak A, Okoń E, Dudziak K. Advancements in Zebrafish Models for Breast Cancer Research: Unveiling Biomarkers, Targeted Therapies, and Personalized Medicine. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 2023;29:e940550-1.
15. Addissouky TA, Ali MM, El Sayed IE, Wang Y. Recent advances in diagnosing and treating helicobacter pylori through botanical extracts and advanced technologies. Archives of Pharmacology and Therapeutics. 2023 Nov 3;5(1):53-66.
16. Addissouky TA, Megahed FA, Elagroudy AE, El Sayed IE. Efficiency of mixture of olives oil and figs as an antiviral agent: a review and perspective. International Journal of Medical Science and Health Research. 2020 Aug;4(4):107-11.
17. Addissouky TA, Khalil AA, El Agroudy AE. Assessing the efficacy of a modified triple drug regimen supplemented with mastic gum in the eradication of Helicobacter Pylori Infection. American Journal of Clinical Pathology. 2023;160(Supplement_1):S19.
18. da Silva JC, Kawassaki AC, Alves FM, Pacholak LM, Ferreira MO, Coletto MI, et al. Animal Models in Cancer Research: Breast Cancer. In: Pathak S, Banerjee A, Bisgin A, (eds). Handbook of Animal Models and its Uses in Cancer Research. Singapore: Springer Nature Singapore; 2023. pp. 247-66.
19. Kolahi Azar H, Gharibshahian M, Rostami M, Mansouri V, Sabouri L, Beheshtizadeh N, et al. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis. Journal of Biological Engineering. 2024 Feb 5;18(1):14.
20. Fan T, Zhang M, Yang J, Zhu Z, Cao W, Dong C. Therapeutic cancer vaccines: advancements, challenges, and prospects. Signal Transduction and Targeted Therapy. 2023 Dec 13;8(1):450.
21. Pandya A, Shah Y, Kothari N, Postwala H, Shah A, Parekh P, et al. The future of cancer immunotherapy: DNA vaccines leading the way. Medical Oncology. 2023 Jun 9;40(7):200.
22. Xie L, Han Y, Liu Y, Zhou Y, Yu J, von Brunn A, et al. Viral vector‐based cancer treatment and current clinical applications. MedComm–Oncology. 2023 Dec;2(4):e55.
23. Ragothaman M, Yoo SY. Engineered phage-based cancer vaccines: current advances and future directions. Vaccines. 2023 Apr 29;11(5):919.
24. Ding YD, Shu LZ, Deng H. Listeria monocytogenes: a promising vector for tumor immunotherapy. Frontiers in Immunology. 2023 Oct 6;14:1278011.
25. Yin G, Liu L, Yu T, Yu L, Feng M, Zhou C, et al. Genomic and transcriptomic analysis of breast cancer identifies novel signatures associated with response to neoadjuvant chemotherapy. Genome Medicine. 2024 Jan 12;16(1):11.
26. Addissouky TA, Wang Y, El Sayed IE, Baz AE, Ali MM, Khalil AA. Recent trends in Helicobacter pylori management: harnessing the power of AI and other advanced approaches. Beni-Suef University Journal of Basic and Applied Sciences. 2023 Sep 2;12(1):80.
27. Miyashita M, Bell JS, Wenric S, Karaesmen E, Rhead B, Kase M, et al. Molecular profiling of a real-world breast cancer cohort with genetically inferred ancestries reveals actionable tumor biology differences between European ancestry and African ancestry patient populations. Breast Cancer Research. 2023 May 25;25(1):58.
28. Prabhu KS, Sadida HQ, Kuttikrishnan S, Junejo K, Bhat AA, Uddin S. Beyond Genetics: Exploring the Role of Epigenetic Alterations in Breast Cancer. Pathology-Research and Practice. 2024 Jan 26:155174.
29. Szczepanek J, Skorupa M, Jarkiewicz-Tretyn J, Cybulski C, Tretyn A. Harnessing epigenetics for breast cancer therapy: The role of DNA methylation, histone modifications, and microRNA. International Journal of Molecular Sciences. 2023 Apr 13;24(8):7235.
