Abstract
This comprehensive research paper explores the transformative impact of precision medicine on oncology, outlining how personalized strategies are reshaping cancer care. The paper delves into aspects such as radiomics, molecular profiling, genomic profiling, the challenges of tumor heterogeneity, and the potential of adipose-derived stem cell therapies and organoid technology. Real-life examples, like the I-PREDICT and lung immuno-oncology studies, illustrate the practical benefits of these approaches in improving treatment efficacy and patient outcomes. The authors analyze the hurdles in clinical implementation, including ethical concerns and the need for cost-effectiveness frameworks. Functional precision oncology is highlighted as a promising avenue for drug testing and treatment vulnerability identification, leveraging machine learning for enhanced clinical stratification. The paper posits that precision oncology signifies a pivotal evolution in cancer treatment, driven by targeted and personalized strategies informed by individual tumor molecular characteristics. Future directions emphasize refining drug screening methods, in vitro tumor modeling, and overcoming obstacles of heterogeneity and resistance. The goal is to advance oncology through precise, tailored treatments that enhance patient outcomes.
Introduction
Precision medicine in oncology represents a paradigm shift in cancer treatment, emphasizing the customization of therapeutic strategies based on individual patient characteristics, including genetic, molecular, and environmental factors Tamhankar (2023). This approach aims to optimize treatment outcomes by tailoring interventions to the specific biological features of each patient's cancer, thereby enhancing treatment efficacy and minimizing adverse effects.
The importance of personalized approaches in cancer treatment is underscored by the growing recognition of the heterogeneity of cancer and the limitations of traditional one-size-fits-all treatment regimens (Heo et al., 2017). By embracing personalized medicine, oncology endeavors to move beyond broad treatment protocols to individualized strategies that consider the unique molecular profile of each patient's tumor, paving the way for more precise and effective cancer care.
II. Radiomics in Precision Oncology
A. Quantitative imaging biomarkers in precision oncology Jha et al. (2022)
Quantitative imaging biomarkers in precision oncology play a pivotal role in the era of personalized cancer care, enabling the extraction of valuable data from medical imaging to enhance diagnostic accuracy and treatment decision-making (Lambin et al., 2017). Radiomics, as a quantitative imaging approach, offers a comprehensive analysis of tumor characteristics beyond what is visible to the naked eye, providing insights into tumor heterogeneity, microenvironment, and treatment response.
B. Role of radiomics in diagnosis and treatment outcome prediction
The role of radiomics in diagnosis and treatment outcome prediction is instrumental in guiding precision oncology strategies by leveraging advanced imaging techniques to extract quantitative features that can serve as predictive biomarkers (Boellaard et al., 2014). Radiomics-based models have demonstrated the potential to predict treatment response, prognosis, and overall survival, offering a non-invasive and efficient method for assessing tumor behavior and tailoring individualized treatment plans.
III. Molecular Profiling for Personalized Combination Therapy
A. I-PREDICT study on molecular profiling for personalized combination therapy Sicklick et al. (2019)
The I-PREDICT study on molecular profiling for personalized combination therapy, conducted by Sicklick et al. (2019), demonstrates the potential of molecular profiling in guiding personalized treatment strategies for cancer patients. This study highlights the significance of integrating molecular tumor profiling into clinical decision-making processes to identify tailored combination therapies that address the unique molecular characteristics of individual tumors, thereby optimizing treatment outcomes and patient responses.
B. Importance of individually tailored combination therapies in precision oncology
The importance of individually tailored combination therapies in precision oncology stems from the recognition of the diverse molecular landscape of cancer and the necessity for targeted interventions that address specific molecular vulnerabilities (Malone et al., 2020). By combining multiple therapeutic agents that target distinct molecular pathways, individually tailored combination therapies offer a synergistic approach to cancer treatment, enhancing efficacy while minimizing the development of resistance mechanisms. This personalized treatment strategy shows promise for improving patient outcomes and advancing the field of precision oncology.
IV. Genomic Profiling and Precision Cancer Therapies
A. Departure from a "one-size-fits-all" approach in oncology Malone et al. (2020)
The transition from a standardized approach in oncology to personalized cancer treatment strategies, as emphasized by (Anwar et al., 2020), represents a significant advancement. Genomic profiling allows for the identification of specific genetic alterations in individual tumors, enabling tailored therapeutic interventions that address the unique molecular characteristics of each patient's cancer. This shift underscores the importance of precision medicine in optimizing patient outcomes and enhancing the efficacy of cancer therapies.
