Pub Date : 2025-01-07DOI: 10.1038/d41591-024-00083-5
Christine Laine, Dianne Babski, Vivienne C. Bachelet, Till W Bärnighausen, Christopher Baethge, Kirsten Bibbins-Domingo, Frank Frizelle, Laragh Gollogy, Sabine Kleinert, Elizabeth Loder, João Monteiro, Eric J Rubin, Peush Sahni, Christina C Wee, Jin-Hong Yoo, Lilia Zakhama
Discover the world’s best science and medicine | Nature.com
{"title":"Predatory journals: what can we do to protect their prey?","authors":"Christine Laine, Dianne Babski, Vivienne C. Bachelet, Till W Bärnighausen, Christopher Baethge, Kirsten Bibbins-Domingo, Frank Frizelle, Laragh Gollogy, Sabine Kleinert, Elizabeth Loder, João Monteiro, Eric J Rubin, Peush Sahni, Christina C Wee, Jin-Hong Yoo, Lilia Zakhama","doi":"10.1038/d41591-024-00083-5","DOIUrl":"https://doi.org/10.1038/d41591-024-00083-5","url":null,"abstract":"Discover the world’s best science and medicine | Nature.com","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"28 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142935018","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07DOI: 10.1038/s41591-024-03451-3
Nicholas A. Vitanza, Rebecca Ronsley, Michelle Choe, Kristy Seidel, Wenjun Huang, Stephanie D. Rawlings-Rhea, Madison Beam, Leonel Steinmetzer, Ashley L. Wilson, Christopher Brown, Adam Beebe, Catherine Lindgren, Joshua A. Gustafson, Amy Wein, Susan Holtzclaw, Corrine Hoeppner, Hannah E. Goldstein, Samuel R. Browd, Jason S. Hauptman, Amy Lee, Jeffrey G. Ojemann, Erin E. Crotty, Sarah E. S. Leary, Francisco A. Perez, Jason N. Wright, Marta M. Alonso, Matthew D. Dun, Jessica B. Foster, Diana Hurst, Ada Kong, Alison Thomsen, Rimas J. Orentas, Catherine M. Albert, Navin Pinto, Colleen Annesley, Rebecca A. Gardner, On Ho, Sowmya Pattabhi, Juliane Gust, Jason P. Wendler, Julie R. Park, Michael C. Jensen
Diffuse intrinsic pontine glioma (DIPG) is a fatal central nervous system (CNS) tumor that confers a median survival of 11 months. As B7-H3 is expressed on pediatric CNS tumors, we conducted BrainChild-03, a single-center, dose-escalation phase 1 clinical trial of repetitive intracerebroventricular (ICV) dosing of B7-H3-targeting chimeric antigen receptor T cells (B7-H3 CAR T cells) for children with recurrent or refractory CNS tumors and DIPG. Here we report results from Arm C, restricted to patients with DIPG. The primary objectives were to assess feasibility and tolerability, which were both met. Secondary objectives included assessments of CAR T cell distribution and survival. A total of 23 patients with DIPG enrolled, and 21 were treated with repeated doses of ICV B7-H3 CAR T cells using intra-patient dose-escalation regimens without previous lymphodepletion. Concurrent tumor-directed therapy, including re-irradiation, was not allowed while on protocol therapy. We delivered a total of 253 ICV doses and established the highest planned dose regimen, DR4, which escalated up to 10 × 107 cells per dose, as the maximally tolerated dose regimen. Common adverse events included headache, fatigue and fever. There was one dose-limiting toxicity (intratumoral hemorrhage) during DR2. For all treated patients (n = 21), the median survival from their initial CAR T cell infusion was 10.7 months and the median survival from diagnosis was 19.8 months with 3 patients still alive at 44, 45 and 52 months from diagnosis. Ultimately, this completed first-in-human trial shows that repetitive ICV dosing of B7-H3 CAR T cells in pediatric and young adult patients with DIPG is tolerable, including multiyear repeated dosing, and may have clinical efficacy that warrants further investigation on a multisite phase 2 trial. ClinicalTrials.gov registration: NCT04185038.
