{"title":"State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer.","authors":"Wassim Abida, Himisha Beltran, Ruben Raychaudhuri","doi":"10.1200/EDBK-25-473636","DOIUrl":"10.1200/EDBK-25-473636","url":null,"abstract":"<p><p>Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (<sup>177</sup>Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473636"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143671336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Michael Jefford, Larissa Nekhlyudov, Andrea L Smith, Raymond J Chan, Julia Lai-Kwon, Nicolas H Hart
{"title":"Survivorship Care for People Affected by Advanced or Metastatic Cancer: Building on the Recent Multinational Association of Supportive Care in Cancer-ASCO Standards and Practice Recommendations.","authors":"Michael Jefford, Larissa Nekhlyudov, Andrea L Smith, Raymond J Chan, Julia Lai-Kwon, Nicolas H Hart","doi":"10.1200/EDBK-25-471752","DOIUrl":"https://doi.org/10.1200/EDBK-25-471752","url":null,"abstract":"<p><p>Although there is a growing number of people living with advanced or metastatic cancer, primarily because of more effective treatment regimens, there are limited estimates of the actual number of people living with advanced or metastatic cancer. Many people will have treatable but not curable cancers, may have survival measured in years, and may have periods on and off therapy. People with advanced or metastatic disease, as well as their families and caregivers, may experience significant unmet needs, overlapping yet distinct to those with potentially curable cancer. Recently, the Multinational Association of Supportive Care in Cancer and ASCO developed standards and practice recommendations relevant to the delivery of quality survivorship care for people living with advanced or metastatic cancer. The recommendations included seven domains: (1) person-centered care; (2) coordinated and integrated care; (3) evidence-based and comprehensive care; (4) evaluated and communicated care; (5) accessible and equitable care; (6) sustainable and resourced care; and (7) research and data-driven care. Immediate priorities to improve clinical care include focusing on (1) discussions regarding prognosis and goals of care; (2) routinely assessing physical, psychological, and social unmet needs with referral to appropriate supportive care services; and (3) creating blended models of care, incorporating elements of palliative care and survivorship services. Additional areas for focus include (1) advocacy and policy; (2) system design and health care delivery; (3) defining, measuring, and managing quality; (4) addressing inequity; and (5) research specifically focused on these cancer populations.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e471752"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143989916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"New Treatment Approaches for Triple-Negative Breast Cancer.","authors":"Fauzia Riaz, Joshua J Gruber, Melinda L Telli","doi":"10.1200/EDBK-25-481154","DOIUrl":"https://doi.org/10.1200/EDBK-25-481154","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) was first described as a distinct disease entity 20 years ago. Since that time, there has been tremendous effort invested in understanding the clinical features and biology of this breast cancer subtype and developing novel therapeutics specifically targeted for this group of tumors. This review will focus on therapeutic advances in the treatment of metastatic TNBC, outlining successes that contributed to expanded treatment options for advanced TNBC at present and highlight areas of ongoing investigation with potential to further advance treatment paradigms for this aggressive breast cancer subtype. Since 2018, five new therapies have been introduced into clinical practice for the treatment of advanced TNBC. Poly(ADP-ribose) polymerase inhibitors represent the only success for genomically targeted therapy, and this is an option only for a small subgroup of patients with TNBC and a germline <i>BRCA1</i> or <i>BRCA2</i> pathogenic variant. Pembrolizumab is currently the only PD-1 checkpoint inhibitor approved in the United States in combination with chemotherapy in the first-line setting and is an option for roughly 40% of patients with PD-L1 positive tumors. Antibody-drug conjugates have been an important advance in the treatment of advanced TNBC, although these drugs do not represent a TNBC-specific advance as both sacituzumab govitecan and trastuzumab deruxtecan have activity in breast tumors beyond TNBC. Thus, despite significant strides over the past decade, significant unmet clinical need persists, and novel therapeutics remain a pressing need for the treatment of advanced TNBC.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e481154"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144217165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Danielle Brazel, Vera Kazakova, Magdalena Fay, Kathryn Bollin, Kriti Mittal, Kerry L Reynolds, Mazie Tsang