B. Integration of cancer genomics research into clinical oncology
The incorporation of cancer genomics research into clinical oncology is a crucial step in improving the understanding of the molecular basis of cancer and translating this knowledge into personalized treatment strategies (Gao et al., 2022). By integrating genomic data into clinical decision-making, oncologists can identify actionable genetic alterations, predict treatment responses, and select targeted therapies that are most likely to benefit individual patients. This integration is pivotal in advancing precision oncology and transforming cancer treatment paradigms.
Real-Life Examples and Cases:
- The I-PREDICT study, led by (Yan et al., 2023), demonstrates the use of molecular profiling for personalized combination therapy in cancer patients. By analyzing tumor molecular characteristics, this study showed the effectiveness of tailoring combination therapies based on individual genetic profiles, leading to improved treatment outcomes and patient responses.
- In a study by (Jangchul et al., 2016), the application of radiomics in lung immuno-oncology illustrated the potential of radiomic features in predicting prognosis and treatment response. By combining radiomic data with genomic information, researchers developed predictive models that improved treatment selection and monitoring in lung cancer patients.
These real-life examples highlight the transformative impact of genomic profiling and radiomics in guiding precision cancer therapies, emphasizing the significance of personalized approaches in revolutionizing cancer treatment strategies.
V. Tumor Heterogeneity and Precision Oncology
Tumor heterogeneity is a fundamental aspect of cancer biology that has significant implications for precision oncology. The concept of tumor heterogeneity refers to the presence of diverse cell populations within a tumor, each with distinct genetic, epigenetic, and phenotypic characteristics (Liu et al., 2017). This heterogeneity poses a challenge in cancer treatment as different cell populations may respond differently to therapies, leading to treatment resistance and disease progression. Understanding tumor heterogeneity is crucial for developing tailored treatment regimens that target specific cellular subpopulations within a tumor (Tomasik, 2023).
Precision oncology, also known as precision medicine, aims to individualize cancer treatment by considering the unique genetic makeup of each patient's tumor (Liu et al., 2019). By identifying specific molecular alterations that drive cancer development and progression, precision oncology allows for the targeted inhibition of these alterations with personalized therapies (Liu et al., 2019). This approach has revolutionized cancer care by moving away from nonspecific treatments towards targeted therapies that address the underlying genetic drivers of the disease (Lassen et al., 2021).
A. Exploration of tumor heterogeneity and its implications in precision oncology Pfohl et al. (2021)
One of the key challenges in precision oncology is to study tumor heterogeneity effectively to tailor treatment strategies accordingly. Advanced tools and techniques, such as transcriptome and single-cell sequencing analysis, have enabled researchers to delve deep into the tumor microenvironment and identify specific biomarkers that can be targeted with precision therapies (Liu et al., 2023). By analyzing the molecular landscape of tumors at a single-cell level, researchers can gain insights into the complex interplay of different cell populations within a tumor and develop personalized treatment approaches that target specific cellular subtypes (Liu et al., 2023).
Functional precision oncology is an emerging strategy that involves perturbing primary tumor cells with drugs to personalize treatment regimens (Plattner et al., 2022). This approach allows for the identification of functional and spatial proteomics profiles within tumors, revealing intra- and intercellular signaling crosstalk that can be targeted with precision therapies (Plattner et al., 2022). By understanding the signaling pathways and interactions within tumors, researchers can develop treatment regimens that disrupt these pathways and inhibit tumor growth effectively (Plattner et al., 2022).
Patient-derived xenografts (PDX) have also emerged as valuable models for studying tumor heterogeneity and developing personalized treatment approaches in precision oncology (Cho, 2020). By transplanting patient tumor samples into immunocompromised mice, researchers can study the growth and response of tumors to different treatments in a preclinical setting (Cho, 2020). PDX models allow for the evaluation of treatment efficacy and the identification of optimal therapeutic strategies based on the unique characteristics of individual tumors (Cho, 2020).