{"title":"Intracerebroventricular B7-H3-targeting CAR T cells for diffuse intrinsic pontine glioma: a phase 1 trial","authors":"Nicholas A. Vitanza, Rebecca Ronsley, Michelle Choe, Kristy Seidel, Wenjun Huang, Stephanie D. Rawlings-Rhea, Madison Beam, Leonel Steinmetzer, Ashley L. Wilson, Christopher Brown, Adam Beebe, Catherine Lindgren, Joshua A. Gustafson, Amy Wein, Susan Holtzclaw, Corrine Hoeppner, Hannah E. Goldstein, Samuel R. Browd, Jason S. Hauptman, Amy Lee, Jeffrey G. Ojemann, Erin E. Crotty, Sarah E. S. Leary, Francisco A. Perez, Jason N. Wright, Marta M. Alonso, Matthew D. Dun, Jessica B. Foster, Diana Hurst, Ada Kong, Alison Thomsen, Rimas J. Orentas, Catherine M. Albert, Navin Pinto, Colleen Annesley, Rebecca A. Gardner, On Ho, Sowmya Pattabhi, Juliane Gust, Jason P. Wendler, Julie R. Park, Michael C. Jensen","doi":"10.1038/s41591-024-03451-3","DOIUrl":"https://doi.org/10.1038/s41591-024-03451-3","url":null,"abstract":"<p>Diffuse intrinsic pontine glioma (DIPG) is a fatal central nervous system (CNS) tumor that confers a median survival of 11 months. As B7-H3 is expressed on pediatric CNS tumors, we conducted BrainChild-03, a single-center, dose-escalation phase 1 clinical trial of repetitive intracerebroventricular (ICV) dosing of B7-H3-targeting chimeric antigen receptor T cells (B7-H3 CAR T cells) for children with recurrent or refractory CNS tumors and DIPG. Here we report results from Arm C, restricted to patients with DIPG. The primary objectives were to assess feasibility and tolerability, which were both met. Secondary objectives included assessments of CAR T cell distribution and survival. A total of 23 patients with DIPG enrolled, and 21 were treated with repeated doses of ICV B7-H3 CAR T cells using intra-patient dose-escalation regimens without previous lymphodepletion. Concurrent tumor-directed therapy, including re-irradiation, was not allowed while on protocol therapy. We delivered a total of 253 ICV doses and established the highest planned dose regimen, DR4, which escalated up to 10 × 10<sup>7</sup> cells per dose, as the maximally tolerated dose regimen. Common adverse events included headache, fatigue and fever. There was one dose-limiting toxicity (intratumoral hemorrhage) during DR2. For all treated patients (<i>n</i> = 21), the median survival from their initial CAR T cell infusion was 10.7 months and the median survival from diagnosis was 19.8 months with 3 patients still alive at 44, 45 and 52 months from diagnosis. Ultimately, this completed first-in-human trial shows that repetitive ICV dosing of B7-H3 CAR T cells in pediatric and young adult patients with DIPG is tolerable, including multiyear repeated dosing, and may have clinical efficacy that warrants further investigation on a multisite phase 2 trial. ClinicalTrials.gov registration: NCT04185038.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"78 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934989","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07DOI: 10.1038/s41591-024-03411-x
Reinhard Dummer, Caroline Robert, Richard A. Scolyer, Janis M. Taube, Michael T. Tetzlaff, Alexander M. Menzies, Andrew Hill, Jean-Jacques Grob, David C. Portnoy, Celeste Lebbe, Muhammad A. Khattak, Jonathan Cohen, Gil Bar-Sela, Inderjit Mehmi, Ronnie Shapira-Frommer, Nicolas Meyer, Andrea L. Webber, Yixin Ren, Mizuho Fukunaga-Kalabis, Clemens Krepler, Georgina V. Long
Neoadjuvant immunotherapies have shown antitumor activity in melanoma. Substudy 02C of the global, rolling-arm, phase 1/2, adaptive-design KEYMAKER-U02 trial is evaluating neoadjuvant pembrolizumab (anti-PD-1) alone or in combination, followed by adjuvant pembrolizumab, for stage IIIB–D melanoma. Here we report results from the first three arms: pembrolizumab plus vibostolimab (anti-TIGIT), pembrolizumab plus gebasaxturev (coxsackievirus A21) and pembrolizumab monotherapy. Pathologic complete responses occurred in 10 of 26 patients (38%) with pembrolizumab plus vibostolimab, 7 of 25 (28%) with pembrolizumab plus gebasaxturev and 6 of 15 (40%) with pembrolizumab monotherapy. Major pathologic responses occurred in 13 (50%), 10 (40%) and 7 (47%) patients, respectively. Safety was manageable. Treatment-related adverse events occurred in 24 of 26 patients (92%) with pembrolizumab plus vibostolimab, 21 of 25 (84%) with pembrolizumab plus gebasaxturev and 12 of 15 (80%) with pembrolizumab monotherapy; grade 3 or 4 treatment-related adverse events occurred in 2 (8%), 7 (28%) and 1 (7%) patient in each arm, respectively. No deaths due to adverse events occurred. Exploratory objective responses per RECIST v1.1 were observed in 13 (50%), 8 (32%) and 4 (27%) patients, in each arm, respectively. In a post hoc analysis, scores for tumor mutational burden and an 18-gene T cell-inflamed gene expression profile were generally higher in patients with major pathologic response. Longer follow-up will provide insight into the incremental benefit of combining neoadjuvant pembrolizumab with other therapies in stage IIIB–D melanoma. ClinicalTrials.gov registration: NCT04303169.
{"title":"Neoadjuvant anti-PD-1 alone or in combination with anti-TIGIT or an oncolytic virus in resectable stage IIIB–D melanoma: a phase 1/2 trial","authors":"Reinhard Dummer, Caroline Robert, Richard A. Scolyer, Janis M. Taube, Michael T. Tetzlaff, Alexander M. Menzies, Andrew Hill, Jean-Jacques Grob, David C. Portnoy, Celeste Lebbe, Muhammad A. Khattak, Jonathan Cohen, Gil Bar-Sela, Inderjit Mehmi, Ronnie Shapira-Frommer, Nicolas Meyer, Andrea L. Webber, Yixin Ren, Mizuho Fukunaga-Kalabis, Clemens Krepler, Georgina V. Long","doi":"10.1038/s41591-024-03411-x","DOIUrl":"https://doi.org/10.1038/s41591-024-03411-x","url":null,"abstract":"<p>Neoadjuvant immunotherapies have shown antitumor activity in melanoma. Substudy 02C of the global, rolling-arm, phase 1/2, adaptive-design KEYMAKER-U02 trial is evaluating neoadjuvant pembrolizumab (anti-PD-1) alone or in combination, followed by adjuvant pembrolizumab, for stage IIIB–D melanoma. Here we report results from the first three arms: pembrolizumab plus vibostolimab (anti-TIGIT), pembrolizumab plus gebasaxturev (coxsackievirus A21) and pembrolizumab monotherapy. Pathologic complete responses occurred in 10 of 26 patients (38%) with pembrolizumab plus vibostolimab, 7 of 25 (28%) with pembrolizumab plus gebasaxturev and 6 of 15 (40%) with pembrolizumab monotherapy. Major pathologic responses occurred in 13 (50%), 10 (40%) and 7 (47%) patients, respectively. Safety was manageable. Treatment-related adverse events occurred in 24 of 26 patients (92%) with pembrolizumab plus vibostolimab, 21 of 25 (84%) with pembrolizumab plus gebasaxturev and 12 of 15 (80%) with pembrolizumab monotherapy; grade 3 or 4 treatment-related adverse events occurred in 2 (8%), 7 (28%) and 1 (7%) patient in each arm, respectively. No deaths due to adverse events occurred. Exploratory objective responses per RECIST v1.1 were observed in 13 (50%), 8 (32%) and 4 (27%) patients, in each arm, respectively. In a post hoc analysis, scores for tumor mutational burden and an 18-gene T cell-inflamed gene expression profile were generally higher in patients with major pathologic response. Longer follow-up will provide insight into the incremental benefit of combining neoadjuvant pembrolizumab with other therapies in stage IIIB–D melanoma. ClinicalTrials.gov registration: NCT04303169.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"117 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07DOI: 10.1038/s41591-024-03421-9
Pablo Garcia-Manau, Erika Bonacina, Raquel Martin-Alonso, Lourdes Martin, Ana Palacios, Maria Luisa Sanchez-Camps, Cristina Lesmes, Ivan Hurtado, Esther Perez, Albert Tubau, Patricia Ibañez, Marina Alcoz, Nuria Valiño, Elena Moreno, Carlota Borrero, Esperanza Garcia, Eva Lopez-Quesada, Sonia Diaz, Jose Roman Broullon, Mireia Teixidor, Carolina Chulilla, Roser Ferrer-Costa, Maria M. Gil, Monica Lopez, Gemma M. Ramos-Forner, José Eliseo Blanco, Anna Moreno, Marta Lázaro-Rodríguez, Oscar Vaquerizo, Beatriz Soriano, Marta Fabre, Elena Gomez-Valencia, Ana Cuiña, Nicolas Alayon, Jose Antonio Sainz-Bueno, Angels Vives, Esther Esteve, Vanesa Ocaña, Miguel Ángel López, Anna Maroto, Elena Carreras, Manel Mendoza
Small fetuses, with estimated fetal weight (EFW) below the tenth percentile, are classified as fetal growth restriction (FGR) or small for gestational age (SGA) based on prenatal ultrasound. FGR fetuses have a greater risk of stillbirth and perinatal complications and may benefit from serial ultrasound scans to guide early delivery. Abnormal serum angiogenic factors, such as the soluble fms-like tyrosine kinase-1 (sFlt-1):placental growth factor (PlGF) ratio, have shown potential to more accurately distinguish FGR from SGA, with fewer false positives. This randomized controlled trial compared a management protocol based on the sFlt-1:PlGF with EFW and Doppler ultrasound in avoiding adverse perinatal outcomes in small fetuses after 36 weeks of gestation. A total of 1,088 pregnant women with singleton pregnancies were randomized to either the Doppler-based (control) or the sFlt-1:PlGF-based (intervention) protocol. The primary outcome, neonatal acidosis or Cesarean delivery as a result of abnormal cardiotocography, was assessed in 1,013 participants. The incidence was 10.5% in the intervention group and 10.0% in the control group (absolute difference, 0.53 (−3.21 to 4.26)), with the upper limit of the confidence interval <8.5%, confirming noninferiority. Thus, the sFlt-1:PlGF was noninferior to EFW and Doppler ultrasound in avoiding neonatal acidosis or Cesarean delivery owing to nonreassuring fetal status in small fetuses after 36 weeks (ClinicalTrials.gov registration: NCT04502823).
{"title":"Angiogenic factors versus fetomaternal Doppler for fetal growth restriction at term: an open-label, randomized controlled trial","authors":"Pablo Garcia-Manau, Erika Bonacina, Raquel Martin-Alonso, Lourdes Martin, Ana Palacios, Maria Luisa Sanchez-Camps, Cristina Lesmes, Ivan Hurtado, Esther Perez, Albert Tubau, Patricia Ibañez, Marina Alcoz, Nuria Valiño, Elena Moreno, Carlota Borrero, Esperanza Garcia, Eva Lopez-Quesada, Sonia Diaz, Jose Roman Broullon, Mireia Teixidor, Carolina Chulilla, Roser Ferrer-Costa, Maria M. Gil, Monica Lopez, Gemma M. Ramos-Forner, José Eliseo Blanco, Anna Moreno, Marta Lázaro-Rodríguez, Oscar Vaquerizo, Beatriz Soriano, Marta Fabre, Elena Gomez-Valencia, Ana Cuiña, Nicolas Alayon, Jose Antonio Sainz-Bueno, Angels Vives, Esther Esteve, Vanesa Ocaña, Miguel Ángel López, Anna Maroto, Elena Carreras, Manel Mendoza","doi":"10.1038/s41591-024-03421-9","DOIUrl":"https://doi.org/10.1038/s41591-024-03421-9","url":null,"abstract":"<p>Small fetuses, with estimated fetal weight (EFW) below the tenth percentile, are classified as fetal growth restriction (FGR) or small for gestational age (SGA) based on prenatal ultrasound. FGR fetuses have a greater risk of stillbirth and perinatal complications and may benefit from serial ultrasound scans to guide early delivery. Abnormal serum angiogenic factors, such as the soluble fms-like tyrosine kinase-1 (sFlt-1):placental growth factor (PlGF) ratio, have shown potential to more accurately distinguish FGR from SGA, with fewer false positives. This randomized controlled trial compared a management protocol based on the sFlt-1:PlGF with EFW and Doppler ultrasound in avoiding adverse perinatal outcomes in small fetuses after 36 weeks of gestation. A total of 1,088 pregnant women with singleton pregnancies were randomized to either the Doppler-based (control) or the sFlt-1:PlGF-based (intervention) protocol. The primary outcome, neonatal acidosis or Cesarean delivery as a result of abnormal cardiotocography, was assessed in 1,013 participants. The incidence was 10.5% in the intervention group and 10.0% in the control group (absolute difference, 0.53 (−3.21 to 4.26)), with the upper limit of the confidence interval <8.5%, confirming noninferiority. Thus, the sFlt-1:PlGF was noninferior to EFW and Doppler ultrasound in avoiding neonatal acidosis or Cesarean delivery owing to nonreassuring fetal status in small fetuses after 36 weeks (ClinicalTrials.gov registration: NCT04502823).</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"27 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07DOI: 10.1038/s41591-024-03375-y
