{"title":"PARP inhibitors for prostate cancer: for whom and when?","authors":"Matthew R Cooperberg","doi":"10.1016/s1470-2045(25)00125-1","DOIUrl":"https://doi.org/10.1016/s1470-2045(25)00125-1","url":null,"abstract":"No Abstract","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"47 33 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745367","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":"The Global Platform for Access to Childhood Cancer Medicines: addressing inequities in childhood cancer care","authors":"James R Downing, Tedros Adhanom Ghebreyesus","doi":"10.1016/s1470-2045(25)00146-9","DOIUrl":"https://doi.org/10.1016/s1470-2045(25)00146-9","url":null,"abstract":"No Abstract","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"12 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745368","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}
Emmanuelle Vigarios, Saman Warnakulasuriya, Philippe Pomar, Delphine Maret
{"title":"Integrating dental and oral care in oncology: a crucial step towards comprehensive cancer treatment","authors":"Emmanuelle Vigarios, Saman Warnakulasuriya, Philippe Pomar, Delphine Maret","doi":"10.1016/s1470-2045(25)00098-1","DOIUrl":"https://doi.org/10.1016/s1470-2045(25)00098-1","url":null,"abstract":"No Abstract","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"91 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745374","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}
Pamela Espinoza, Henry E Rice, Paul H Wise, Nickhill Bhakta, Alexandra Mueller, Taisiya Yakimkova, Lisa M Force, Emily R Smith, Asya Agulnik
{"title":"The impact of armed conflict on global patterns of childhood cancer","authors":"Pamela Espinoza, Henry E Rice, Paul H Wise, Nickhill Bhakta, Alexandra Mueller, Taisiya Yakimkova, Lisa M Force, Emily R Smith, Asya Agulnik","doi":"10.1016/s1470-2045(24)00559-x","DOIUrl":"https://doi.org/10.1016/s1470-2045(24)00559-x","url":null,"abstract":"<h2>Section snippets</h2><section><section><section><h2>Incidence of cancer and related mortality in children in conflict zones</h2>Children living in countries with armed conflict account for an increasing proportion of global paediatric cancer deaths over the past 30 years. This pattern has resulted from both a growing number of children living in countries with conflict over time and higher cancer mortality rates in countries in conflict compared with countries not in conflict. During the past three decades, approximately half of the children (aged 0–19 years) around the world lived in countries experiencing armed</section></section></section><section><section><section><h2>Great divergence in global childhood cancer mortality</h2>Many countries have successfully reduced mortality rates over recent decades, leading to aspirational projections of a great convergence in mortality rates for children.<sup>10</sup> However, we found that for children with cancer in conflict settings, there is a great divergence, with higher cancer mortality rates and an increasing proportion of global cancer deaths for children in countries with armed conflict compared with countries without conflict.<sup>11</sup> Without urgent action, this trend suggests that as</section></section></section><section><section><h2>Limitations</h2>There are several limitations to this study. First, our analysis risks stability bias: most sampling data in conflict settings come from the more stable and accessible regions, and the least data from where there is poor access to information, which can lead to falsely lowered measured levels of the impact of conflict on health services.<sup>15</sup> Second, missing data might have biased our findings. In our study, 17·6% of the countries had missing conflict data in 1990, although this represented only</section></section><section><section><h2>Conclusion</h2>Countries experiencing armed conflict account for a large and increasing proportion of all global cancer cases and deaths among children. Continued efforts to reduce paediatric cancer mortality worldwide must confront the complex challenges inherent in providing health services in areas of armed conflict. Reaching the 60% survival goal for all children with cancer globally as set forth by the GICC cannot be achieved without improving childhood cancer care in areas of armed conflict. To meet</section></section><section><section><h2>Declaration of interests</h2>We declare no competing interests.</section></section>","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"16 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745526","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}
Andre P Fay, Karim Fizazi, Nobuaki Matsubara, Arun A Azad, Fred Saad, Ugo De Giorgi, Jae Young Joung, Peter C C Fong, Robert J Jones, Stefanie Zschäbitz, Jan Oldenburg, Neal D Shore, Curtis Dunshee, Joan Carles, Paul Cislo, Jane Chang, Cynthia G Healy, Alexander Niyazov, Neeraj Agarwal