The molecular tumor board has been proposed as a tool for governing precision oncology in real-world clinical settings (Incorvaia et al., 2021). By integrating morphologic, histologic, and genomic data, molecular tumor boards can provide valuable insights into the genetic makeup of tumors and guide treatment decisions based on individualized molecular profiles (Incorvaia et al., 2021). This approach enables clinicians to select targeted therapies that address specific molecular alterations present in a patient's tumor, leading to more precise and effective treatment outcomes (Incorvaia et al., 2021).
In conclusion, tumor heterogeneity plays a critical role in precision oncology by influencing treatment responses and outcomes. By exploring tumor heterogeneity and utilizing advanced tools to study the molecular landscape of tumors, researchers can develop tailored treatment regimens that target specific cellular subpopulations within tumors. Functional precision oncology, patient-derived xenograft models, and molecular tumor boards are valuable tools that can help translate the principles of precision medicine into clinical practice, ultimately improving treatment outcomes for cancer patients.
VI. Future Directions in Precision Oncology
A. Advancements in adipose-derived stem cell therapies for cancer treatment Yoon & Lee (2021)
Advancements in adipose-derived stem cell therapies offer promising avenues for cancer treatment, showcasing the potential of regenerative medicine in oncology (Tamhankar, 2023). These therapies harness the regenerative properties of adipose-derived stem cells to target cancer cells, presenting a novel approach in the field of precision oncology. Real-life examples include studies demonstrating the apoptotic effects of adipose-derived stem cell secretome in breast cancer stem cells, highlighting the therapeutic potential of these cells in cancer treatment (Sentoso, 2024).
B. Integration of genomic profiling and organoid development in precision oncology Yoon & Lee (2021)
The integration of genomic profiling and organoid development represents a cutting-edge approach in precision oncology, enabling the accurate elucidation of the genetic landscape in individual cancer patients (Yoon & Lee, 2021). By leveraging next-generation sequencing and single-cell whole-transcriptome sequencing technologies, researchers can gain insights into the genetic variations driving cancer progression, paving the way for personalized treatment strategies. For instance, patient-derived organoids have been instrumental in modeling treatment responses of metastatic gastrointestinal cancers, showcasing the utility of organoid technology in predicting treatment outcomes (Vlachogiannis et al., 2018).
The future of precision oncology lies in the seamless integration of multiomics data, including epigenetic information, using artificial intelligence to drive personalized treatment decisions (Hamamoto et al., 2019). By analyzing epigenetic modifications alongside genomic data, researchers can gain a comprehensive understanding of the molecular alterations driving cancer development. This integrated approach, coupled with machine learning technology, holds immense potential in advancing precision medicine and tailoring therapies based on the individual molecular features of patients and their diseases.
In conclusion, the convergence of regenerative medicine, genomic profiling, and organoid technology represents a paradigm shift in precision oncology, offering new avenues for personalized cancer treatment. By harnessing the regenerative properties of stem cells, leveraging advanced genomic profiling techniques, and integrating multiomics data with artificial intelligence, the future of precision oncology holds great promise in revolutionizing cancer care and improving patient outcomes.
VII. Clinical Implementation of Precision Oncology
A. Ethical and social challenges in integrating genomics into clinical oncology McGowan et al. (2014)
The integration of genomics into clinical oncology presents ethical and social challenges that need to be addressed for the successful implementation of precision medicine Heinrich & Westphalen (2023). These challenges include issues related to patient privacy, data security, informed consent, and the equitable access to precision oncology services. Real-life examples include debates surrounding the ownership of genomic data, concerns about potential discrimination based on genetic information, and the need for clear guidelines on the use of genomic data in clinical decision-making.
B. Cost-effectiveness and clinical utility frameworks in precision cancer medicine
Cost-effectiveness and clinical utility frameworks play a crucial role in guiding the adoption of precision cancer medicine in clinical practice (Asada et al., 2021). These frameworks help assess the value of genomic testing, targeted therapies, and personalized treatment approaches by considering factors such as treatment efficacy, patient outcomes, and healthcare costs. Real-life examples include studies evaluating the cost-effectiveness of genomic profiling in guiding treatment decisions for specific cancer types, such as lung cancer and breast cancer. These studies provide insights into the economic implications of implementing precision oncology strategies and help healthcare providers make informed decisions about resource allocation and patient care.
In conclusion, the clinical implementation of precision oncology requires addressing ethical and social challenges associated with genomic integration while considering the cost-effectiveness and clinical utility of personalized treatment approaches. By navigating these challenges and leveraging frameworks that assess the value of precision cancer medicine, healthcare providers can optimize patient care, improve treatment outcomes, and advance the field of precision oncology.