{"title":"Connecting the Dots: Practical Strategies for Academic and Community Oncology Synergy to Advance Multidisciplinary Management in Immunotherapy Toxicity Care.","authors":"Danielle Brazel, Vera Kazakova, Magdalena Fay, Kathryn Bollin, Kriti Mittal, Kerry L Reynolds, Mazie Tsang","doi":"10.1200/EDBK-25-473080","DOIUrl":"10.1200/EDBK-25-473080","url":null,"abstract":"<p><p>Immunotherapy has significantly affected cancer treatment and survival rates, accompanied by an increase in immune-related adverse events (irAEs) requiring new management strategies. irAEs can affect various organ systems and have varying severity levels, with higher rates observed when combining immune checkpoint inhibitors. National organizations such as ASCO, the National Comprehensive Cancer Network, the Society for Immunotherapy of Cancer, and the European Society for Medical Oncology have created guidelines for managing irAEs. This chapter expands on these guidelines by discussing practical strategies to improve the multidisciplinary management in irAE care, focusing on the who, what, and how to bridge gaps in care and enhance collaboration between academic and community oncology practices. Effective irAE management involves early recognition and guideline-adherent approaches using a multidisciplinary team, including oncologists, other subspecialists, primary care clinicians, and all care team members. Institutions are developing methods to integrate irAE care into clinical workflows, such as incorporating urgent care clinics and e-consults for efficient irAE management and developing hub-and-spoke models to extend specialized care from academic centers to community hospitals for equitable care delivery. Additionally, effective patient education is critical for improving irAE recognition and health literacy. The new ASCO Community of Practice called the Alliance for Support and Prevention of Immune-Related Adverse Events consortium and patient advocacy group Standing Together to Optimize Research, Interventions, and Education in irAEs initiatives aim to advance irAE clinical care, research, and education through global collaboration, standardized data collection, and improved outreach to patients and caregivers.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473080"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144133152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuxin Liu, Pinkal Desai, Omar Nadeem, Jennifer A Woyach, Aseel Alsouqi
{"title":"Precursor Hematologic Conditions: Diagnosis, Risk Stratification, Clinical Implications, and Management.","authors":"Yuxin Liu, Pinkal Desai, Omar Nadeem, Jennifer A Woyach, Aseel Alsouqi","doi":"10.1200/EDBK-25-473650","DOIUrl":"https://doi.org/10.1200/EDBK-25-473650","url":null,"abstract":"<p><p>Hematologic precursor conditions include monoclonal gammopathy of undetermined significance (MGUS), monoclonal B-cell lymphocytosis (MBL), and clonal hematopoiesis (CH). These conditions are characterized by a clonal expansion of either plasma cells, lymphoid cells, or myeloid cells without meeting criteria for active hematologic malignancy and are considered premalignant conditions. Diagnosis of these precursor conditions often occur incidentally based on abnormalities on routine laboratory assessments, such as an elevated globulin level leading to diagnosis of MGUS or smoldering multiple myeloma (SMM) or elevated absolute lymphocyte count on blood count checks leading to MBL identification. Diagnosis of CH requires next-generation sequencing of peripheral blood or bone marrow to identify expansion in somatic genetic alterations in hematopoietic cells. Epidemiologic studies of these precursor conditions show that they are relatively common and increase with age, with prevalence of MGUS of 3%-4% in individuals older than 50 years, 3%-17% in adult populations for MBL, and 10% of individuals older than 50 years for CH. Evaluating the risk of progression to overt disease in an individual with a precursor condition is critical in determining management. Often, the risk of progression is quite low, or the latency period is quite long, and for which observation is the current standard of care among these patients. Various risk stratification systems or calculators have been developed for MGUS/SMM, MBL, and CH to better delineate a patient's risk. There is active clinical investigation regarding the role of early intervention among patients who are at highest risk of progression to active hematologic malignancy.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473650"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144276211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Prajakta Adsul, Mackenzi Pergolotti, Kathryn H Schmitz
{"title":"Implementation Science as the Secret Sauce for Integrating Exercise Screening and Triage Pathways in Oncology.","authors":"Prajakta Adsul, Mackenzi Pergolotti, Kathryn H Schmitz","doi":"10.1200/EDBK-25-472854","DOIUrl":"10.1200/EDBK-25-472854","url":null,"abstract":"<p><p>Oncology rehabilitation and exercise (ORE) exist along a continuum of care, providing essential services for patients with cancer to improve health outcomes. Although oncology rehabilitation, typically delivered by licensed medical professionals, is often covered by third-party payers, exercise oncology remains largely unfunded despite its strong evidence base. Research indicates that exercise interventions improve cancer-related fatigue, physical function, mental health, and quality of life, yet referral and implementation remain limited. A geospatial analysis highlights disparity in program availability, particularly