ImmunotherapyPub Date : 2025-05-06DOI: 10.1080/1750743X.2025.2501929
Ilias Giannakodimos, Afroditi Ziogou, Alexios Giannakodimos, Konstantinos Tzelepis, Zisis Kratiras, Evangelos Fragkiadis, Ioannis Zachos, Vasileios Tzortzis, Michael Chrisofos, Nikolaos Charalampakis
{"title":"Neoadjuvant immunotherapy for muscle-invasive bladder cancer: a 2025 update.","authors":"Ilias Giannakodimos, Afroditi Ziogou, Alexios Giannakodimos, Konstantinos Tzelepis, Zisis Kratiras, Evangelos Fragkiadis, Ioannis Zachos, Vasileios Tzortzis, Michael Chrisofos, Nikolaos Charalampakis","doi":"10.1080/1750743X.2025.2501929","DOIUrl":"https://doi.org/10.1080/1750743X.2025.2501929","url":null,"abstract":"<p><p>Urothelial bladder cancer constitutes one of the most common malignancies of the urinary tract, comprising 90-95% of urothelial carcinomas. Only 25% of patients present with muscle-invasive bladder cancer (MIBC), a neoplasm associated with higher morbidity and mortality. Concerning localized MIBC, cisplatin-based chemotherapy remains the standard neoadjuvant treatment; however, its survival benefits are limited, and its use is restricted to patients with adequate performance status and renal function. Current clinical guidelines recommend neoadjuvant immunotherapy as a first- or second-line option, especially for cisplatin-ineligible patients. Neoadjuvant immunotherapy as monotherapy or in combination with chemotherapy or other immune checkpoint inhibitors is under active investigation. In the ABACUS trial, atezolizumab monotherapy achieved a 31% pathological complete response (pCR). The NCT03520491 trial showed a pCR rate of up to 46% with nivolumab and ipilimumab. The KCT0003804 trial, evaluating immunotherapy plus chemotherapy, reported a 59% pCR and 81.8% 1-year disease-free survival. This review provides an updated overview of clinical trials on neoadjuvant immunotherapy for MIBC, highlighting its therapeutic potential and safety.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"1-9"},"PeriodicalIF":2.7,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144011652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-05-05DOI: 10.1080/1750743X.2025.2500908
Robert A Needleman, Alesha A Thai
{"title":"The complexities of PD-L1 expression as an indicator of immunotherapy outcomes.","authors":"Robert A Needleman, Alesha A Thai","doi":"10.1080/1750743X.2025.2500908","DOIUrl":"https://doi.org/10.1080/1750743X.2025.2500908","url":null,"abstract":"","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"1-4"},"PeriodicalIF":2.7,"publicationDate":"2025-05-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144008878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effect of <i>Helicobacter Pylori</i> infection on immunotherapy for gastrointestinal cancer: a narrative review.","authors":"Afroditi Ziogou, Alexios Giannakodimos, Ilias Giannakodimos, Dimitrios Schizas, Nikolaos Charalampakis","doi":"10.1080/1750743X.2025.2479410","DOIUrl":"10.1080/1750743X.2025.2479410","url":null,"abstract":"<p><p>Immunotherapy for gastrointestinal cancers has elicited considerable amount of attention as a viable therapeutic option for several cancer types. Gut microbiome as a whole plays a critical role in shaping immune responses and influencing cancer progression. Recent evidence suggests that <i>Helicobacter pylori</i> (<i>H. pylori</i>), may influence immunotherapy efficacy by modulating the tumor microenvironment. Infection with <i>H</i>. <i>pylori</i> is common as it affects approximately 50% of the global population and remains the leading risk factor for gastric cancer. Interestingly, recent clinical and preclinical data has associated <i>H. pylori</i> with colorectal cancer carcinogenesis. Gut microbiome appears to be a modulator of the relationship between the immune system, gastrointestinal cancer development and existing therapies. Infection with <i>H. pylori</i> may affect immunotherapy results in both gastroesophageal and colorectal cancer; favorable results were noticed in <i>H. pylori</i> positive patients with gastric cancer, while in colorectal cancer patients the pathogen seemed to impede immunotherapy's action. This article aims to review current data on the role of <i>H. pylori</i> in triggering gastric inflammation and cancer, as well as its potential involvement in colorectal cancer development. Additionally, it seeks to highlight the impact of <i>H. pylori</i> infection on the response to immunotherapy in gastrointestinal cancers.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"355-368"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143630365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-04-02DOI: 10.1080/1750743X.2025.2488609
