Naranie Shanmuganathan, Michael Osborn, Timothy P Hughes
{"title":"Which Is the Best Tyrosine Kinase Inhibitor for Newly Diagnosed Chronic Myelogenous Leukemia?","authors":"Naranie Shanmuganathan, Michael Osborn, Timothy P Hughes","doi":"10.1200/EDBK-25-473082","DOIUrl":"https://doi.org/10.1200/EDBK-25-473082","url":null,"abstract":"<p><p>The choice of frontline therapy for a patient with chronic phase chronic myeloid leukemia (CP-CML) can have a profound effect on the long-term clinical outcome. Currently, five tyrosine kinase inhibitors (TKIs-imatinib, dasatinib, nilotinib, bosutinib, and asciminib) are available for frontline therapy, but no single TKI is optimal for all patients. EUTOS long-term survival (ELTS) risk score, comorbidities, and treatment-free remission (TFR) priority are the key determinants of frontline TKI selection. Higher ELTS score, low age and comorbidity score, and a high priority for achievement of TFR would all favor the frontline use of a more potent TKI than imatinib. However, no TKI has improved survival compared with imatinib. In children with CP-CML, imatinib, dasatinib, and nilotinib have similar long-term efficacy, with ease of administration and impact of toxicities on quality of life being key considerations. Recent adult trials of reduced-dose dasatinib frontline showed that efficacy may be equivalent to standard-dose dasatinib with a better tolerability and safety profile, but experience is limited in patients with high-risk ELTS scores. The ASC4FIRST trial has confirmed that tolerability and molecular response with asciminib are superior to those with both imatinib and the second-generation (2G)-TKIs. While the overall treatment failure rate was lower with asciminib, the rate of BCR::ABL1 mutations that emerged with asciminib appeared to be higher. The risk of emergent mutations appears to be highly associated with the presence of <i>ASXL1</i> mutations in the CML cells at diagnosis, but more work is needed to understand the implications of this finding.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473082"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022622","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}
Vivek Subbiah, Megan Othus, Jim Palma, Branko Cuglievan, Razelle Kurzrock
{"title":"Designing Clinical Trials for Patients With Rare Cancers: Connecting the Zebras.","authors":"Vivek Subbiah, Megan Othus, Jim Palma, Branko Cuglievan, Razelle Kurzrock","doi":"10.1200/EDBK-25-100051","DOIUrl":"https://doi.org/10.1200/EDBK-25-100051","url":null,"abstract":"<p><p>The field of rare cancer research is rapidly transforming, marked by significant progress in clinical trials and treatment strategies. Rare cancers, as defined by the National Cancer Institute, occur in fewer than 150 cases per million people each year, yet they collectively represent a significant portion of all cancer diagnoses. Because of their infrequency, these cancers pose distinct challenges for clinical trials, including limited patient populations, geographical dispersion, and a general lack of awareness of treatment options. Economic limitations further complicate drug development, making initiatives such as the Orphan Drug Act essential for incentivizing research. The advent of next-generation sequencing (NGS) and precision medicine has been instrumental in identifying actionable genetic alterations in parallel with an explosion in the development of genomically targeted therapies, immunotherapies, and antibody drug conjugates. Advances in clinical NGS, precision medicine, and tumor-agnostic therapies have become central to the progress in rare cancer research. The development and approval of tumor-agnostic drugs, such as BRAF, NTRK, and RET inhibitors, and immunotherapy for mismatch repair deficient/microsatellite instability-high status cancers highlight the potential of personalized treatments across diverse cancer types and across the age spectrum. Collaborative trials from cooperative groups including SWOG DART, ASCO TAPUR, NCI-MATCH, pediatric COG-match, DRUP, IMPRESS, and innovative registrational basket and platform trials (eg, VE-Basket, ROAR, LIBRETTO-001, ARROW), along with patient advocacy group-run trials like TRACK, are enhancing access to clinical trials. In addition, artificial intelligence has the potential to improve the trial matching process. An integrated approach, combining these innovations in collaboration with multiple stakeholders, is crucial for advancing rare cancer research, offering hope for better patient outcomes and quality of life.