Manuel Dote-Montero, Antonio Clavero-Jimeno, Elisa Merchán-Ramírez, Maddi Oses, Jon Echarte, Alba Camacho-Cardenosa, Mara Concepción, Francisco J. Amaro-Gahete, Juan M. A. Alcántara, Alejandro López-Vázquez, Rocío Cupeiro, Jairo H. Migueles, Alejandro De-la-O, Patricia V. García Pérez, Victoria Contreras-Bolivar, Araceli Muñoz-Garach, Ana Zugasti, Estrella Petrina, Natalia Alvarez de Eulate, Elena Goñi, Cristina Armendariz-Brugos, Maria T. González Cejudo, Jose L. Martín-Rodríguez, Fernando Idoate, Rafael Cabeza, Almudena Carneiro-Barrera, Rafael de Cabo, Manuel Muñoz-Torres, Idoia Labayen, Jonatan R. Ruiz
The optimal eating window for time-restricted eating (TRE) remains unclear, particularly its impact on visceral adipose tissue (VAT), which is associated with cardiometabolic morbidity and mortality. We investigated the effects of three TRE schedules (8 h windows in the early day, late day and participant-chosen times) combined with usual care (UC, based on education about the Mediterranean diet) versus UC alone over 12 weeks in adults with overweight or obesity. The primary outcome was VAT changes measured by magnetic resonance imaging. A total of 197 participants were randomized to UC (n = 49), early TRE (n = 49), late TRE (n = 52) or self-selected TRE (n = 47). No significant differences were found in VAT changes between early TRE (mean difference (MD): −4%; 95% confidence interval (CI), −12 to 4; P = 0.87), late TRE (MD: −6%; 95% CI, −13 to 2; P = 0.31) and self-selected TRE (MD: −3%; 95% CI, −11 to 5; P ≥ 0.99) compared with UC, nor among the TRE groups (all P ≥ 0.99). No serious adverse events occurred; five participants reported mild adverse events. Adherence was high (85–88%) across TRE groups. These findings suggest that adding TRE, irrespective of eating window timing, offers no additional benefit over a Mediterranean diet alone in reducing VAT. TRE appears to be a safe, well-tolerated and feasible dietary approach for adults with overweight or obesity. ClinicalTrials.gov registration: NCT05310721.
{"title":"Effects of early, late and self-selected time-restricted eating on visceral adipose tissue and cardiometabolic health in participants with overweight or obesity: a randomized controlled trial","authors":"Manuel Dote-Montero, Antonio Clavero-Jimeno, Elisa Merchán-Ramírez, Maddi Oses, Jon Echarte, Alba Camacho-Cardenosa, Mara Concepción, Francisco J. Amaro-Gahete, Juan M. A. Alcántara, Alejandro López-Vázquez, Rocío Cupeiro, Jairo H. Migueles, Alejandro De-la-O, Patricia V. García Pérez, Victoria Contreras-Bolivar, Araceli Muñoz-Garach, Ana Zugasti, Estrella Petrina, Natalia Alvarez de Eulate, Elena Goñi, Cristina Armendariz-Brugos, Maria T. González Cejudo, Jose L. Martín-Rodríguez, Fernando Idoate, Rafael Cabeza, Almudena Carneiro-Barrera, Rafael de Cabo, Manuel Muñoz-Torres, Idoia Labayen, Jonatan R. Ruiz","doi":"10.1038/s41591-024-03375-y","DOIUrl":"https://doi.org/10.1038/s41591-024-03375-y","url":null,"abstract":"<p>The optimal eating window for time-restricted eating (TRE) remains unclear, particularly its impact on visceral adipose tissue (VAT), which is associated with cardiometabolic morbidity and mortality. We investigated the effects of three TRE schedules (8 h windows in the early day, late day and participant-chosen times) combined with usual care (UC, based on education about the Mediterranean diet) versus UC alone over 12 weeks in adults with overweight or obesity. The primary outcome was VAT changes measured by magnetic resonance imaging. A total of 197 participants were randomized to UC (<i>n</i> = 49), early TRE (<i>n</i> = 49), late TRE (<i>n</i> = 52) or self-selected TRE (<i>n</i> = 47). No significant differences were found in VAT changes between early TRE (mean difference (MD): −4%; 95% confidence interval (CI), −12 to 4; <i>P</i> = 0.87), late TRE (MD: −6%; 95% CI, −13 to 2; <i>P</i> = 0.31) and self-selected TRE (MD: −3%; 95% CI, −11 to 5; <i>P</i> ≥ 0.99) compared with UC, nor among the TRE groups (all <i>P</i> ≥ 0.99). No serious adverse events occurred; five participants reported mild adverse events. Adherence was high (85–88%) across TRE groups. These findings suggest that adding TRE, irrespective of eating window timing, offers no additional benefit over a Mediterranean diet alone in reducing VAT. TRE appears to be a safe, well-tolerated and feasible dietary approach for adults with overweight or obesity. ClinicalTrials.gov registration: NCT05310721.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"99 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934701","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06DOI: 10.1038/s41591-024-03364-1
Melissa D. McCradden, Alex John London, Judy Wawira Gichoya, Mark Sendak, Lauren Erdman, Ian Stedman, Lauren Oakden-Rayner, Ismail Akrout, James A. Anderson, Lesley-Anne Farmer, Robert Greer, Anna Goldenberg, Yvonne Ho, Shalmali Joshi, Jennie Louise, Muhammad Mamdani, Mjaye L. Mazwi, Abdullahi Mohamud, Lyle J. Palmer, Antonios Peperidis, Stephen R. Pfohl, Mandy Rickard, Carolyn Semmler, Karandeep Singh, Devin Singh, Seyi Soremekun, Lana Tikhomirov, Anton H. van der Vegt, Karin Verspoor, Xiaoxuan Liu
Over the past few years, authoritative, trustworthy guidance for the clinical translation of artificial intelligence (AI) has formed mainly around two areas: responsible model development and validation; and prospective clinical trials. Initial work focused on building a good model, which generally means a model that demonstrates good performance, addresses an important clinical task, trains on the right data to address that task and could be used for some meaningful goal1. The model should then be assessed against sets of unseen cases to ensure it can generalize beyond the test set and can potentially be tested externally. Further practices are emerging around ongoing monitoring to detect model drift and performance changes. Collectively, these practices are characterized as responsible machine learning.
