Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Clinical innovation and scientific progress in GLP-1 medicine (Nov 2025)
    • Pancreatic Cancer (Jul 2025)
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact

Oncology

  • 1,389 Articles
  • 14 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 138
  • 139
  • Next →
HDAC5 deficiency induces intrinsic resistance to KRAS inhibition by disrupting c-Myc acetylation-ubiquitination homeostasis
Taoyu Chen, Haixin Yu, Keshan Wang, Gengdu Qin, Yuhan Zhao, Xueyi Liang, Yuxuan Li, Tianhao Zou, Jiaying Liu, Jingyuan Zhao, Zhiqiang Liu, Ruozheng Wei, Bo Wang, Shanmiao Gou, Tao Yin, Heshui Wu, Xin Jin, Yingke Zhou
Taoyu Chen, Haixin Yu, Keshan Wang, Gengdu Qin, Yuhan Zhao, Xueyi Liang, Yuxuan Li, Tianhao Zou, Jiaying Liu, Jingyuan Zhao, Zhiqiang Liu, Ruozheng Wei, Bo Wang, Shanmiao Gou, Tao Yin, Heshui Wu, Xin Jin, Yingke Zhou
View: Text | PDF

HDAC5 deficiency induces intrinsic resistance to KRAS inhibition by disrupting c-Myc acetylation-ubiquitination homeostasis

  • Text
  • PDF
Abstract

KRAS mutations serve as key oncogenic drivers in the initiation and progression of pancreatic ductal adenocarcinoma (PDAC). Despite the advancement of KRAS inhibitors like MRTX1133 for PDAC treatment, intrinsic and acquired resistance remain major barriers to their clinical efficacy. This study underscored the role of histone deacetylase 5 (HDAC5) loss in mediating intrinsic resistance to KRASG12D inhibitors. Mechanistically, HDAC5 promoted c-Myc degradation by deacetylating K148, thereby facilitating NEDD4-mediated ubiquitination at this site. The loss of HDAC5 resulted in hyperacetylation of c-Myc at K148, impeding the ubiquitination and subsequent degradation process of c-Myc following deacetylation. Consequently, c-Myc stability and transcriptional activity were sustained even under KRAS-MEK-ERK pathway inhibition, reinforcing MAPK signaling and promoting cell survival despite KRAS suppression. Our data further demonstrated that pharmacological or genetic inhibition of c-Myc effectively reversed the resistance phenotype mediated by HDAC5 loss, suggesting a therapeutic strategy centered on "KRAS-MYC dual-node blockade." Furthermore, the expression levels of HDAC5 and the acetylation status of c-Myc may serve as potential biomarkers for predicting the therapeutic response to MRTX1133. These findings provide insights into overcoming resistance to KRASG12D inhibitors and offer potential biomarkers and combinatorial therapeutic strategies for precision treatment of PDAC.

Authors

Taoyu Chen, Haixin Yu, Keshan Wang, Gengdu Qin, Yuhan Zhao, Xueyi Liang, Yuxuan Li, Tianhao Zou, Jiaying Liu, Jingyuan Zhao, Zhiqiang Liu, Ruozheng Wei, Bo Wang, Shanmiao Gou, Tao Yin, Heshui Wu, Xin Jin, Yingke Zhou

