Nature Reviews Clinical Oncology Nature Reviews Clinical Oncology is a peer-reviewed journal for oncologists and affiliated health-care professionals. The journal delivers timely interpretations of key scientific developments in oncology and related areas of study. Nature Reviews Clinical Oncology is published monthly in print and online, and includes news, commentary and opinion pieces, and comprehensive reviews. Articles are subject to rigorous peer-review and/or review by in-house editors. Topics covered include epidemiology, screening, diagnosis, pathology, prevention, chemotherapy, radiotherapy, surgical oncology, medical oncology, targeted therapies, hormonal therapies, hematology, immunotherapy, imaging, palliative care, pediatric oncology, genetics and pharmacology.           http://feeds.nature.com/nrclinonc/rss/current Nature Publishing Group en © 2024 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. Nature Reviews Clinical Oncology © 2024 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. permissions@nature.com Nature Reviews Clinical Oncology https://www.nature.com/uploads/product/nrclinonc/rss.gif http://feeds.nature.com/nrclinonc/rss/current <![CDATA[Targeting cuproplasia and cuproptosis in cancer]]> https://www.nature.com/articles/s41571-024-00876-0 Nature Reviews Clinical Oncology, Published online: 14 March 2024; doi:10.1038/s41571-024-00876-0

Copper is an essential trace element with inherent redox properties and fundamental roles in a diverse range of biological processes; therefore, maintaining copper homeostasis is crucial. In this Review, the authors discuss new insights into the mechanisms by which disrupted copper homeostasis contributes to tumour initiation and development, including the recently defined concepts of cuproplasia (copper-dependent cell growth and proliferation) and cuproptosis (a mitochondrial pathway of cell death triggered by excessive copper exposure). They also discuss potential strategies to exploit cuproplasia and cuproptosis for the treatment of cancer.]]>
Daolin TangGuido KroemerRui Kang doi:10.1038/s41571-024-00876-0 Nature Reviews Clinical Oncology, Published online: 2024-03-14; | doi:10.1038/s41571-024-00876-0 2024-03-14 Nature Reviews Clinical Oncology 10.1038/s41571-024-00876-0 https://www.nature.com/articles/s41571-024-00876-0
<![CDATA[The promise of AI in personalized breast cancer screening: are we there yet?]]> https://www.nature.com/articles/s41571-024-00877-z Nature Reviews Clinical Oncology, Published online: 12 March 2024; doi:10.1038/s41571-024-00877-z

The benefits and potential harms of mammography-based screening for breast cancer are often a matter of debate. Here, I discuss the promises and limitations of a recent study that tested an artificial intelligence-based tool for the detection of breast cancer in digital mammograms in a large, prospective screening setting.]]>
Despina Kontos doi:10.1038/s41571-024-00877-z Nature Reviews Clinical Oncology, Published online: 2024-03-12; | doi:10.1038/s41571-024-00877-z 2024-03-12 Nature Reviews Clinical Oncology 10.1038/s41571-024-00877-z https://www.nature.com/articles/s41571-024-00877-z
<![CDATA[FIRSTMAPPP prospectively charts the efficacy of sunitinib for phaeochromocytoma and paraganglioma]]> https://www.nature.com/articles/s41571-024-00880-4 Nature Reviews Clinical Oncology, Published online: 08 March 2024; doi:10.1038/s41571-024-00880-4

FIRSTMAPPP prospectively charts the efficacy of sunitinib for phaeochromocytoma and paraganglioma]]>
David Killock doi:10.1038/s41571-024-00880-4 Nature Reviews Clinical Oncology, Published online: 2024-03-08; | doi:10.1038/s41571-024-00880-4 2024-03-08 Nature Reviews Clinical Oncology 10.1038/s41571-024-00880-4 https://www.nature.com/articles/s41571-024-00880-4
<![CDATA[Non-inferiority of simple versus radical hysterectomy in low-risk cervical cancer]]> https://www.nature.com/articles/s41571-024-00879-x Nature Reviews Clinical Oncology, Published online: 06 March 2024; doi:10.1038/s41571-024-00879-x

Non-inferiority of simple versus radical hysterectomy in low-risk cervical cancer]]>
Diana Romero doi:10.1038/s41571-024-00879-x Nature Reviews Clinical Oncology, Published online: 2024-03-06; | doi:10.1038/s41571-024-00879-x 2024-03-06 Nature Reviews Clinical Oncology 10.1038/s41571-024-00879-x https://www.nature.com/articles/s41571-024-00879-x
<![CDATA[Pegargiminase improves outcomes in nonepithelioid MPM]]> https://www.nature.com/articles/s41571-024-00878-y Nature Reviews Clinical Oncology, Published online: 04 March 2024; doi:10.1038/s41571-024-00878-y