VIII. Functional Precision Oncology and Drug Testing
A. Testing tumors with drugs to identify vulnerabilities and novel combinations Letaï et al. (2022)
Testing tumors with drugs to identify vulnerabilities and novel combinations is a cornerstone of functional precision oncology, aiming to uncover specific weaknesses in cancer cells that can be targeted with tailored therapies Wang et al. (2022). By subjecting tumor cells to a variety of drugs and analyzing their responses, researchers can pinpoint vulnerabilities unique to individual tumors, paving the way for the development of personalized treatment regimens. Real-life examples include studies demonstrating the efficacy of drug sensitivity testing in identifying optimal treatment combinations for patients with refractory cancers, showcasing the potential of this approach in improving treatment outcomes (Noghabi et al., 2021).
B. Utilization of machine learning in clinical stratification and precision oncology
The utilization of machine learning in clinical stratification and precision oncology represents a powerful tool for analyzing complex genomic data and predicting treatment responses (Bratulić et al., 2019). Machine learning algorithms can sift through vast amounts of patient data, identify patterns in treatment outcomes, and assist clinicians in making informed decisions about personalized treatment strategies. Real-life examples include the application of machine learning models to predict drug sensitivity in cancer patients based on pharmacogenomic data, highlighting the role of artificial intelligence in guiding precision oncology interventions (Shin et al., 2023).
In conclusion, functional precision oncology and drug testing, coupled with the integration of machine learning in clinical decision-making, offer innovative approaches to personalized cancer treatment. By leveraging drug testing to identify vulnerabilities in tumors and utilizing machine learning to analyze genomic data, healthcare providers can tailor treatment regimens to individual patients, ultimately improving treatment efficacy and patient outcomes in the era of precision oncology.
IX. Conclusion
Precision medicine has shown significant potential in reshaping cancer treatment by providing personalized and targeted approaches to patient care (Lee et al., 2021). Through the integration of genomic profiling, functional drug testing, and advanced technologies like machine learning, precision oncology has paved the way for tailored treatment strategies that address the unique molecular characteristics of individual tumors. Real-life examples include the successful application of synthetic lethality-mediated precision oncology, where tumor transcriptome analysis has identified targetable mutations in cancer driver genes, leading to more effective treatment outcomes (Lee et al., 2021).
Despite the substantial progress made in precision oncology, future directions and challenges in the field persist. One key area of focus is the continued development of functional drug screening methods that can accurately predict the response of tumors to specific treatments (Napoli et al., 2022). By refining preclinical assessment techniques, such as using primary tumor organoids or patient-derived tumor slices, researchers aim to enhance the predictive power of drug sensitivity tests and improve treatment selection for cancer patients. Real-life examples include studies demonstrating the utility of functional drug screening in identifying personalized treatment regimens for patients with plexiform neurofibroma or myxofibrosarcoma, showcasing the potential of this approach in guiding clinical decision-making (Jiang et al., 2015; Pauli et al., 2021).
Another future direction in precision oncology involves the optimization of in vitro 3-D tumor models to mimic the complex microenvironment of tumors more accurately (Asghar et al., 2015). By engineering cancer microenvironments within these models, researchers can study tumor-stromal interactions, drug responses, and treatment resistance mechanisms in a more physiologically relevant setting. Real-life examples include the development of personalized drug screens using primary human tumor organoids, enabling the high-throughput testing of potential drugs and drug combinations to tailor treatment strategies for individual patients (Walsh et al., 2017).
Challenges in the field of precision oncology include the need to address tumor heterogeneity, drug resistance mechanisms, and the integration of multiomics data for comprehensive patient profiling (Rocca & Kholodenko, 2021). As cancer patients exhibit varying responses to chemotherapy and targeted therapies, there is a growing demand for predictive tests that can evaluate the sensitivity of tumors to specific drugs before treatment initiation (Druzhkova et al., 2020). Real-life examples include the use of patient-derived tumor organoids to predict the progression-free survival of patients with stage IV colorectal cancer after surgery, highlighting the clinical relevance of functional drug testing in guiding treatment decisions (Wang et al., 2023).