×

CD8+ T cells in the tumor microenvironment modulate response to endocrine therapy in breast cancer
Fabiana Napolitano, Yunguan Wang, Dhivya R. Sudhan, Paula I. Gonzalez-Ericsson, Luigi Formisano, Nisha Unni, Shahbano Shakeel, James Z. Zhu, Khushi Ahuja, Lei Guo, María Rosario Chica-Parrado, Yuki Matsunaga, Pamela Luna, Chang-Ching A. Lin, Yasuaki Uemoto, Kyung-Min Lee, Hongli Ma, Nathaniel J. Evans, Alberto Servetto, Saurabh Mendiratta, Spencer D. Barnes, Roberto Bianco, Yisheng V. Fang, Lin Xu, Jeon Lee, Tao Wang, Justin M. Balko, Gordon B. Mills, Marilyne Labrie, Ariella B. Hanker, Carlos L. Arteaga
Fabiana Napolitano, Yunguan Wang, Dhivya R. Sudhan, Paula I. Gonzalez-Ericsson, Luigi Formisano, Nisha Unni, Shahbano Shakeel, James Z. Zhu, Khushi Ahuja, Lei Guo, María Rosario Chica-Parrado, Yuki Matsunaga, Pamela Luna, Chang-Ching A. Lin, Yasuaki Uemoto, Kyung-Min Lee, Hongli Ma, Nathaniel J. Evans, Alberto Servetto, Saurabh Mendiratta, Spencer D. Barnes, Roberto Bianco, Yisheng V. Fang, Lin Xu, Jeon Lee, Tao Wang, Justin M. Balko, Gordon B. Mills, Marilyne Labrie, Ariella B. Hanker, Carlos L. Arteaga
View: Text | PDF

CD8+ T cells in the tumor microenvironment modulate response to endocrine therapy in breast cancer

  • Text
  • PDF
Abstract

The role of the tumor immune microenvironment (TIME) in modulating responses to antiestrogen therapy in hormone receptor-positive (HR+) breast cancers remains unclear. We analyzed pre- and on-treatment biopsies from patients with HR+ breast cancer treated with letrozole to induce estrogen deprivation (ED). Stromal tumor-infiltrating lymphocytes, assessed by H&E-staining, and immune-related gene sets, including IFNɣ signaling, measured by RNA sequencing, were increased in ED-resistant tumors. Cyclic immunofluorescence and spatial transcriptomics revealed an abundance of CD8+ T cells and enhanced antigen processing and immune gene signatures in ED-resistant tumors. In this group, the expression of CXCL9, CXCL10, and CXCL11 — chemokine genes involved in CD8+ T cell recruitment — and the CXCR3 receptor were upregulated both before and after letrozole. CXCL11 levels were higher in conditioned media from HR+ breast cancer cells co-cultured with CD8+ T cells. Both recombinant CXCL11 and co-culture with CD8+ T cells promoted MCF7 and T47D cell growth in estrogen-free conditions. Finally, deletion combined with silencing of the CXCL11 receptors CXCR3 and CXCR7 in MCF7 cells impaired proliferation in response to exogenous CXCL11 and to co-culture with CD8+ T cells in estrogen-free conditions. These findings suggest that CD8+ T cell-associated CXCL11 in the TIME modulates the response of HR+ breast cancer cells to estrogen suppression.

Authors

Fabiana Napolitano, Yunguan Wang, Dhivya R. Sudhan, Paula I. Gonzalez-Ericsson, Luigi Formisano, Nisha Unni, Shahbano Shakeel, James Z. Zhu, Khushi Ahuja, Lei Guo, María Rosario Chica-Parrado, Yuki Matsunaga, Pamela Luna, Chang-Ching A. Lin, Yasuaki Uemoto, Kyung-Min Lee, Hongli Ma, Nathaniel J. Evans, Alberto Servetto, Saurabh Mendiratta, Spencer D. Barnes, Roberto Bianco, Yisheng V. Fang, Lin Xu, Jeon Lee, Tao Wang, Justin M. Balko, Gordon B. Mills, Marilyne Labrie, Ariella B. Hanker, Carlos L. Arteaga

×

A fungal-derived cyclic peptide enhances Th9-mediated antitumor immunity by targeting ZAP70 and SREBP1
Wenli Zhao, Yang Zhou, Yuyang Chen, Yicheng Sun, Jiaxin Tang, Yihan Zhu, Jie Ren, Tianxu Du, Handuo Wang, Yuan Gao, Yu Hu, Ling Jiang, Tomohiko Ohwada, Qi Luo, Enguang Bi
Wenli Zhao, Yang Zhou, Yuyang Chen, Yicheng Sun, Jiaxin Tang, Yihan Zhu, Jie Ren, Tianxu Du, Handuo Wang, Yuan Gao, Yu Hu, Ling Jiang, Tomohiko Ohwada, Qi Luo, Enguang Bi
View: Text | PDF