Bahadır Köylü, Cevat İlteriş Kıkılı, Öner Dikensoy, Fatih Selçukbiricik
{"title":"Late-onset recurrent immune checkpoint inhibitor-related pneumonitis after cessation of pembrolizumab: a case report.","authors":"Bahadır Köylü, Cevat İlteriş Kıkılı, Öner Dikensoy, Fatih Selçukbiricik","doi":"10.1080/1750743X.2025.2488609","DOIUrl":"10.1080/1750743X.2025.2488609","url":null,"abstract":"<p><p>Immune-related adverse events typically occur during the early phases of immune checkpoint inhibitor therapy. However, late-onset immune-related adverse events can still arise long after the immune checkpoint inhibitor therapy has ended. Immune checkpoint inhibitor-related pneumonitis warrants special attention for risk assessment and early detection due to its potential for serious outcomes, including hospitalization and death. Despite its rarity, late-onset immune checkpoint inhibitor-related pneumonitis should be considered in the differential diagnosis for dyspnea in patients with a history of immune checkpoint inhibitor therapy to prevent morbidity and mortality. In this case report, we present a case of an 84-year-old female patient suffering from locally advanced triple-negative breast cancer and late-onset immune checkpoint inhibitor-related pneumonitis requiring hospitalization 104 days after the last cycle of pembrolizumab. Following successful treatment of late-onset immune checkpoint inhibitor-related pneumonitis with corticosteroids, a recurrence of immune checkpoint inhibitor-related pneumonitis occurred a month later. Corticosteroid therapy was reinitiated, gradually tapered after radiological improvement, and eventually discontinued. The patient remains in remission from breast cancer. For patients with a history of immune checkpoint inhibitor therapy, medical vigilance, accurate diagnosis, and timely management of late-onset immune checkpoint inhibitor-related pneumonitis are crucial.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"317-320"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045559/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143763611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Liquid biopsy for guiding breast cancer immunotherapy.","authors":"Emanuela Fina, Elsa Vitale, Simona De Summa, Gennaro Gadaleta-Caldarola, Stefania Tommasi, Raffaella Massafra, Oronzo Brunetti, Alessandro Rizzo","doi":"10.1080/1750743X.2025.2479426","DOIUrl":"10.1080/1750743X.2025.2479426","url":null,"abstract":"<p><p>Liquid biopsy is a laboratory test used to detect and analyze circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and other tumor-derived components, in a blood sample. In the context of breast cancer (BC), liquid biopsies hold significant promise for guiding the use of immune checkpoint inhibitors and immune-based combinations, offering real-time insights into tumor dynamics, treatment response, and resistance mechanisms. This review explores the role of liquid biopsy in BC immunotherapy, focusing on its applications, benefits, issues, and current and future research directions.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"369-383"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045575/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143624368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-04-23DOI: 10.1080/1750743X.2025.2492541
Veera Nurmela, Anni Juntunen, Tuomas Selander, Sanna Pasonen-Seppänen, Outi Kuittinen, Satu Tiainen, Aino Rönkä