However, the distinctions between the in silico context and the clinical environment are substantial, which highlights the need for clinical evaluations. In 2020, the SPIRIT-AI and CONSORT-AI reporting guidelines were published to establish the minimum reporting criteria for the conduct of prospective, interventional clinical trials evaluating the impact of an AI model2,3. The DECIDE-AI guidelines were published shortly thereafter to address first-in-human feasibility trials of AI tools4. Regulatory frameworks emphasize the importance of clinical evidence, but precisely what kind and degree of evidence is needed for the approval of clinical AI applications is a matter of ongoing uncertainty5.
{"title":"CANAIRI: the Collaboration for Translational Artificial Intelligence Trials in healthcare","authors":"Melissa D. McCradden, Alex John London, Judy Wawira Gichoya, Mark Sendak, Lauren Erdman, Ian Stedman, Lauren Oakden-Rayner, Ismail Akrout, James A. Anderson, Lesley-Anne Farmer, Robert Greer, Anna Goldenberg, Yvonne Ho, Shalmali Joshi, Jennie Louise, Muhammad Mamdani, Mjaye L. Mazwi, Abdullahi Mohamud, Lyle J. Palmer, Antonios Peperidis, Stephen R. Pfohl, Mandy Rickard, Carolyn Semmler, Karandeep Singh, Devin Singh, Seyi Soremekun, Lana Tikhomirov, Anton H. van der Vegt, Karin Verspoor, Xiaoxuan Liu","doi":"10.1038/s41591-024-03364-1","DOIUrl":"https://doi.org/10.1038/s41591-024-03364-1","url":null,"abstract":"<p>Over the past few years, authoritative, trustworthy guidance for the clinical translation of artificial intelligence (AI) has formed mainly around two areas: responsible model development and validation; and prospective clinical trials. Initial work focused on building a good model, which generally means a model that demonstrates good performance, addresses an important clinical task, trains on the right data to address that task and could be used for some meaningful goal<sup>1</sup>. The model should then be assessed against sets of unseen cases to ensure it can generalize beyond the test set and can potentially be tested externally. Further practices are emerging around ongoing monitoring to detect model drift and performance changes. Collectively, these practices are characterized as responsible machine learning.</p><p>However, the distinctions between the in silico context and the clinical environment are substantial, which highlights the need for clinical evaluations. In 2020, the SPIRIT-AI and CONSORT-AI reporting guidelines were published to establish the minimum reporting criteria for the conduct of prospective, interventional clinical trials evaluating the impact of an AI model<sup>2,3</sup>. The DECIDE-AI guidelines were published shortly thereafter to address first-in-human feasibility trials of AI tools<sup>4</sup>. Regulatory frameworks emphasize the importance of clinical evidence, but precisely what kind and degree of evidence is needed for the approval of clinical AI applications is a matter of ongoing uncertainty<sup>5</sup>.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"25 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142929742","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06DOI: 10.1038/s41591-024-03398-5
Seong-Keun Yoo, Conall W. Fitzgerald, Byuri Angela Cho, Bailey G. Fitzgerald, Catherine Han, Elizabeth S. Koh, Abhinav Pandey, Hannah Sfreddo, Fionnuala Crowley, Michelle Rudshteyn Korostin, Neha Debnath, Yan Leyfman, Cristina Valero, Mark Lee, Joris L. Vos, Andrew Sangho Lee, Karena Zhao, Stanley Lam, Ezekiel Olumuyide, Fengshen Kuo, Eric A. Wilson, Pauline Hamon, Clotilde Hennequin, Miriam Saffern, Lynda Vuong, A. Ari Hakimi, Brian Brown, Miriam Merad, Sacha Gnjatic, Nina Bhardwaj, Matthew D. Galsky, Eric E. Schadt, Robert M. Samstein, Thomas U. Marron, Mithat Gönen, Luc G. T. Morris, Diego Chowell
Predicting whether a patient with cancer will benefit from immune checkpoint inhibitors (ICIs) without resorting to advanced genomic or immunologic assays is an important clinical need. To address this, we developed and evaluated SCORPIO, a machine learning system that utilizes routine blood tests (complete blood count and comprehensive metabolic profile) alongside clinical characteristics from 9,745 ICI-treated patients across 21 cancer types. SCORPIO was trained on data from 1,628 patients across 17 cancer types from Memorial Sloan Kettering Cancer Center. In two internal test sets comprising 2,511 patients across 19 cancer types, SCORPIO achieved median time-dependent area under the receiver operating characteristic curve (AUC(t)) values of 0.763 and 0.759 for predicting overall survival at 6, 12, 18, 24 and 30 months, outperforming tumor mutational burden (TMB), which showed median AUC(t) values of 0.503 and 0.543. Additionally, SCORPIO demonstrated superior predictive performance for predicting clinical benefit (tumor response or prolonged stability), with AUC values of 0.714 and 0.641, compared to TMB (AUC = 0.546 and 0.573). External validation was performed using 10 global phase 3 trials (4,447 patients across 6 cancer types) and a real-world cohort from the Mount Sinai Health System (1,159 patients across 18 cancer types). In these external cohorts, SCORPIO maintained robust performance in predicting ICI outcomes, surpassing programmed death-ligand 1 immunostaining. These findings underscore SCORPIO’s reliability and adaptability, highlighting its potential to predict patient outcomes with ICI therapy across diverse cancer types and healthcare settings.