A fungal-derived cyclic peptide enhances Th9-mediated antitumor immunity by targeting ZAP70 and SREBP1

  • Text
  • PDF
Abstract

Adoptive cell therapy (ACT) relies on durable and functional T cells to mediate tumor clearance. Th9 cells are a metabolically fit CD4+ T cell subset with strong persistence but limited cytotoxicity. Here, we identified endomelipeptide A (EpA), a cyclic peptide isolated from Ganoderma lucidum–associated endophytic fungi, as a potent enhancer of Th9 differentiation. EpA promoted a cytotoxic Th9 phenotype with enhanced mitochondrial function and metabolic fitness. Mechanistically, EpA dually targeted ZAP70 and SREBP1, coupling T cell receptor (TCR) signaling activation with lipid metabolism suppression. EpA-treated Th9 cells mediated robust, CD8+ T cell–dependent tumor control and enhanced the efficacy of human Th9 CAR-T therapy in vivo. These findings establish EpA as a distinct cyclic peptide that reprograms Th9 cells and provides a potential approach to boost ACT efficacy.

Authors

Wenli Zhao, Yang Zhou, Yuyang Chen, Yicheng Sun, Jiaxin Tang, Yihan Zhu, Jie Ren, Tianxu Du, Handuo Wang, Yuan Gao, Yu Hu, Ling Jiang, Tomohiko Ohwada, Qi Luo, Enguang Bi

×

Osimertinib activates a TGFβ2-dependent secretory program that drives lung adenocarcinoma progression
Madhurima Ghosh, Chao Wu, Abhishek Kumar, Monique B. Nilsson, John V. Heymach, Weina Zhao, Jiang Yu, Xin Liu, Na Ding, Shike Wang, Guan-Yu Xiao, Angelo Chen, Kate V. Grimley, William K. Russell, Chad J. Creighton, Xiaochao Tan, Jonathan M. Kurie
Madhurima Ghosh, Chao Wu, Abhishek Kumar, Monique B. Nilsson, John V. Heymach, Weina Zhao, Jiang Yu, Xin Liu, Na Ding, Shike Wang, Guan-Yu Xiao, Angelo Chen, Kate V. Grimley, William K. Russell, Chad J. Creighton, Xiaochao Tan, Jonathan M. Kurie
View: Text | PDF

Osimertinib activates a TGFβ2-dependent secretory program that drives lung adenocarcinoma progression

  • Text
  • PDF
Abstract

EGFR-mutant lung adenocarcinomas (LUADs) that are vulnerable to the EGFR antagonist Osimertinib (Osi) eventually relapse owing in part to the emergence of drug tolerant persister (DTP) cells that arise through epigenetic mechanisms. Intra-tumoral DTP cells can herald a worse clinical outcome, but the way in which DTP cells influence LUAD progression remains unclear. Osi-resistant (OR) cells exhibit typical DTP cell features, including a propensity to undergo senescence and epithelial-to-mesenchymal transition (EMT), which can activate heightened secretory states. Therefore, we postulated that OR cells influence LUAD progression through paracrine mechanisms. To test this hypothesis, we utilized congenic pairs of EGFR-mutant LUAD cell lines in which drug naive (DN) cells were rendered OR by chronic exposure to escalating doses of Osi. Co-cultured in vitro or co-injected into mice, paracrine signals from OR cells enhanced the growth and metastatic properties of DN cells. EMT and senescence activated non-overlapping secretomes, and OR cells governed DN cells by undergoing EMT but not senescence. Mechanistically, Osi rapidly increased TGFβ2 levels to initiate EMT, which triggered a Golgi remodeling process that accelerated the biogenesis and anterograde trafficking of secretory vesicles. The pro-tumorigenic activity of OR cells was diminished by depletion of EMT-dependent secreted proteins or the EMT-activating transcription factor ZEB1. These findings identify paracrine mechanisms by which OR cells drive LUAD progression.