Pegargiminase improves outcomes in nonepithelioid MPM]]>
Peter Sidaway doi:10.1038/s41571-024-00878-y Nature Reviews Clinical Oncology, Published online: 2024-03-04; | doi:10.1038/s41571-024-00878-y 2024-03-04 Nature Reviews Clinical Oncology 10.1038/s41571-024-00878-y https://www.nature.com/articles/s41571-024-00878-y
<![CDATA[FGFR-targeted therapeutics: clinical activity, mechanisms of resistance and new directions]]> https://www.nature.com/articles/s41571-024-00869-z Nature Reviews Clinical Oncology, Published online: 29 February 2024; doi:10.1038/s41571-024-00869-z

FGFR inhibitors are now approved for use in patients with advanced-stage urothelial carcinoma, cholangiocarcinoma and myeloid or lymphoid neoplasms that harbour certain FGFR alterations. Nonetheless, challenges such as tolerability and acquired resistance limit the clinical potential of these agents. In this Review, the authors summarize the available clinical data on FGFR inhibitors, describe promising novel agents and highlight future research directions that might optimize the efficacy of FGFR-targeted therapies.]]>
Masuko KatohYohann LoriotGiovanni BrandiSimona TavolariZev A. WainbergMasaru Katoh doi:10.1038/s41571-024-00869-z Nature Reviews Clinical Oncology, Published online: 2024-02-29; | doi:10.1038/s41571-024-00869-z 2024-02-29 Nature Reviews Clinical Oncology 10.1038/s41571-024-00869-z https://www.nature.com/articles/s41571-024-00869-z
<![CDATA[Adjuvant and neoadjuvant immunotherapies in hepatocellular carcinoma]]> https://www.nature.com/articles/s41571-024-00868-0 Nature Reviews Clinical Oncology, Published online: 29 February 2024; doi:10.1038/s41571-024-00868-0

Patients with early stage hepatocellular carcinoma typically undergo resection, liver transplantation or local ablation; however, 30–50% will have disease recurrence at 3 years. The authors of this Review describe the tumour immune microenvironment and mechanism of action of immunotherapies, and discuss the available evidence from phase II/III trials of neoadjuvant and adjuvant treatment approaches in this setting.]]>
Josep M. LlovetRoser PinyolMark YarchoanAmit G. SingalThomas U. MarronMyron SchwartzEli PikarskyMasatoshi KudoRichard S. Finn doi:10.1038/s41571-024-00868-0 Nature Reviews Clinical Oncology, Published online: 2024-02-29; | doi:10.1038/s41571-024-00868-0 2024-02-29 Nature Reviews Clinical Oncology 10.1038/s41571-024-00868-0 https://www.nature.com/articles/s41571-024-00868-0
<![CDATA[Regulatory T cell-mediated immunosuppression orchestrated by cancer: towards an immuno-genomic paradigm for precision medicine]]> https://www.nature.com/articles/s41571-024-00870-6 Nature Reviews Clinical Oncology, Published online: 29 February 2024; doi:10.1038/s41571-024-00870-6

Increasing evidence indicates that signalling networks activated downstream of oncogenic alterations contribute fundamentally to cancer immune evasion, including by promoting the accumulation of regulatory T (Treg) cells and other immunosuppressive cells in the tumour microenvironment (TME). Herein, the authors discuss the mechanisms via which cancers engage Treg cells to evade antitumour immunity, as well as the characteristics of Treg cells in the TME and their roles in resistance to immune-checkpoint inhibitors. Considering these aspects, they propose the concept of ‘immuno-genomic cancer evolution’ for tumorigenesis and the related paradigm of ‘immuno-genomic precision medicine’, postulating that the specific characteristics of cancer, especially genetic profiles that correlate with particular immunosuppressive networks in the TME, are likely to inform individualized strategies for combining molecularly targeted agents with immunotherapies.]]>
Shogo KumagaiKota ItahashiHiroyoshi Nishikawa doi:10.1038/s41571-024-00870-6 Nature Reviews Clinical Oncology, Published online: 2024-02-29; | doi:10.1038/s41571-024-00870-6 2024-02-29 Nature Reviews Clinical Oncology 10.1038/s41571-024-00870-6 https://www.nature.com/articles/s41571-024-00870-6