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e100051"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144062317","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":"Perioperative Therapy in Oncogene-Driven Non-Small Cell Lung Cancer: Current Strategies and Unanswered Questions.","authors":"Teja Voruganti, Rosalyn Marar, Benjamin Bleiberg, Edoardo Garbo, Biagio Ricciuti, Kaushal Parikh, Charu Aggarwal","doi":"10.1200/EDBK-25-472804","DOIUrl":"https://doi.org/10.1200/EDBK-25-472804","url":null,"abstract":"<p><p>Perioperative therapy has become a critical component in the management of resectable non-small cell lung cancer (NSCLC), particularly in the era of precision medicine. Although molecular testing is standard in metastatic NSCLC, its incorporation into early-stage disease remains essential for guiding treatment decisions. Reflex molecular testing pathways are necessary to optimize tissue utilization and ensure timely results. However, liquid biopsies, although valuable in advanced disease, have limited sensitivity in early-stage NSCLC, reinforcing the need for tissue-based next-generation sequencing. Targeted therapies have revolutionized treatment for oncogene-driven NSCLC, with adjuvant osimertinib now standard for EGFR-mutant disease and ongoing investigations into ALK tyrosine kinase inhibitors (TKIs). However, unanswered questions remain regarding the inclusion of perioperative TKI therapy, the role of molecular residual disease assessment, and whether specific TKIs offer greater benefit for high-risk subgroups. The role of immunotherapy (IO) in oncogene-driven NSCLC remains controversial. Although perioperative chemo-IO has demonstrated survival benefits in unselected NSCLC, its efficacy in EGFR, ALK, and other actionable alterations is unclear. Tumors harboring KRAS and BRAF mutations may respond better because of a more immune-inflamed microenvironment, and remains an active area of investigation. As the landscape of perioperative therapy continues to evolve, ongoing trials will help define the optimal integration of targeted therapies and IO in oncogene-driven NSCLC. Addressing these unanswered questions will be crucial in refining treatment strategies and 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":"e472804"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144053817","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":"Pegylated Interferons: Still a Major Player for the Treatment of Myeloproliferative Neoplasms.","authors":"Michael Daunov, Rebecca B Klisovic","doi":"10.1200/EDBK-25-473912","DOIUrl":"https://doi.org/10.1200/EDBK-25-473912","url":null,"abstract":"<p><p>Over the past 35 years, interferons have been explored in various formulations for the management of Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), such as essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis, and remain a key tool in caring for patients with these diseases. These agents are excellent cytoreductive agents with high rates of hematologic response, are helpful in symptom management, and have a long track record of safety and manageable toxicities. More recently, they have shown promise in sustaining responses over many years, with associated reductions in driver mutations (<i>JAK2, MPL, CALR</i>) of these diseases, particularly in PV and ET. Since reductions in molecular mutant allele burden have been correlated with several response outcomes such as reductions in both thrombotic risk and disease progression, there is emerging proof that interferons may offer disease-modifying activity. These long-term benefits and their use as the preferred agent in young pregnant women who need cytoreduction make interferons often the first choice in young adult population who harbor a lifetime risk of progression. Looking forward, the prospect of sustained treatment-free responses, like chronic myeloid leukemia after deep molecular response, and normal life expectancy may also be on the frontier. Despite relative rookies such as JAK inhibitors in the MPN landscape, the veteran in the game, interferon, remains a key player.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473912"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144000816","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}
Kamaria L Lee, Alexandru Eniu, Christopher M Booth, Molly MacDonald, Fumiko Chino