in rural and underserved areas. Effective ORE programs require systematic planning, institutional support, clinical workflow integration, and sustainable funding. Case analyses suggest that program adoption depends on leadership buy-in, organizational readiness, and structured referral pathways. Implementation science (IS) provides a framework to address real-world barriers, ensuring efficient integration of exercise services into oncology care. Screening and triage models can aid in patient assessment, streamlining referrals to appropriate levels of exercise intervention. Despite growing policy efforts, widespread third-party reimbursement remains elusive. Leveraging IS strategies can facilitate the adoption and sustainability of ORE programs, bridging the gap between research and practice. Future directions should focus on improving clinician education, expanding access through policy initiatives, and integrating exercise interventions into standard oncology care to optimize patient outcomes.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e472854"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144286705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kim A Reiss, Kevin C Soares, Robert J Torphy, Bishal Gyawali
{"title":"Treatment Innovations in Pancreatic Cancer: Putting Patient Priorities First.","authors":"Kim A Reiss, Kevin C Soares, Robert J Torphy, Bishal Gyawali","doi":"10.1200/EDBK-25-473204","DOIUrl":"10.1200/EDBK-25-473204","url":null,"abstract":"<p><p>Pancreatic adenocarcinoma remains one of the most aggressive and difficult-to-treat solid tumor malignancies, with a high mortality-to-incidence ratio. Globally, pancreatic cancer ranks 12th in terms of incidence but sixth for mortality signifying its aggressive behavior and limited treatment options. While the mortality rates for many other solid tumors have substantially improved over the past few decades, temporal trends in pancreatic cancer mortality rates are quite sobering. In the United States, from 2000 to 2020, the mortality rates from pancreatic cancer have increased, whereas at the same time, mortality rates from other cancers, such as lung, colorectal, or kidney, have fallen appreciably. Is this for lack of treatment innovation? How do we improve survival for patients with pancreatic cancer? In this chapter, we discuss the recent advances and future directions with targeted therapies and immunotherapies in the treatment of pancreatic cancer, and provide the reasons for both optimism and caution for the future of systemic treatment of pancreatic cancer.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473204"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143774422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Heikki Joensuu, Hirotaka Miyashita, Suzanne George, Jason Sicklick
{"title":"Navigating Ongoing Challenges in GI Stromal Tumors.","authors":"Heikki Joensuu, Hirotaka Miyashita, Suzanne George, Jason Sicklick","doi":"10.1200/EDBK-25-473224","DOIUrl":"10.1200/EDBK-25-473224","url":null,"abstract":"<p><p>GI stromal tumors (GISTs) are mesenchymal neoplasms with variable natural histories, originating in the GI tract, most commonly in the stomach. They are frequently characterized by <i>KIT</i> or <i>platelet-derived growth factor receptor alpha</i> (<i>PDGFRA</i>) oncogenic mutations. Surgical resection remains the cornerstone treatment for localized GISTs. Clinical trials have demonstrated the benefits of adjuvant imatinib in patients selected on the basis of recurrence risk and gene mutations, although the optimal duration of therapy is yet to be established. Some data suggest that longer durations of adjuvant imatinib (>3 years) may provide additional benefit, which is being investigated in ongoing trials. Management of imatinib-related adverse effects is essential during treatment, and longitudinal abdominal imaging is mandatory both during and after adjuvant therapy. Once GISTs are more advanced and unresectable, KIT- and PDGFRA-directed tyrosine kinase inhibitors (TKIs) become the key treatment in most patients with <i>KIT</i> mutation. Several TKIs have regulatory approval for advanced GISTs, but in most patients, resistance to TKIs eventually emerges, mainly from secondary resistance mutations in <i>KIT</i>. Each TKI has different coverage of oncogenic <i>KIT</i> mutations, suggested by preclinical and clinical findings, which has given rationale to an ongoing clinical trial that includes molecular selection as eligibility criteria. Furthermore, novel treatment approaches, from TKI combinations to an antibody-drug conjugate, are being investigated. Despite the significant advance in managing GISTs with <i>KIT</i> mutations, those without <i>KIT</i> or <i>PDGFRA</i> mutation, which consists of 10%-15% of patients with GIST, can be a clinical challenge in the advanced setting. These non-<i>KIT/PDGFRA</i> GISTs could be driven by genomic or epigenomic alterations in <i>SDHx</i>, <i>NF1</i> mutations, and other genomic alterations. Non-<i>KIT/PDGFRA</i> GISTs are less responsive to currently available TKIs than GISTs driven by <i>KIT/PDGFRA</i> mutations, and each subset of non-<i>KIT/PDGFRA</i> GIST has distinctive biology and clinical features. Therefore, individualized, multidisciplinary, biology-based management and consideration for clinical trial enrollment are critical for non-<i>KIT/PDGRFA</i> GISTs.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473224"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144112079","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Tali Azenkot, Donna R Rivera, Mark D Stewart, Sandip P Patel