{"title":"Prediction of checkpoint inhibitor immunotherapy efficacy for cancer using routine blood tests and clinical data","authors":"Seong-Keun Yoo, Conall W. Fitzgerald, Byuri Angela Cho, Bailey G. Fitzgerald, Catherine Han, Elizabeth S. Koh, Abhinav Pandey, Hannah Sfreddo, Fionnuala Crowley, Michelle Rudshteyn Korostin, Neha Debnath, Yan Leyfman, Cristina Valero, Mark Lee, Joris L. Vos, Andrew Sangho Lee, Karena Zhao, Stanley Lam, Ezekiel Olumuyide, Fengshen Kuo, Eric A. Wilson, Pauline Hamon, Clotilde Hennequin, Miriam Saffern, Lynda Vuong, A. Ari Hakimi, Brian Brown, Miriam Merad, Sacha Gnjatic, Nina Bhardwaj, Matthew D. Galsky, Eric E. Schadt, Robert M. Samstein, Thomas U. Marron, Mithat Gönen, Luc G. T. Morris, Diego Chowell","doi":"10.1038/s41591-024-03398-5","DOIUrl":"https://doi.org/10.1038/s41591-024-03398-5","url":null,"abstract":"<p>Predicting whether a patient with cancer will benefit from immune checkpoint inhibitors (ICIs) without resorting to advanced genomic or immunologic assays is an important clinical need. To address this, we developed and evaluated SCORPIO, a machine learning system that utilizes routine blood tests (complete blood count and comprehensive metabolic profile) alongside clinical characteristics from 9,745 ICI-treated patients across 21 cancer types. SCORPIO was trained on data from 1,628 patients across 17 cancer types from Memorial Sloan Kettering Cancer Center. In two internal test sets comprising 2,511 patients across 19 cancer types, SCORPIO achieved median time-dependent area under the receiver operating characteristic curve (AUC(t)) values of 0.763 and 0.759 for predicting overall survival at 6, 12, 18, 24 and 30 months, outperforming tumor mutational burden (TMB), which showed median AUC(t) values of 0.503 and 0.543. Additionally, SCORPIO demonstrated superior predictive performance for predicting clinical benefit (tumor response or prolonged stability), with AUC values of 0.714 and 0.641, compared to TMB (AUC = 0.546 and 0.573). External validation was performed using 10 global phase 3 trials (4,447 patients across 6 cancer types) and a real-world cohort from the Mount Sinai Health System (1,159 patients across 18 cancer types). In these external cohorts, SCORPIO maintained robust performance in predicting ICI outcomes, surpassing programmed death-ligand 1 immunostaining. These findings underscore SCORPIO’s reliability and adaptability, highlighting its potential to predict patient outcomes with ICI therapy across diverse cancer types and healthcare settings.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"42 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142929743","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06DOI: 10.1038/s41591-024-03361-4
Andrea M. Gross, Geraldine O’Sullivan Coyne, Eva Dombi, Cecilia Tibery, William G. Herrick, Staci Martin, Steven P. Angus, Jack F. Shern, Steven D. Rhodes, Jared C. Foster, Larry V. Rubinstein, Andrea Baldwin, Christopher Davis, Shelley A. H. Dixon, Margaret Fagan, Mary Jane Ong, Pamela L. Wolters, Mary Anne Tamula, Olivia Reid, Hari Sankaran, Fang Fang, Jeevan Prasaad Govindharajulu, Alice T. Browne, Rosandra N. Kaplan, Kara Heisey, Thomas J. On, Xiaoling Xuei, Xiyuan Zhang, Barry C. Johnson, Ralph E. Parchment, D. Wade Clapp, Apurva K. Srivastava, James H. Doroshow, Alice P. Chen, Brigitte C. Widemann
The MEK inhibitor selumetinib induces objective responses and provides clinical benefit in children with neurofibromatosis type 1 (NF1) and inoperable plexiform neurofibromas (PNs). To evaluate whether similar outcomes were possible in adult patients, in whom PN growth is generally slower than in pediatric patients, we conducted an open-label phase 2 study of selumetinib in adults with NF1 PNs. The study was designed to evaluate objective response rate (primary objective), tumor volumetric responses, patient-reported outcomes and pharmacodynamic effects in PN biopsies. The objective response rate was 63.6% (21/33 participants). Median maximal PN volume decrease was 23.6% (range: −48.1% to 5.5%). No disease progression relative to baseline PN volumes occurred before data cutoff, with a median of 28 cycles completed (range: 1–78, 28 d per cycle). Participants experienced decreased tumor pain intensity and pain interference. Adverse events (AEs) were similar to those of the pediatric trial; acneiform rash was the most prevalent AE. Phosphorylation ratios of ERK1/2 decreased significantly (ERK1 median change: −64.6% (range: −99.5% to 90.7%), ERK2 median change: −57.3% (range: −99.9% to 84.4%)) in paired PN biopsies (P ≤ 0.001 for both isoforms) without compensatory phosphorylation of AKT1/2/3. The sustained PN volume decreases, associated improvement in pain and manageable AE profile indicate that selumetinib provides benefit to adults with NF1 and inoperable PNs. ClinicalTrials.gov identifier: NCT02407405.