{"title":"Financial Toxicity and Breast Cancer: Why Does It Matter, Who Is at Risk, and How Do We Intervene?","authors":"Kamaria L Lee, Alexandru Eniu, Christopher M Booth, Molly MacDonald, Fumiko Chino","doi":"10.1200/EDBK-25-473450","DOIUrl":"https://doi.org/10.1200/EDBK-25-473450","url":null,"abstract":"<p><p>Financial toxicity, or the financial burden patients experience because of medical costs, can lead to negative patient effects including lower quality of life, compromised clinical care, and worse health outcomes. People with cancer and survivors are more likely to have financial toxicity than those without cancer, and patients with breast cancer are uniquely at risk. Patients with breast cancer often require multimodal treatment (surgery, radiation, and/or systemic therapy) and adjuvant hormonal therapy can continue for years after primary treatment. With improved disease outcomes, patients with breast cancer have prolonged survivorship often lasting decades but may carry chronic toxicities from treatment; both ongoing treatment of metastatic disease and long-term surveillance include continued tests, imaging, and medical visits that add to the cumulative burden on patients and their families. Additionally, breast cancer predominately affects women, who are more likely to have dual caregiver responsibilities, and increasingly is diagnosed in younger patients, who may have fertility preservation expenses and are more likely to experience education and/or employment disruption. When faced with high costs, patients may face difficult decisions regarding what sacrifices they are willing to endure to receive care. Interventions designed to reduce financial toxicity are moving out of the pilot phase, and ongoing randomized trials are expected to provide evidence into the effectiveness of financial navigation programs. Further work to address financial toxicity in breast cancer at the patient-provider, institutional, and governmental levels is needed for comprehensively better financial outcomes and quality of life.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473450"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143988589","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}
Cheryl M Czerlanis, Navneet Singh, Florian J Fintelmann, Vikram Damaraju, Allison E B Chang, MacKenzie White, Nasser Hanna
{"title":"Broadening the Net: Overcoming Challenges and Embracing Novel Technologies in Lung Cancer Screening.","authors":"Cheryl M Czerlanis, Navneet Singh, Florian J Fintelmann, Vikram Damaraju, Allison E B Chang, MacKenzie White, Nasser Hanna","doi":"10.1200/EDBK-25-473778","DOIUrl":"https://doi.org/10.1200/EDBK-25-473778","url":null,"abstract":"<p><p>Lung cancer is one of the leading causes of cancer-related mortality worldwide, with most cases diagnosed at advanced stages where curative treatment options are limited. Low-dose computed tomography (LDCT) for lung cancer screening (LCS) of individuals selected based on age and smoking history has shown a significant reduction in lung cancer-specific mortality. The number needed to screen to prevent one death from lung cancer is lower than that for breast cancer, cervical cancer, and colorectal cancer. Despite the substantial impact on reducing lung cancer-related mortality and proof that LCS with LDCT is effective, uptake of LCS has been low and LCS eligibility criteria remain imperfect. While LCS programs have historically faced patient recruitment challenges, research suggests that there are novel opportunities to both identify and improve screening for at-risk populations. In this review, we discuss the global obstacles to implementing LCS programs and strategies to overcome barriers in resource-limited settings. We explore successful approaches to promote LCS through robust engagement with community partners. Finally, we examine opportunities to enhance LCS in at-risk populations not captured by current eligibility criteria, including never smokers and individuals with a family history of lung cancer, with a focus on early detection through novel artificial intelligence technologies.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e473778"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144053796","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":"Decoding Clinical Trials in Metastatic Breast Cancer: Practical Insights for Optimal Therapy Sequencing.","authors":"Chiara Corti, Hope S Rugo, Sara M Tolaney","doi":"10.1200/EDBK-25-100053","DOIUrl":"https://doi.org/10.1200/EDBK-25-100053","url":null,"abstract":"<p><p>The art of sequencing therapy in the management of breast cancer is a multifaceted challenge that demands the careful integration of clinical trial data, real-world evidence, and individualized patient factors to guide treatment decisions. As the therapeutic landscape evolves rapidly with new agents