30. Brožová K, Hantusch B, Kenner L, Kratochwill K. Spatial proteomics for the molecular characterization of breast cancer. Proteomes. 2023 May 3;11(2):17.
31. El Agroudy AE, Elghareb MS, Addissouky TA, Elshahat EH, Hafez EH. Serum hyaluronic acid as non invasive biomarker to predict liver fibrosis in viral hepatitis patients. Journal of Bioscience and Applied Research. 2016 May 24;2(5):326-33.
32. El Agroudy AE, Elghareb MS, Addissouky TA, Elshahat EH, Hafez EH. Biochemical study of some non invasive markers in liver fibrosis patients. Journal of Bioscience and Applied Research. 2016 May 23;2(5):319-25.
33. Addissouky TA, Khalil AA, El Agroudy AE. Assessment of potential biomarkers for early detection and management of Glomerulonephritis Patients with Diabetic Diseases. American Journal of Clinical Pathology. 2023 Nov;160(Supplement_1):S18-S19.
34. Addissouky T, Ali M, El Sayed IE, Wang Y. Revolutionary innovations in diabetes research: from biomarkers to genomic medicine. Iranian Journal of Diabetes and Obesity. 2023 Dec 28; 15(4):228-42.
35. Garcia-Guasch M, Escrich E, Moral R, Duarte IF. Metabolomics insights into the differential response of breast cancer cells to the phenolic compounds Hydroxytyrosol and Luteolin. Molecules. 2023 May 4;28(9):3886.
36. Chung HW, Park KS, Lim I, Noh WC, Yoo YB, Nam SE, et al. PET/MRI and Novel Targets for Breast Cancer. Biomedicines. 2024 Jan 12;12(1):172.
37. Singh S, Saini H, Sharma A, Gupta S, Huddar VG, Tripathi R. Breast cancer: miRNAs monitoring chemoresistance and systemic therapy. Frontiers in Oncology. 2023 Jun 16;13:1155254.
38. Shrode RL, Knobbe JE, Cady N, Yadav M, Hoang J, Cherwin C, et al. Breast cancer patients from the Midwest region of the United States have reduced levels of short-chain fatty acid-producing gut bacteria. Scientific Reports. 2023 Jan 11;13(1):526.
39. Viswanathan S, Parida S, Lingipilli BT, Krishnan R, Podipireddy DR, Muniraj N. Role of gut microbiota in breast cancer and drug resistance. Pathogens. 2023 Mar 16;12(3):468.
40. Thu MS, Ondee T, Nopsopon T, Farzana IA, Fothergill JL, Hirankarn N, et al. Effect of probiotics in breast cancer: A systematic review and meta-analysis. Biology. 2023 Feb 9;12(2):280.
41. Addissouky TA, Ali MM, El Sayed IE, Wang Y, El Baz A, Elarabany N, et al. Preclinical promise and clinical challenges for innovative therapies targeting liver fibrogenesis. Archives of Gastroenterology Research. 2023 Nov 14;4(1):14-23.
42. Addissouky TA, Wang Y, Megahed FA, El Agroudy AE, El Sayed IE, El-Torgoman AM. Novel biomarkers assist in detection of liver fibrosis in HCV patients. Egyptian Liver Journal. 2021 Dec;11:1-5.
43. Addissouky TA, Ayman E. El-Agroudy, Abdel Moneim AK El-Torgoman and 1Ibrahim E. El-Sayed, Efficacy of biomarkers in detecting fibrosis levels of Liver Diseases. World Journal of Medical Sciences. 2019 Mar;16(1):11-8.
44. Addissouky TA, El Agroudy AE, El-Torgoman AM, El Sayed IE, Ibrahim EM. Efficiency of alternative markers to assess liver fibrosis levels in viral hepatitis B patients. Biomedical Research. 2019 Jan 15;30(2):351-6.