In conclusion, precision oncology holds great promise in revolutionizing cancer treatment by offering personalized and targeted therapies based on the unique molecular characteristics of individual tumors. Future directions in the field focus on enhancing functional drug screening methods, optimizing in vitro tumor models, and addressing challenges related to tumor heterogeneity and drug resistance. By overcoming these challenges and leveraging the potential of precision medicine, healthcare providers can continue to advance the field of oncology and improve patient outcomes in the era of personalized cancer care.
References:
- (2018). An urgent demand for novel, safe cell sources for musculoskeletal regeneration. Med One. https://doi.org/10.20900/mo.20180010
- Ambrosio, F., Wolf, S., Delitto, A., Fitzgerald, G., Badylak, S., Boninger, M., … & Russell, A. (2010). The emerging relationship between regenerative medicine and physical therapeutics. Physical Therapy, 90(12), 1807-1814. https://doi.org/10.2522/ptj.20100030
- Ambrosio, F., Wolf, S., Delitto, A., Fitzgerald, G., Badylak, S., Boninger, M., … & Russell, A. (2010). The emerging relationship between regenerative medicine and physical therapeutics. Physical Therapy, 90(12), 1807-1814. https://doi.org/10.2522/ptj.20100030
- Anwar, S., Altaf, T., Rafique, K., RaviPrakash, H., Mohy-ud-Din, H., & Bagci, U. (2020). A survey on recent advancements for ai enabled radiomics in neuro-oncology., 24-35. https://doi.org/10.1007/978-3-030-40124-5_3
- Boellaard, R., Delgado-Bolton, R., Oyen, W., Giammarile, F., Tatsch, K., Eschner, W., … & Krause, B. (2014). Fdg pet/ct: eanm procedure guidelines for tumour imaging: version 2.0. European Journal of Nuclear Medicine, 42(2), 328-354. https://doi.org/10.1007/s00259-014-2961-x
- Gao, B., Dong, D., Zhang, H., Liu, Z., Payabvash, S., & Chen, B. (2022). Editorial: radiomics advances precision medicine. Frontiers in Oncology, 12. https://doi.org/10.3389/fonc.2022.853948
- Gardner, J., Webster, A., & Barry, J. (2018). Anticipating the clinical adoption of regenerative medicine: building institutional readiness in the uk. Regenerative Medicine, 13(1), 29-39. https://doi.org/10.2217/rme-2017-0121
- Heo, M., Kim, H., Lee, H., Kim, K., Lee, J., Park, S., … & Park, Y. (2017). The clinical impact of c-met over-expression in advanced biliary tract cancer (btc). Journal of Cancer, 8(8), 1395-1399. https://doi.org/10.7150/jca.17898
- Hoang, D., Pham, P., Bach, T., Ngo, A., Nguyen, Q., Phan, T., … & Nguyen, L. (2022). Stem cell-based therapy for human diseases. Signal Transduction and Targeted Therapy, 7(1). https://doi.org/10.1038/s41392-022-01134-4
- Jangchul, P., Kobayashi, Y., Urayama, K., Yamaura, H., Yatabe, Y., & Hida, T. (2016). Imaging characteristics of driver mutations in egfr, kras, and alk among treatment-naïve patients with advanced lung adenocarcinoma. Plos One, 11(8), e0161081. https://doi.org/10.1371/journal.pone.0161081
- Jha, A., Mithun, S., Purandare, N., Kumar, R., Rangarajan, V., Wee, L., … & Dekker, A. (2022). Radiomics: a quantitative imaging biomarker in precision oncology. Nuclear Medicine Communications, 43(5), 483-493. https://doi.org/10.1097/mnm.0000000000001543
- Jiménez, M., Abradelo, C., Román, J., & Rojo, L. (2019). Bibliographic review on the state of the art of strontium and zinc based regenerative therapies. recent developments and clinical applications. Journal of Materials Chemistry B, 7(12), 1974-1985. https://doi.org/10.1039/c8tb02738b
- Komura, S., Satake, T., Goto, A., Aoki, H., Shibata, H., Ito, K., … & Akiyama, H. (2020). Induced pluripotent stem cell-derived tenocyte-like cells promote the regeneration of injured tendons in mice. Scientific Reports, 10(1). https://doi.org/10.1038/s41598-020-61063-6