{"title":"First-line talazoparib plus enzalutamide versus placebo plus enzalutamide in men with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: patient-reported outcomes from the randomised, double-blind, placebo-controlled, phase 3 TALAPRO-2 trial","authors":"Andre P Fay, Karim Fizazi, Nobuaki Matsubara, Arun A Azad, Fred Saad, Ugo De Giorgi, Jae Young Joung, Peter C C Fong, Robert J Jones, Stefanie Zschäbitz, Jan Oldenburg, Neal D Shore, Curtis Dunshee, Joan Carles, Paul Cislo, Jane Chang, Cynthia G Healy, Alexander Niyazov, Neeraj Agarwal","doi":"10.1016/s1470-2045(25)00031-2","DOIUrl":"https://doi.org/10.1016/s1470-2045(25)00031-2","url":null,"abstract":"<h3>Background</h3>In the phase 3 TALAPRO-2 trial, talazoparib plus enzalutamide significantly improved radiographic progression-free survival compared with placebo plus enzalutamide in men with metastatic castration-resistant prostate cancer harbouring alterations in genes involved in homologous recombination repair (HRR). We aimed to assess patient-reported outcomes in patients with HRR-deficient metastatic castration-resistant prostate cancer in TALAPRO-2.<h3>Methods</h3>TALAPRO-2 is a randomised, double-blind, placebo-controlled, phase 3 trial conducted at 223 hospitals, cancer centres, and medical centres in 26 countries worldwide. Eligible participants were male patients aged 18 years or older (≥20 years in Japan) who were receiving ongoing androgen deprivation therapy, had asymptomatic or mildly symptomatic metastatic castration-resistant prostate cancer, an Eastern Cooperative Oncology Group performance status of 0 or 1, and had not received previous life-prolonging systemic therapy for castration-resistant prostate cancer or metastatic castration-resistant prostate cancer. Patients with HRR gene alterations were randomly assigned (1:1) using a centralised interactive web response system and a permuted block size of 4 to oral talazoparib 0·5 mg once daily or placebo, plus oral enzalutamide 160 mg once daily, stratified by previous second-generation androgen receptor pathway inhibitor (abiraterone or orteronel) or docetaxel (yes vs no) in the castration-sensitive setting. The sponsor, patients, and investigators were masked to allocation of talazoparib or placebo; enzalutamide was open-label. The primary endpoint was radiographic progression-free survival by blinded independent central review and has been reported previously. Patient-reported outcomes were assessed as secondary outcomes in the patient-reported outcomes population, which comprised patients from the intention-to-treat population with a baseline patient-reported outcome assessment and at least one post-baseline patient-reported outcome assessment. Patient-reported outcomes included time to definitive deterioration in global health status/quality of life (GHS/QoL) per European Organisation for Research and Treatment of Cancer (EORTC) Core Quality of Life Questionnaire (QLQ-C30) and prostate cancer-specific urinary symptoms per EORTC Quality of Life Questionnaire-Prostate (QLQ-PR25), and time to deterioration in pain symptoms per Brief Pain Inventory-Short Form (BPI-SF). Mean change from baseline in GHS/QoL, overall cancer and prostate cancer-specific functioning and symptoms (per EORTC QLQ-C30 and QLQ-PR25), in pain symptoms per BPI-SF, and in general health status per EQ-5D-5L were also patient-reported secondary outcomes. This study is registered with <span><span>ClinicalTrials.gov</span><svg aria-label=\"Opens in new window\" focusable=\"false\" height=\"20\" viewbox=\"0 0 8 8\"><path d=\"M1.12949 2.1072V1H7V6.85795H5.89111V2.90281L0.784057 8L0 7.21635L5.11902 2.1072H1.12949Z\"></pat","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"37 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745527","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}
Ajay Aggarwal, David Cromwell, Julie Nossiter, Jan van der Meulen, Kate Walker
{"title":"The National Cancer Audit Collaborating Centre (NATCAN): improving the quality of National Health Service cancer care in England and Wales","authors":"Ajay Aggarwal, David Cromwell, Julie Nossiter, Jan van der Meulen, Kate Walker","doi":"10.1016/s1470-2045(24)00665-x","DOIUrl":"https://doi.org/10.1016/s1470-2045(24)00665-x","url":null,"abstract":"The National Cancer Audit Collaborating Centre (NATCAN) was launched on Oct 1, 2022, and is delivering ten national cancer audits to assess and assure the quality of National Health Service (NHS) cancer care in England and Wales. These audits are a collaboration between clinical leaders, methodological experts, professional organisations, civil society, and policy makers to develop and implement performance measures across all NHS cancer care and inform quality improvement of the care