{"title":"Poor survival of metastatic cancer patients hospitalized due to immune checkpoint inhibitor-related adverse events.","authors":"Veera Nurmela, Anni Juntunen, Tuomas Selander, Sanna Pasonen-Seppänen, Outi Kuittinen, Satu Tiainen, Aino Rönkä","doi":"10.1080/1750743X.2025.2492541","DOIUrl":"https://doi.org/10.1080/1750743X.2025.2492541","url":null,"abstract":"<p><strong>Aims: </strong>Immune-related adverse events (irAEs) are common side effects of immune checkpoint inhibitor (ICI) cancer therapy, affecting approximately half of ICI-treated patients. irAEs may be severe and result in hospitalization. This study examined the risk factors and outcomes of irAE-related hospitalization.</p><p><strong>Methods: </strong>We conducted a retrospective study including 202 metastatic cancer patients treated with ICIs at Kuopio University Hospital, Finland, in 2015-2022.</p><p><strong>Results: </strong>IrAEs occurred in 57.4% of the patients. About 26.0% of them required inpatient treatment. Hospitalization was associated with severe (grades III - IV) toxicities and need for systemic corticosteroids. Median overall survival (mOS) for hospitalized patients was 12.9 months and for outpatients with irAEs 26.9 months (<i>p</i> = 0.006). The duration of ICI therapy was 1.8 months in hospitalized patients and 5.0 months in outpatients (<i>p</i> < 0.001). The median maximum glucocorticoid doses were 52 mg and 100 mg, respectively (<i>p</i> < 0.001).</p><p><strong>Conclusions: </strong>IrAE-related hospitalization deteriorated the survival of ICI-treated patients, likely due to decreased biological efficacy of ICIs resulting from short therapy periods and strong immunosuppression by glucocorticoids.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":"17 5","pages":"339-346"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045564/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143998287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-04-04DOI: 10.1080/1750743X.2025.2488728
Xiao Hu, Angie Mae Rodday, Anastasia Gurinovich, Stacey Pan, Yana V Salei, Jeffrey H Lin, Margaret M Byrne, Yu Cao, Lori Pai, Susan K Parsons
{"title":"Real-world data of immune-related adverse events in lung cancer patients receiving immune-checkpoint inhibitors.","authors":"Xiao Hu, Angie Mae Rodday, Anastasia Gurinovich, Stacey Pan, Yana V Salei, Jeffrey H Lin, Margaret M Byrne, Yu Cao, Lori Pai, Susan K Parsons","doi":"10.1080/1750743X.2025.2488728","DOIUrl":"10.1080/1750743X.2025.2488728","url":null,"abstract":"<p><strong>Background: </strong>Immune-checkpoint inhibitors (ICIs) have revolutionized lung cancer (LC) treatment; however, immune-related adverse effects (irAEs) may occur. The risk factors of irAEs and the impact of irAEs on patient outcomes in LC remain uncertain.</p><p><strong>Materials and methods: </strong>irAEs within 12 months of ICI initiation in LC patients who initiated ICIs 2018-2021 were identified. Cause-specific Cox regression was used to assess risk factors for irAEs with the competing risk of death; a subset analysis was done among non-small cell lung cancer (NSCLC) group. Multivariable Cox regressions were used to evaluate the impact of irAEs on progression-free survival (PFS) and overall survival (OS).</p><p><strong>Results: </strong>Of 125 patients, 50 irAEs occurred in 39 patients. Small cell lung cancer (SCLC) histology was associated with a higher risk of irAEs (Hazard ratio (HR) = 2.73, 95% CI [1.17, 6.35], <i>p</i> = 0.020) than NSCLC. In NSCLC subset, programmed death-ligand 1 (PDL1) positivity (HR = 2.68, 95% CI [1.10. 6.53], <i>p</i> = 0.030) was identified as a risk factor. irAEs were not significantly associated with PFS (HR = 0.69, <i>p</i> = 0.204) or OS (HR = 0.72, <i>p</i> = 0.353).</p><p><strong>Conclusion: </strong>SCLC histology and PDL1 positivity were associated with irAEs, and the occurrence of irAEs showed no impact on survival in LC patients. Future studies are required to validate the findings.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"321-329"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045565/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143779978","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-04-25DOI: 10.1080/1750743X.2025.2491295
Alexandria Matic, Vera Bril