{"title":"Selumetinib in adults with NF1 and inoperable plexiform neurofibroma: a phase 2 trial","authors":"Andrea M. Gross, Geraldine O’Sullivan Coyne, Eva Dombi, Cecilia Tibery, William G. Herrick, Staci Martin, Steven P. Angus, Jack F. Shern, Steven D. Rhodes, Jared C. Foster, Larry V. Rubinstein, Andrea Baldwin, Christopher Davis, Shelley A. H. Dixon, Margaret Fagan, Mary Jane Ong, Pamela L. Wolters, Mary Anne Tamula, Olivia Reid, Hari Sankaran, Fang Fang, Jeevan Prasaad Govindharajulu, Alice T. Browne, Rosandra N. Kaplan, Kara Heisey, Thomas J. On, Xiaoling Xuei, Xiyuan Zhang, Barry C. Johnson, Ralph E. Parchment, D. Wade Clapp, Apurva K. Srivastava, James H. Doroshow, Alice P. Chen, Brigitte C. Widemann","doi":"10.1038/s41591-024-03361-4","DOIUrl":"https://doi.org/10.1038/s41591-024-03361-4","url":null,"abstract":"<p>The MEK inhibitor selumetinib induces objective responses and provides clinical benefit in children with neurofibromatosis type 1 (NF1) and inoperable plexiform neurofibromas (PNs). To evaluate whether similar outcomes were possible in adult patients, in whom PN growth is generally slower than in pediatric patients, we conducted an open-label phase 2 study of selumetinib in adults with NF1 PNs. The study was designed to evaluate objective response rate (primary objective), tumor volumetric responses, patient-reported outcomes and pharmacodynamic effects in PN biopsies. The objective response rate was 63.6% (21/33 participants). Median maximal PN volume decrease was 23.6% (range: −48.1% to 5.5%). No disease progression relative to baseline PN volumes occurred before data cutoff, with a median of 28 cycles completed (range: 1–78, 28 d per cycle). Participants experienced decreased tumor pain intensity and pain interference. Adverse events (AEs) were similar to those of the pediatric trial; acneiform rash was the most prevalent AE. Phosphorylation ratios of ERK1/2 decreased significantly (ERK1 median change: −64.6% (range: −99.5% to 90.7%), ERK2 median change: −57.3% (range: −99.9% to 84.4%)) in paired PN biopsies (<i>P</i> ≤ 0.001 for both isoforms) without compensatory phosphorylation of AKT1/2/3. The sustained PN volume decreases, associated improvement in pain and manageable AE profile indicate that selumetinib provides benefit to adults with NF1 and inoperable PNs. ClinicalTrials.gov identifier: NCT02407405.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"34 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142929805","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06DOI: 10.1038/s41591-024-03345-4
Laura Lara-Castor, Meghan O’Hearn, Frederick Cudhea, Victoria Miller, Peilin Shi, Jianyi Zhang, Julia R. Sharib, Sean B. Cash, Simon Barquera, Renata Micha, Dariush Mozaffarian
The consumption of sugar-sweetened beverages (SSBs) is associated with type 2 diabetes (T2D) and cardiovascular diseases (CVD). However, an updated and comprehensive assessment of the global burden attributable to SSBs remains scarce. Here we estimated SSB-attributable T2D and CVD burdens across 184 countries in 1990 and 2020 globally, regionally and nationally, incorporating data from the Global Dietary Database, jointly stratified by age, sex, educational attainment and urbanicity. In 2020, 2.2 million (95% uncertainty interval 2.0–2.3) new T2D cases and 1.2 million (95% uncertainty interval 1.1–1.3) new CVD cases were attributable to SSBs worldwide, representing 9.8% and 3.1%, respectively, of all incident cases. Globally, proportional SSB-attributable burdens were higher among men versus women, younger versus older adults, higher- versus lower-educated adults, and adults in urban versus rural areas. By world region, the highest SSB-attributable percentage burdens were in Latin America and the Caribbean (T2D: 24.4%; CVD: 11.3%) and sub-Saharan Africa (T2D: 21.5%; CVD: 10.5%). From 1990 to 2020, the largest proportional increases in SSB-attributable incident T2D and CVD cases were in sub-Saharan Africa (+8.8% and +4.4%, respectively). Our study highlights the countries and subpopulations most affected by cardiometabolic disease associated with SSB consumption, assisting in shaping effective policies and interventions to reduce these burdens globally.