45. Khazaei Y, Basi A, Fernandez ML, Foudazi H, Bagherzadeh R, Shidfar F. The effects of synbiotics supplementation on reducing chemotherapy-induced side effects in women with breast cancer: A randomized placebo-controlled double-blind clinical trial. BMC Complementary Medicine and Therapies. 2023 Sep 26;23(1):339.
46. Stoff R, Wolf Y, Boursi B. Fecal microbiota transplantation as a cancer therapeutic. The Cancer Journal. 2023 Mar 1;29(2):102-8.
47. Bernardo G, Le Noci V, Di Modica M, Montanari E, Triulzi T, Pupa SM, et al. The emerging role of the microbiota in breast cancer progression. Cells. 2023 Jul 27;12(15):1945.
48. Sun L, Liu H, Ye Y, Lei Y, Islam R, Tan S, et al. Smart nanoparticles for cancer therapy. Signal Transduction and Targeted Therapy. 2023 Nov 3;8(1):418.
49. Addissouky T. Detecting liver fibrosis by recent reliable biomarkers in viral hepatitis patients. American Journal of Clinical Pathology. 2019 Oct 1;152:S85.
50. Nag S, Mitra O, Tripathi G, Adur I, Mohanto S, Nama M, et al. Nanomaterials-assisted Photothermal Therapy for Breast Cancer: State-of-the-art Advances and Future Perspectives. Photodiagnosis and Photodynamic Therapy. 2024 Feb:45:103959.
51. Lin X, Wang Y, Fang K, Guo Z, Lin N, Li L. The application of nanoparticles in theranostic systems targeting breast cancer stem cells: current progress and future challenges. Stem Cell Research & Therapy. 2023 Dec 10;14(1):356.
52. Patel P, Kumar K, Jain VK, Popli H, Yadav AK, Jain K. Nanotheranostics for diagnosis and treatment of breast cancer. Current Pharmaceutical Design. 2023 Mar 1;29(10):732-47.
53. Kinnel B, Singh SK, Oprea-Ilies G, Singh R. Targeted therapy and mechanisms of drug resistance in breast cancer. Cancers. 2023 Feb 19;15(4):1320.
54. Abdullah S. The prevention of multi-drug resistance in cancers through the application of nanotechnology-based targeted delivery systems for combination therapies involving Traditional Chinese medicine. Pharmacological Research-Modern Chinese Medicine. 2024 Feb 4:100386.
55. Addissouky TA, Ali M, Sayed IE, Wang Y. Emerging advanced approaches for diagnosis and inhibition of liver fibrogenesis. The Egyptian Journal of Internal Medicine. 2024 Dec;36(1):19.
56. Addissouky TA, Sayed IE, Ali MM, Wang Y, Baz AE, Khalil AA, et al. Latest advances in hepatocellular carcinoma management and prevention through advanced technologies. Egyptian Liver Journal. 2024 Jan 2;14(1):2.
57. Taylor CR, Monga N, Johnson C, Hawley JR, Patel M. Artificial intelligence applications in breast imaging: current status and future directions. Diagnostics. 2023 Jun 13;13(12):2041.
58. You C, Shen Y, Sun S, Zhou J, Li J, Su G, et al. Artificial intelligence in breast imaging: Current situation and clinical challenges. Exploration. 2023 Oct;3(5):20230007.
59. Wang ZZ, Li XH, Wen XL, Wang N, Guo Y, Zhu X, et al. Integration of multi-omics data reveals a novel hybrid breast cancer subtype and its biomarkers. Frontiers in Oncology. 2023 Mar 21;13:1130092.
60. Addissouky TA, El Sayed IE, Ali MM, Wang Y, El Baz A, Elarabany N, et al. Shaping the future of cardiac wellness: exploring revolutionary approaches in disease management and prevention. Journal of Clinical Cardiology. 2024 Jan 5;5(1):6-29.
61. Liu Y, Deng S, Zhang Q, Guo Y, Wang Y, Li T, et al. MLIF modulates microglia polarization in ischemic stroke by targeting eEF1A1. Frontiers in Pharmacology. 2021 Sep 7;12:725268.