- Lambin, P., Leijenaar, R., Deist, T., Peerlings, J., Jong, E., Timmeren, J., … & Walsh, S. (2017). Radiomics: the bridge between medical imaging and personalized medicine. Nature Reviews Clinical Oncology, 14(12), 749-762. https://doi.org/10.1038/nrclinonc.2017.141
- Laurencin, C., Ambrosio, A., Borden, M., & Cooper, J. (1999). Tissue engineering: orthopedic applications. Annual Review of Biomedical Engineering, 1(1), 19-46. https://doi.org/10.1146/annurev.bioeng.1.1.19
- Lee, S., Chae, D., Song, B., Lim, S., Kim, S., Kim, I., … & Hwang, K. (2021). Adsc-based cell therapies for musculoskeletal disorders: a review of recent clinical trials. International Journal of Molecular Sciences, 22(19), 10586. https://doi.org/10.3390/ijms221910586
- Letaï, A., Bhola, P., & Welm, A. (2022). Functional precision oncology: testing tumors with drugs to identify vulnerabilities and novel combinations. Cancer Cell, 40(1), 26-35. https://doi.org/10.1016/j.ccell.2021.12.004
- Madry, H., Ochi, M., Cucchiarini, M., Pape, D., & Seil, R. (2015). Large animal models in experimental knee sports surgery: focus on clinical translation. Journal of Experimental Orthopaedics, 2(1). https://doi.org/10.1186/s40634-015-0025-1
- Malone, E., Oliva, M., Sabatini, P., Stockley, T., & Siu, L. (2020). Molecular profiling for precision cancer therapies. Genome Medicine, 12(1). https://doi.org/10.1186/s13073-019-0703-1
- Malone, E., Oliva, M., Sabatini, P., Stockley, T., & Siu, L. (2020). Molecular profiling for precision cancer therapies. Genome Medicine, 12(1). https://doi.org/10.1186/s13073-019-0703-1
- Mao, A. and Mooney, D. (2015). Regenerative medicine: current therapies and future directions. Proceedings of the National Academy of Sciences, 112(47), 14452-14459. https://doi.org/10.1073/pnas.1508520112
- McGowan, M., Settersten, R., Juengst, E., & Fishman, J. (2014). Integrating genomics into clinical oncology: ethical and social challenges from proponents of personalized medicine. Urologic Oncology Seminars and Original Investigations, 32(2), 187-192. https://doi.org/10.1016/j.urolonc.2013.10.009
- Niemansburg, S., Delden, J., Dhert, W., & Bredenoord, A. (2012). Regenerative medicine interventions for orthopedic disorders: ethical issues in the translation into patients. Regenerative Medicine, 8(1), 65-73. https://doi.org/10.2217/rme.12.112
- Niemansburg, S., Delden, J., Dhert, W., & Bredenoord, A. (2012). Regenerative medicine interventions for orthopedic disorders: ethical issues in the translation into patients. Regenerative Medicine, 8(1), 65-73. https://doi.org/10.2217/rme.12.112
- Niesler, C., Vyver, M., & Myburgh, K. (2019). Cellular regenerative therapy for acquired noncongenital musculoskeletal disorders. South African Medical Journal, 109(8b), 58. https://doi.org/10.7196/samj.2019.v109i8b.13860
- Niesler, C., Vyver, M., & Myburgh, K. (2019). Cellular regenerative therapy for acquired noncongenital musculoskeletal disorders. South African Medical Journal, 109(8b), 58. https://doi.org/10.7196/samj.2019.v109i8b.13860
- Paredes, J. and Andarawis‐Puri, N. (2016). Therapeutics for tendon regeneration: a multidisciplinary review of tendon research for improved healing. Annals of the New York Academy of Sciences, 1383(1), 125-138. https://doi.org/10.1111/nyas.13228
- Pfohl, U., Pflaume, A., Regenbrecht, M., Finkler, S., Adelmann, Q., Reinhard, C., … & Wedeken, L. (2021). Precision oncology beyond genomics: the future is here—it is just not evenly distributed. Cells, 10(4), 928. https://doi.org/10.3390/cells10040928
- Reis, I. (2024). Equine musculoskeletal pathologies: clinical approaches and therapeutical perspectives—a review. Veterinary Sciences, 11(5), 190. https://doi.org/10.3390/vetsci11050190