pathway. The aims of NATCAN are to provide transparent and timely feedback to hospitals about their practices and outcomes of cancer care, identify opportunities for improvement of cancer care, and support hospitals to conduct quality-improvement initiatives. The key methods used to achieve these aims were use of routinely collected data that did not necessitate bespoke collection by health-care staff specifically for the audit; public reporting of outcomes of care at hospital and regional levels; assessment of determinants of variation in cancer care to target quality improvement; and a central governance structure to ensure that unsafe care or outlying performance were managed effectively and promptly. This Personal View provides comparisons between NATCAN and other international audit programmes, including how other countries could consider and incorporate components of NATCAN, alongside scope for future development.","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"41 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745530","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}
Toon Van Gorp, Kathleen N Moore, Gottfried E Konecny, Alexandra Leary, Yolanda García-García, Susana Banerjee, Domenica Lorusso, Jung-Yun Lee, John W Moroney, Giuseppe Caruso, Dagmara Klasa-Mazurkiewicz, Jacqueline Tromp, Lainie P Martin, Shani Breuer, Charles A Leath, David Cibula, S John Weroha, Purificación Estévez-García, David M O’Malley, Rowan E Miller, Felix Hilpert
{"title":"Patient-reported outcomes from the MIRASOL trial evaluating mirvetuximab soravtansine versus chemotherapy in patients with folate receptor α-positive, platinum-resistant ovarian cancer: a randomised, open-label, phase 3 trial","authors":"Toon Van Gorp, Kathleen N Moore, Gottfried E Konecny, Alexandra Leary, Yolanda García-García, Susana Banerjee, Domenica Lorusso, Jung-Yun Lee, John W Moroney, Giuseppe Caruso, Dagmara Klasa-Mazurkiewicz, Jacqueline Tromp, Lainie P Martin, Shani Breuer, Charles A Leath, David Cibula, S John Weroha, Purificación Estévez-García, David M O’Malley, Rowan E Miller, Felix Hilpert","doi":"10.1016/s1470-2045(25)00021-x","DOIUrl":"https://doi.org/10.1016/s1470-2045(25)00021-x","url":null,"abstract":"<h3>Background</h3>Mirvetuximab soravtansine-gynx (MIRV) is a first-in-class antibody–drug conjugate targeting folate receptor α (FRα), approved by the US Food and Drug Administration for the treatment of platinum-resistant ovarian cancer in the USA. Here, we report patient-reported outcomes for participants treated with MIRV compared with investigator's choice of chemotherapy from the phase 3 MIRASOL trial, which met its primary endpoint of progression-free survival and key secondary endpoints of objective response rate and overall survival.<h3>Methods</h3>The MIRASOL trial was a confirmatory, phase 3, randomised, controlled, open-label trial, building on the phase 2 SORAYA trial which had previously demonstrated the safety and efficacy of MIRV in platinum-resistant ovarian cancer. Patients 18 years or older with a confirmed platinum-resistant, recurrent high-grade serous epithelial ovarian cancer diagnosis were recruited from 253 sites including hospitals, academic centres, and community centres in 21 countries. Patients must have received one to three previous systemic anticancer therapies, and have high FRα tumour expression (≥75% tumour cells with an immunohistochemistry score of ≥2+ membrane staining using the PS2+ scoring method), one or more lesions with measurable disease, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) to MIRV or investigator's choice of chemotherapy, stratified by number of previous therapy lines and the type of investigator's choice of chemotherapy. Therapies were administered in an open-label manner; MIRV was administered intravenously at 6 mg/kg of adjusted ideal bodyweight every 3 weeks. The primary endpoint was progression-free survival. Key secondary endpoints were objective response rate, overall survival, and a 15·0-point or greater improvement at week 8 or 9 in abdominal and gastrointestinal symptoms using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Ovarian Cancer Module (EORTC QLQ-OV28) in the intention-to-treat population. The MIRASOL trial was registered at <span><span>ClinicalTrials.gov</span><svg aria-label=\"Opens in new window\" focusable=\"false\" height=\"20\" viewbox=\"0 0 8 8\"><path d=\"M1.12949 2.1072V1H7V6.85795H5.89111V2.90281L0.784057 8L0 7.21635L5.11902 2.1072H1.12949Z\"></path></svg></span> (<span><span>NCT04209855</span><svg aria-label=\"Opens in new window\" focusable=\"false\" height=\"20\" viewbox=\"0 0 8 8\"><path d=\"M1.12949 2.1072V1H7V6.85795H5.89111V2.90281L0.784057 8L0 7.21635L5.11902 2.1072H1.12949Z\"></path></svg></span>), the Gynecologic Oncology Group (GOG 3045), and the European Network of Gynaecological Oncological Trial Groups (ENGOT-ov55), and is complete.<h3>Findings</h3>Between Feb 3, 2020, and Aug 3, 2022, 453 patients were enrolled and randomly assigned to treatment (227 to the MIRV group and 226 to the investigator's choice of chemotherapy group). All patients were female; 301 (66","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"33 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745533","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}