Authors

Madhurima Ghosh, Chao Wu, Abhishek Kumar, Monique B. Nilsson, John V. Heymach, Weina Zhao, Jiang Yu, Xin Liu, Na Ding, Shike Wang, Guan-Yu Xiao, Angelo Chen, Kate V. Grimley, William K. Russell, Chad J. Creighton, Xiaochao Tan, Jonathan M. Kurie

×

Non-canonical functions of UGT2B17 promote castrate-resistant prostate cancer progression
Tingting Feng, Ning Xie, Lin Gao, Qiongqiong Jia, Sonia Kung, Tunc Morova, Yinan Li, Lin Wang, Ladan Fazli, Louis Lacombe, Chantal Guillemette, Eric Lévesque, Nathan A. Lack, Jianfei Qi, Bo Han, Xuesen Dong
Tingting Feng, Ning Xie, Lin Gao, Qiongqiong Jia, Sonia Kung, Tunc Morova, Yinan Li, Lin Wang, Ladan Fazli, Louis Lacombe, Chantal Guillemette, Eric Lévesque, Nathan A. Lack, Jianfei Qi, Bo Han, Xuesen Dong
View: Text | PDF

Non-canonical functions of UGT2B17 promote castrate-resistant prostate cancer progression

  • Text
  • PDF
Abstract

Androgen deprivation therapy is the primary treatment for advanced prostate tumors. While initially effective, tumor progression to the therapy-resistant stage is inevitable. Paradoxically, UDP-glucuronosyltransferase 2B17 (UGT2B17), the key enzyme responsible for androgen catabolism in prostate tumor cells, is upregulated in therapy-resistant tumors, though its role in tumor progression remains unclear. Here, we demonstrate that UGT2B17 possesses multiple oncogenic functions independent of androgen catabolism. It modulates protein-folding pathways, allowing tumor cells to endure therapy-induced stress. UGT2B17 also regulates transcription associated with cell division and the DNA damage response, enabling unchecked cell proliferation. Targeting the newly identified UGT2B17 functions using a combination of inhibitors reduces tumor growth in therapy-resistant tumor models, highlighting a promising therapeutic strategy. Collectively, these findings reveal a mechanism by which prostate tumors exploit UGT2B17 to evade therapy and highlight its potential as a therapeutic target in advanced prostate cancer.

Authors

Tingting Feng, Ning Xie, Lin Gao, Qiongqiong Jia, Sonia Kung, Tunc Morova, Yinan Li, Lin Wang, Ladan Fazli, Louis Lacombe, Chantal Guillemette, Eric Lévesque, Nathan A. Lack, Jianfei Qi, Bo Han, Xuesen Dong

×

Oncogenic KRAS/ERK/JUNB signaling suppresses differentiation regulator GATA6 in pancreatic cancer
Zheng Zhong, Xinang Cao, Pei-Ju Liao, Raman Sethi, Jeffrey A. Klomp, Clint A. Stalnecker, Jinmiao Chen, Yue Wan, Channing J. Der, David M. Virshup
Zheng Zhong, Xinang Cao, Pei-Ju Liao, Raman Sethi, Jeffrey A. Klomp, Clint A. Stalnecker, Jinmiao Chen, Yue Wan, Channing J. Der, David M. Virshup
View: Text | PDF

Oncogenic KRAS/ERK/JUNB signaling suppresses differentiation regulator GATA6 in pancreatic cancer

  • Text
  • PDF
Abstract

GATA6 is a master regulator of differentiation in the pancreas and its expression levels determine the two main molecular subtypes of pancreatic cancer. High GATA6 contributes to the “classical” pancreatic cancer subtype, which is associated with a higher degree of tumor differentiation and better disease prognosis. However, why GATA6 expression varies across pancreatic cancers and what regulate GATA6 expression remain elusive. Here we report that the oncogenic KRAS-activated ERK signaling suppresses GATA6 transcription in pancreatic cancers. GATA6 mRNA levels inversely correlated with KRAS/ERK activity in pancreatic tumors. A genome-wide CRISPR screen in a GATA6-EGFP reporter knockin cell line identified JUNB as the ERK-regulated transcriptional repressor for GATA6. Active ERK stabilizes JUNB protein while KRAS/ERK inhibition led to ubiquitin-independent proteasomal degradation of JUNB and increased transcription of GATA6. Up-regulation of GATA6 enhanced chemosensitivity of pancreatic cancer cells and KRAS/ERK inhibitors synergized with chemotherapy in a GATA6-dependent manner. Our study identifies how oncogenic KRAS/ERK signaling suppresses GATA6 to cause dedifferentiation in pancreatic cancer. Combining KRAS/ERK inhibitors with standard-of-care chemotherapies could be a promising therapeutic strategy for treating pancreatic cancers.