{"title":"Rozanolixizumab for Myasthenia Gravis: a breakthrough treatment and future prospects.","authors":"Alexandria Matic, Vera Bril","doi":"10.1080/1750743X.2025.2491295","DOIUrl":"https://doi.org/10.1080/1750743X.2025.2491295","url":null,"abstract":"<p><p>Myasthenia gravis is a rare chronic autoimmune disorder affecting the post-synaptic neuromuscular junction, primarily mediated by pathogenic immunoglobulin G (IgG) targeting specific proteins like acetylcholine receptor (AChR), muscle-specific tyrosine kinase (MuSK), and low-density lipoprotein receptor-related protein 4 (LRP4). Modulating pathogenic IgG is a promising approach for disease management. Rozanolixizumab, a human IgG4 neonatal Fc receptor (FcRn) inhibitor, enhances the degradation of pathogenic IgG by 78%, marking a significant advancement in treating generalized myasthenia gravis. It offers effective management for patients with AChR or MuSK antibodies and is administered subcutaneously with mild to moderate adverse events. However, the safety and efficacy of rozanolixizumab require further validation through real-world post-marketing studies. If current trial results are confirmed, rozanolixizumab may become a preferred treatment option for myasthenia gravis in the near future. This review examines clinical trials evaluating the pharmacokinetics, efficacy, and safety of rozanolixizumab in patients with generalized myasthenia gravis and discusses ongoing trials and future research directions.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":"17 5","pages":"309-316"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045563/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143963277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-03-31DOI: 10.1080/1750743X.2025.2485670
Tarek Assi, Tania Moussa, Axel Le Cesne
{"title":"Exploring the role of immune checkpoint inhibitors in solitary fibrous tumors.","authors":"Tarek Assi, Tania Moussa, Axel Le Cesne","doi":"10.1080/1750743X.2025.2485670","DOIUrl":"10.1080/1750743X.2025.2485670","url":null,"abstract":"","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":" ","pages":"305-307"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045572/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143752408","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunotherapyPub Date : 2025-04-01Epub Date: 2025-04-24DOI: 10.1080/1750743X.2025.2493038
Vivek M Shastri, Srideshikan Sargur Madabushi, Susanta K Hui, Jatinder K Lamba
{"title":"CD33-D2 isoform characterization for advancement of its therapeutic potential.","authors":"Vivek M Shastri, Srideshikan Sargur Madabushi, Susanta K Hui, Jatinder K Lamba","doi":"10.1080/1750743X.2025.2493038","DOIUrl":"https://doi.org/10.1080/1750743X.2025.2493038","url":null,"abstract":"<p><strong>Purpose: </strong>While CD33 directed immunotherapies have caught significant interest in recent years, the only approved antibody-drug conjugate targeting this antigen for AML is gemtuzumab ozogamicin, which targets the IgV-domain of CD33. Unfortunately, in its current form, these are not effective in a significant proportion of patients due to the presence of a splicing SNP resulting in the loss of IgV-domain. This, however, can be mitigated by targeting the IgC2-domain of CD33; thus, this study aimed to characterize CD33-D2 isoform using the recently developed CD33-D2-targeting antibody HL2541.</p><p><strong>Methods: </strong>Genetically engineered AML cell lines expressing CD33 isoforms were tested for antibody-bound internalization and response to GO <i>in vitro</i>. AML-bearing NSG-SGM3 mice were used to evaluate CD33-D2 localization and targeting by the HL2541 antibody <i>in vivo</i>.</p><p><strong>Results: </strong>HL2541-bound-CD33-D2 is internalized similar to CD33-FL upon binding the antibody component of GO. Co-existence of both isoforms compromises the internalization by >2.5-3-fold for each isoform in the AML cell lines, further resulting in 7-9.5-fold higher IC<sub>50</sub> values compared to cells expressing only CD33-FL. Finally, we demonstrate that AML cells expressing CD33-D2 localize to bones in mice and are targeted by HL2541antibody <i>in vivo</i>.</p><p><strong>Conclusion: </strong>The results establish the relevance of targeting IgC domain as an alternative immunotarget to supplement AML chemotherapy.</p>","PeriodicalId":13328,"journal":{"name":"Immunotherapy","volume":"17 5","pages":"347-354"},"PeriodicalIF":2.7,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12045573/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143999780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}