Andrew D J Pearson, Claudia Rossig, Crystal L Mackall, Nirali N Shah, André Baruchel, Sam Daems, John Anderson, Andrea Biondi, Nicholas Bird, Nicole Bodmer, Erica Brivio, Jochen Buechner, Friso G Calkoen, Todd Cooper, Teresa de Rojas, Elizabeth Fox, Rebecca Gardner, Sara Ghorashian, Delphine Heenen, Marianne Ifversen, Gilles Vassal
{"title":"New models for the development of and access to CAR T-cell therapies for children and adolescents with cancer: an ACCELERATE multistakeholder analysis","authors":"Andrew D J Pearson, Claudia Rossig, Crystal L Mackall, Nirali N Shah, André Baruchel, Sam Daems, John Anderson, Andrea Biondi, Nicholas Bird, Nicole Bodmer, Erica Brivio, Jochen Buechner, Friso G Calkoen, Todd Cooper, Teresa de Rojas, Elizabeth Fox, Rebecca Gardner, Sara Ghorashian, Delphine Heenen, Marianne Ifversen, Gilles Vassal","doi":"10.1016/s1470-2045(24)00736-8","DOIUrl":"https://doi.org/10.1016/s1470-2045(24)00736-8","url":null,"abstract":"Realising the potentially substantial benefits of chimeric antigen receptor (CAR) T-cell therapy for children with cancer is hindered by non-scientific barriers that are also relevant for other rare diseases. A solely commercial development model will not deliver optimally due to insufficient return on investment for pharmaceutical companies. Access to therapies is restricted for patients who might benefit and advancing innovation in the academic research setting is difficult. Challenges relating to CAR T-cell therapies in paediatric malignancies and how they might be addressed were discussed in a meeting convened by ACCELERATE—an international multistakeholder organisation aiming to advance the timely investigation of new anticancer drugs. New academic and biopharma hybrid development models could benefit rare populations and coordination of early development can promote synergy and avoid duplicative efforts. Following promising first-in-child trials, new models are needed to support pivotal trials, decentralised manufacturing, registration, and reduced costs. The European Medicines Agency and the US Food and Drug Administration encourage academic development and early discussions. A biotech company funded via a pooled investment vehicle could provide access to safe and effective products for children and adolescents with cancer through registration and reimbursement.","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"5 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745528","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}
Antonin Levy, Christophe Massard, Stefan Michiels, Eric Deutsch
{"title":"Innovative, early-phase clinical trials of drug–radiotherapy combinations","authors":"Antonin Levy, Christophe Massard, Stefan Michiels, Eric Deutsch","doi":"10.1016/s1470-2045(24)00664-8","DOIUrl":"https://doi.org/10.1016/s1470-2045(24)00664-8","url":null,"abstract":"Over the past few decades, breakthroughs in cancer biology at the molecular level have revolutionised cancer treatment. Enhanced precision in radiotherapy has not only reduced patient side-effects, but also enabled the delivery of high-dose stereotactic extracranial irradiation with unprecedented accuracy. Simultaneously, the number of medical therapies available for clinical care continues to grow. Despite the progress made with combined chemoradiotherapy, only a few drug–radiotherapy combinations have received clinical approval, leaving a vast landscape of untapped opportunities for basic, translational, and clinical research, particularly in early-phase drug–radiotherapy trials. New and promising pharmaceutical therapies, paired with advanced radiotherapy technologies, are now being tested in innovative clinical trial designs. Moreover, the integration of biological and imaging markers—both tumour-specific and peripheral—holds the potential to personalise drug–radiotherapy combinations, thereby enhancing the therapeutic index for specific patient populations. In this Review, we highlight the latest developments and future directions for early-phase clinical trials that combine precision drug–radiotherapy strategies in adult patients, with the aims of improving outcomes and expanding treatment options.","PeriodicalId":22865,"journal":{"name":"The Lancet Oncology","volume":"8 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143745529","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}