and combinations, clinicians are confronted with critical decisions on how best to order treatments to maximize benefit, minimize toxicity, and preserve future options. For patients with estrogen receptor-positive (ER+) disease, this review discusses how emerging resistance patterns after cyclin-dependent kinase 4 and 6 inhibitors require careful re-evaluation of subsequent endocrine and targeted therapies, as well as chemotherapy, emphasizing the need for evidence-based strategies and ethical crossover designs in clinical trials. In addition, for both ER+ and ER- metastatic breast cancer (MBC) with nonoverexpressed human epidermal growth factor receptor 2 (HER2), this review highlights pivotal trials investigating antibody-drug conjugates (ADCs)-including trastuzumab deruxtecan, sacituzumab govitecan, and datopotamab deruxtecan-and the challenges related to control arm selection and crossover that may affect outcome interpretation. Finally, for patients with HER2-positive disease, the review explores first-line and maintenance strategies-including insights from landmark trials like CLEOPATRA and PATINA-and addresses the impact of brain metastases on sequencing decisions. By critically appraising current data and identifying gaps in biomarker-guided and sequencing-specific strategies, this review provides practical insights to inform clinical practice and optimize personalized treatment plans for patients with MBC.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e100053"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144079748","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}
Arielle Elkrief, Bertrand Routy, Lisa Derosa, Laura Bolte, Jennifer A Wargo, Jennifer L McQuade, Laurence Zitvogel
{"title":"Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship.","authors":"Arielle Elkrief, Bertrand Routy, Lisa Derosa, Laura Bolte, Jennifer A Wargo, Jennifer L McQuade, Laurence Zitvogel","doi":"10.1200/EDBK-25-472902","DOIUrl":"https://doi.org/10.1200/EDBK-25-472902","url":null,"abstract":"<p><p>The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e472902"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144049886","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}
Erica L Braverman, Giuliana P Mognol, Andy J Minn, Dario A A Vignali, Judith A Varner
{"title":"One Step Ahead: Preventing Tumor Adaptation to Immune Therapy.","authors":"Erica L Braverman, Giuliana P Mognol, Andy J Minn, Dario A A Vignali, Judith A Varner","doi":"10.1200/EDBK-25-481556","DOIUrl":"https://doi.org/10.1200/EDBK-25-481556","url":null,"abstract":"<p><p>Immune checkpoint inhibitors are cancer therapeutics that have shown remarkable success in extending lives in many cancers, including melanoma, MSI-high cancers, and other cancers. However, these therapeutics have not shown benefit for many patients with cancer, especially those with advanced cancer diagnoses. In addition, many patients develop resistance to these therapeutics and/or life-altering adverse events that can include cardiotoxicity, pneumonitis, thyroiditis, pancreatitis, and hepatitis. Extensive efforts to improve cancer care by uncovering mechanisms of resistance to immune therapy in solid tumors have led to identification of new sources of resistance and to the development of new approaches to activate or sustain antitumor immunity. Chronic stimulation of T cells by tumors and by checkpoint inhibitors can lead to a progressive state of T-cell exhaustion. Chronic T-cell activation by the tumor microenvironment (TME) or immune therapeutics can upregulate the expression and function of alternate checkpoints, including the T-cell protein LAG-3. Persistent interferon signaling in the TME can drive epigenetic changes in cancer cells that enable tumors to counter immune activation and disrupt tumor cell elimination. In addition, immune-suppressive macrophages can flood tumors in response to signals from dying tumor cells, further preventing effective immune responses. New clinical developments and/or approvals for therapies that target alternate immune checkpoints, such as the T-cell checkpoint LAG-3; myeloid cell proteins, such as the kinase phosphoinositide 3-kinase gamma isoform; and chronic interferon signaling, such as Jak 1 inhibitors, have been approved for cancer care or shown promise in recent clinical trials.</p>","PeriodicalId":37969,"journal":{"name":"American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting","volume":"45 3","pages":"e481556"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022878","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}