{"title":"Artificial Intelligence and Machine Learning Innovations to Improve Design and Representativeness in Oncology Clinical Trials.","authors":"Tali Azenkot, Donna R Rivera, Mark D Stewart, Sandip P Patel","doi":"10.1200/EDBK-25-473590","DOIUrl":"https://doi.org/10.1200/EDBK-25-473590","url":null,"abstract":"<p><p>The integration of artificial intelligence (AI) and machine learning (ML) in oncology clinical trials is rapidly evolving alongside the broader field. For example, AI-driven adaptive trial designs may allow for real-time modifications based on emerging safety and efficacy signals, enabling more responsive and efficient trials. AI-powered diagnostic tools, including radiomics, computational pathology, and spatial omics, can improve trial patient selection and response assessments. ML-based patient outcome simulations can similarly enhance patient stratification strategies and statistical power. Application of AI can also improve the accessibility of real-world data, including opportunities to enhance data extraction, standardization, and harmonization of data from routine clinical practice. Data generated from digital health technologies (eg, wearable devices, electronic sensors, computing platforms, software applications) may enable a more comprehensive understanding of patient populations to support clinical trials from enrollment to assessment. Automation of trial operations and data management can also improve data fidelity and decrease investigator burden, which has the potential to streamline trial execution and increase potential use of decentralization. There are ongoing efforts to enhance regulatory clarity, mitigate bias, and uphold ethical use of these novel technologies. In this article, we review use cases of AI and ML in oncology clinical trials, including their role in patient recruitment, trial design and operations, data management, and diagnostics. Although these technologies can have applications across all phases of drug development including early discovery, we focus on phase II and III trials, where AI and ML may have a pronounced ability to enhance trial efficiency, patient stratification, and regulatory decision making. By integrating AI and ML, clinical trials can become more adaptive, data-driven, and inclusive in the pursuit of improving patient outcomes.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473590"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144128794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bolin Song, Ipsa Yadav, Jillian C Tsai, Anant Madabhushi, Benjamin H Kann
{"title":"Artificial Intelligence for Head and Neck Squamous Cell Carcinoma: From Diagnosis to Treatment.","authors":"Bolin Song, Ipsa Yadav, Jillian C Tsai, Anant Madabhushi, Benjamin H Kann","doi":"10.1200/EDBK-25-472464","DOIUrl":"https://doi.org/10.1200/EDBK-25-472464","url":null,"abstract":"<p><p>Head and neck squamous cell carcinoma (HNSCC) remains a globally prevalent malignancy with high morbidity and mortality. Despite therapeutic advances, patient outcomes are hindered by tumor heterogeneity, treatment-related toxicity, and the limitations of traditional prognostic tools. Artificial intelligence (AI) offers the opportunity to improve personalized HNSCC management by integrating complex radiologic, pathologic, and molecular data into actionable information insights. This review synthesizes recent developments in AI applications across the HNSCC care continuum, from diagnosis through treatment planning, emphasizing their clinical relevance and translational potential. AI has shown promise in enhancing diagnostic accuracy through automated tumor burden assessment, extranodal extension prediction, and endoscopic image analysis. Deep learning applied to radiology and digital pathology enables the extraction of prognostic features that may inform risk stratification and treatment de-escalation, particularly in human papillomavirus-associated oropharyngeal carcinoma. Multimodal AI models that fuse imaging, histopathology, and electronic health records have demonstrated superior performance in predicting survival outcomes compared with unimodal approaches. Additional applications include early toxicity detection during radiotherapy, adaptive treatment planning, and surgical complication forecasting. AI also holds potential in predicting immunotherapy response by identifying imaging and histologic correlates of tumor immunogenicity. Barriers to clinical translation remain, and continued development of explainable models, prospective trials, and seamless integration into clinical workflows will be critical for broad adoption. AI has already begun to affect HNSCC radiotherapy and surgical planning, and with thoughtful implementation, it may enable safer, more personalized care across the HNSCC treatment landscape.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e472464"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144259083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}