{"title":"Burdens of type 2 diabetes and cardiovascular disease attributable to sugar-sweetened beverages in 184 countries","authors":"Laura Lara-Castor, Meghan O’Hearn, Frederick Cudhea, Victoria Miller, Peilin Shi, Jianyi Zhang, Julia R. Sharib, Sean B. Cash, Simon Barquera, Renata Micha, Dariush Mozaffarian","doi":"10.1038/s41591-024-03345-4","DOIUrl":"https://doi.org/10.1038/s41591-024-03345-4","url":null,"abstract":"<p>The consumption of sugar-sweetened beverages (SSBs) is associated with type 2 diabetes (T2D) and cardiovascular diseases (CVD). However, an updated and comprehensive assessment of the global burden attributable to SSBs remains scarce. Here we estimated SSB-attributable T2D and CVD burdens across 184 countries in 1990 and 2020 globally, regionally and nationally, incorporating data from the Global Dietary Database, jointly stratified by age, sex, educational attainment and urbanicity. In 2020, 2.2 million (95% uncertainty interval 2.0–2.3) new T2D cases and 1.2 million (95% uncertainty interval 1.1–1.3) new CVD cases were attributable to SSBs worldwide, representing 9.8% and 3.1%, respectively, of all incident cases. Globally, proportional SSB-attributable burdens were higher among men versus women, younger versus older adults, higher- versus lower-educated adults, and adults in urban versus rural areas. By world region, the highest SSB-attributable percentage burdens were in Latin America and the Caribbean (T2D: 24.4%; CVD: 11.3%) and sub-Saharan Africa (T2D: 21.5%; CVD: 10.5%). From 1990 to 2020, the largest proportional increases in SSB-attributable incident T2D and CVD cases were in sub-Saharan Africa (+8.8% and +4.4%, respectively). Our study highlights the countries and subpopulations most affected by cardiometabolic disease associated with SSB consumption, assisting in shaping effective policies and interventions to reduce these burdens globally.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"46 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06DOI: 10.1038/s41591-024-03385-w
Sant-Rayn Pasricha, Ernest Moya, Ricardo Ataíde, Glory Mzembe, Rebecca Harding, Martin N. Mwangi, Truwah Zinenani, Khic-Houy Prang, Justina Kaunda, Owen P. L. Mtambo, Maclean Vokhiwa, Gomezgani Mhango, Elisabeth Mamani-Mategula, Katherine Fielding, Ayşe Demir, Naomi Von Dinklage, Hans Verhoef, Alistair RD McLean, Lucinda Manda-Taylor, Sabine Braat, Kamija S. Phiri
Over 46% of African pregnant women are anemic. Oral iron is recommended but often suboptimal, particularly late in pregnancy. Intravenous ferric carboxymaltose (FCM) could treat anemia in women in the third trimester in sub-Saharan Africa. In an open-label, individually randomized trial in antenatal clinics in southern Malawi, we randomized 590 women at 27–35 weeks of gestation with capillary hemoglobin <10.0 g dl−1 to FCM (20 mg kg−1 up to 1,000 mg, once at enrollment) or standard of care (60 mg elemental iron, twice daily for 90 days). Participants and their infants were followed to 4 weeks postpartum. Primary outcomes were maternal anemia at 36 weeks’ gestation or delivery (whichever occurred first) and neonatal birthweight. At the primary timepoint, 126 of 270 (46.7%) of women in the FCM group were anemic, compared to 170 of 271 (67.3%) women in the standard-of-care group (PR, 0.74 (95% CI 0.64, 0.87); P = 0.0002). There was no difference between groups in birthweight (mean difference 10.9 g (−65.7, 87.5 g); P = 0.78). No serious infusion-related reactions occurred, and there were no differences in adverse events between groups. In Malawian women in late pregnancy, FCM effectively and safely reduced anemia before childbirth. Australia New Zealand Clinical Trial registration: ANZCTR12621001239853
{"title":"Ferric carboxymaltose for anemia in late pregnancy: a randomized controlled trial","authors":"Sant-Rayn Pasricha, Ernest Moya, Ricardo Ataíde, Glory Mzembe, Rebecca Harding, Martin N. Mwangi, Truwah Zinenani, Khic-Houy Prang, Justina Kaunda, Owen P. L. Mtambo, Maclean Vokhiwa, Gomezgani Mhango, Elisabeth Mamani-Mategula, Katherine Fielding, Ayşe Demir, Naomi Von Dinklage, Hans Verhoef, Alistair RD McLean, Lucinda Manda-Taylor, Sabine Braat, Kamija S. Phiri","doi":"10.1038/s41591-024-03385-w","DOIUrl":"https://doi.org/10.1038/s41591-024-03385-w","url":null,"abstract":"<p>Over 46% of African pregnant women are anemic. Oral iron is recommended but often suboptimal, particularly late in pregnancy. Intravenous ferric carboxymaltose (FCM) could treat anemia in women in the third trimester in sub-Saharan Africa. In an open-label, individually randomized trial in antenatal clinics in southern Malawi, we randomized 590 women at 27–35 weeks of gestation with capillary hemoglobin <10.0 g dl<sup>−1</sup> to FCM (20 mg kg<sup>−1</sup> up to 1,000 mg, once at enrollment) or standard of care (60 mg elemental iron, twice daily for 90 days). Participants and their infants were followed to 4 weeks postpartum. Primary outcomes were maternal anemia at 36 weeks’ gestation or delivery (whichever occurred first) and neonatal birthweight. At the primary timepoint, 126 of 270 (46.7%) of women in the FCM group were anemic, compared to 170 of 271 (67.3%) women in the standard-of-care group (PR, 0.74 (95% CI 0.64, 0.87); <i>P</i> = 0.0002). There was no difference between groups in birthweight (mean difference 10.9 g (−65.7, 87.5 g); <i>P</i> = 0.78). No serious infusion-related reactions occurred, and there were no differences in adverse events between groups. In Malawian women in late pregnancy, FCM effectively and safely reduced anemia before childbirth. Australia New Zealand Clinical Trial registration: ANZCTR12621001239853</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"28 1","pages":""},"PeriodicalIF":82.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142929746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}