62. Orsini A, Diquigiovanni C, Bonora E. Omics technologies improving breast cancer research and diagnostics. International Journal of Molecular Sciences. 2023 Aug 11;24(16):12690.
63. CANDAN H, ŞAHİN H. Breast Cancer Prediction with Artificial Intelligence Based Clinical Decision Support System. Electronic Letters on Science and Engineering. 2023 Jun 6;19(1):29-35.
64. Kočo L, Siebers CC, Schlooz M, Meeuwis C, Oldenburg HS, Prokop M, et al. The Facilitators and Barriers of the Implementation of a Clinical Decision Support System for Breast Cancer Multidisciplinary Team Meetings—An Interview Study. Cancers. 2024 Jan 17;16(2):401.
65. Ali M, Wani SU, Salahuddin M, Manjula SN, Mruthunjaya K, Dey T, et al. Recent advance of herbal medicines in cancer-a molecular approach. Heliyon. 2023 Feb 1;9(2):e13684.
66. Chen PE, Hung HH, Huang WT, Yeh CH, Kao YW, Shia BC, et al. Chinese Herbal Medicine Improved Survival in Stage IV Breast Cancer Patients: Data Mining of the Incorporated Taiwan Cancer Registry Database and Hospital Database. Integrative Cancer Therapies. 2023 Jun;22:15347354231178898.
67. Shabani H, Karami MH, Kolour J, Sayyahi Z, Parvin MA, Soghala S, et al. Anticancer activity of thymoquinone against breast cancer cells: mechanisms of action and delivery approaches. Biomedicine & Pharmacotherapy. 2023 Sep 1;165:114972.
68. Lee SG. Molecular target and action mechanism of Anti-cancer agents. International Journal of Molecular Sciences. 2023 May 4;24(9):8259.
69. Youn BY, Kim JH, Jo YK, Yoon S, Im JY, Kim HJ, et al. Current Characteristics of Herbal Medicine Interventions for Cancer on Clinical Databases: A Cross-Sectional Study. Integrative Cancer Therapies. 2023 Dec;22:15347354231218255.
70. Que S, Ma X, Yang T, He J. Evaluation of the effect of herbal agents as management of radiodermatitis in breast cancer patients: A systematic review and meta‐analysis. Japan Journal of Nursing Science. 2024 Jan;21(1):e12559.
71. Rulten SL, Grose RP, Gatz SA, Jones JL, Cameron AJ. The future of precision oncology. International Journal of Molecular Sciences. 2023 Aug 9;24(16):12613.
72. Rakhshaninejad M, Fathian M, Shirkoohi R, Barzinpour F, Gandomi AH. Refining breast cancer biomarker discovery and drug targeting through an advanced data-driven approach. BMC Bioinformatics. 2024 Jan 22;25(1):33.
73. Restrepo JC, Dueñas D, Corredor Z, Liscano Y. Advances in genomic data and biomarkers: revolutionizing NSCLC diagnosis and treatment. Cancers. 2023 Jul 3;15(13):3474.
74. Kaczmarek M, Poznańska J, Fechner F, Michalska N, Paszkowska S, Napierała A, et al. Cancer vaccine therapeutics: limitations and effectiveness—A literature review. Cells. 2023 Aug 28;12(17):2159.
75. Fan T, Zhang M, Yang J, Zhu Z, Cao W, Dong C. Therapeutic cancer vaccines: advancements, challenges, and prospects. Signal Transduction and Targeted Therapy. 2023 Dec 13;8(1):450.
76. Altinok Dindar D, Chun B, Palma A, Cheney J, Krieger M, Kasschau K, et al. Association between Gut Microbiota and Breast Cancer: Diet as a Potential Modulating Factor. Nutrients. 2023 Oct 31;15(21):4628.
77. German R, Marino N, Hemmerich C, Podicheti R, Rusch DB, Stiemsma LT, et al. Exploring breast tissue microbial composition and the association with breast cancer risk factors. Breast Cancer Research. 2023 Jul 10;25(1):82.