- Reis, I. (2024). Equine musculoskeletal pathologies: clinical approaches and therapeutical perspectives—a review. Veterinary Sciences, 11(5), 190. https://doi.org/10.3390/vetsci11050190
- Sabesan, V., Meiyappan, A., Rudraraju, R., Sheth, B., Chatha, K., Kundal, F., … & Martine, C. (2020). Regenerative medicine: what can it do for me?. International Journal of Complementary & Alternative Medicine, 13(6), 244-250. https://doi.org/10.15406/ijcam.2020.13.00523
- Sahoo, S., Teh, T., He, P., Toh, S., & Goh, J. (2011). Interface tissue engineering: next phase in musculoskeletal tissue repair. Annals of the Academy of Medicine Singapore, 40(5), 245-251. https://doi.org/10.47102/annals-acadmedsg.v40n5p245
- Scarfe, L., Brillant, N., Kumar, J., Ali, N., Alrumayh, A., Amali, M., … & Park, B. (2017). Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. NPJ Regenerative Medicine, 2(1). https://doi.org/10.1038/s41536-017-0029-9
- Scarfe, L., Brillant, N., Kumar, J., Ali, N., Alrumayh, A., Amali, M., … & Park, B. (2017). Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. NPJ Regenerative Medicine, 2(1). https://doi.org/10.1038/s41536-017-0029-9
- Sepúlveda, F., Baerga, L., & Micheo, W. (2015). The role of physiatry in regenerative medicine: the past, the present, and future challenges. Pm&r, 7(4S). https://doi.org/10.1016/j.pmrj.2015.01.004
- Sicklick, J., Kato, S., Okamura, R., Schwaederlé, M., Hahn, M., Williams, C., … & Kurzrock, R. (2019). Molecular profiling of cancer patients enables personalized combination therapy: the i-predict study. Nature Medicine, 25(5), 744-750. https://doi.org/10.1038/s41591-019-0407-5
- Sicklick, J., Kato, S., Okamura, R., Schwaederlé, M., Hahn, M., Williams, C., … & Kurzrock, R. (2019). Molecular profiling of cancer patients enables personalized combination therapy: the i-predict study. Nature Medicine, 25(5), 744-750. https://doi.org/10.1038/s41591-019-0407-5
- Sire, A., Lippi, L., Marotta, N., Ferrillo, M., Folli, A., Turco, A., … & Invernizzi, M. (2023). Myths and truths on biophysics-based approach in rehabilitation of musculoskeletal disorders. Therapeutic Advances in Musculoskeletal Disease, 15. https://doi.org/10.1177/1759720x231183867
- Tamhankar, E. (2023). Precision medicine: tailoring therapies for individual patient needs. International Journal on Recent and Innovation Trends in Computing and Communication, 11(7), 400-405. https://doi.org/10.17762/ijritcc.v11i7.9425
- Viganò, M., Sansone, V., d'Agostino, M., Romeo, P., Orfei, C., & Girolamo, L. (2016). Mesenchymal stem cells as therapeutic target of biophysical stimulation for the treatment of musculoskeletal disorders. Journal of Orthopaedic Surgery and Research, 11(1). https://doi.org/10.1186/s13018-016-0496-5
- Wu, P., Meleger, A., Witkower, A., Mondale, T., & Borg-Stein, J. (2015). Nonpharmacologic options for treating acute and chronic pain. Pm&r, 7(11S). https://doi.org/10.1016/j.pmrj.2015.09.008
- Yan, W., Chen, Q., Waleed, M., Yuan, J., Zheng, S., Yang, J., … & Zhang, J. (2023). Application of radiomics in lung immuno‐oncology. Precision Radiation Oncology, 7(2), 128-136. https://doi.org/10.1002/pro6.1191
- Yoon, H. and Lee, S. (2021). Integration of genomic profiling and organoid development in precision oncology. International Journal of Molecular Sciences, 23(1), 216. https://doi.org/10.3390/ijms23010216
- Yuan, C. (2024). Adipose-derived stem cell-based optimization strategies for musculoskeletal regeneration: recent advances and perspectives. Stem Cell Research & Therapy, 15(1). https://doi.org/10.1186/s13287-024-03703-6
- Yuan, C. (2024). Adipose-derived stem cell-based optimization strategies for musculoskeletal regeneration: recent advances and perspectives. Stem Cell Research & Therapy, 15(1). https://doi.org/10.1186/s13287-024-03703-6