Authors

Zheng Zhong, Xinang Cao, Pei-Ju Liao, Raman Sethi, Jeffrey A. Klomp, Clint A. Stalnecker, Jinmiao Chen, Yue Wan, Channing J. Der, David M. Virshup

×

LC3-dependent intercellular transfer of phosphorylated STAT1/2 elicits CXCL9+ macrophages and enhances radiation-induced antitumor immunity
Jun-Yan Li, Ying-Qing Li, Jia-Hao Dai, Sha Gong, Qing-Mei He, Jie-Wen Bai, Sai-Wei Huang, Ying-Qi Lu, Yu-Fei Duan, Sen-Yu Feng, Xi-Rong Tan, Xiao-Yu Liang, Jun Ma, Rui Guo, Na Liu
Jun-Yan Li, Ying-Qing Li, Jia-Hao Dai, Sha Gong, Qing-Mei He, Jie-Wen Bai, Sai-Wei Huang, Ying-Qi Lu, Yu-Fei Duan, Sen-Yu Feng, Xi-Rong Tan, Xiao-Yu Liang, Jun Ma, Rui Guo, Na Liu
View: Text | PDF

LC3-dependent intercellular transfer of phosphorylated STAT1/2 elicits CXCL9+ macrophages and enhances radiation-induced antitumor immunity

  • Text
  • PDF
Abstract

The efficacy of anticancer treatments, including radiotherapy, depends on the activation of type I IFN signaling. However, its regulatory networks and mechanisms remain to be elucidated. Here, we report that tumor cell–intrinsic type I IFN signaling can be transferred to macrophages via secretory autophagy, inducing CXCL9hi macrophages and enhancing CD8+ T cell–mediated antitumor immunity. Mechanistically, K63-linked ubiquitination at the K167 site of phosphorylated STAT2 (p-STAT2) facilitates its binding to LC3B, promoting the loading of p-STAT1 and p-STAT2 into extracellular vesicles and intercellular transference from tumor cells to macrophages, which, however, is suppressed by USP5-mediated STAT2 deubiquitination. Genetic depletion or pharmacological inhibition of USP5 promotes autophagy-dependent unconventional protein secretion of p-STAT1 and p-STAT2, leading to the induction of CXCL9+ macrophages. This process promotes the expression of T cell chemokines and upregulates the antigen presentation machinery, thereby enhancing radiation-induced CD8+ T cell antitumor immunity and radiotherapy efficacy. Our findings reveal a critical role of USP5 in type I IFN–induced antitumor immunity, providing potential targets for improving the efficacy of radiotherapy.

Authors

Jun-Yan Li, Ying-Qing Li, Jia-Hao Dai, Sha Gong, Qing-Mei He, Jie-Wen Bai, Sai-Wei Huang, Ying-Qi Lu, Yu-Fei Duan, Sen-Yu Feng, Xi-Rong Tan, Xiao-Yu Liang, Jun Ma, Rui Guo, Na Liu

×

CAR-T-cells targeting the glycoprotein GD2 show potent anti-tumor efficacy in high-risk ependymoma models
Antonio Carlos Tallon-Cobos, Konstantinos Vazaios, Piotr Waranecki, Marliek van Hoesel, Annelisa M. Cornel, Benjamin Schwalm, Norman Mack, Ella de Boed, Jasper van der Lugt, Stefan Nierkens, Marcel Kool, Eelco W. Hoving, Dennis S. Metselaar, Esther Hulleman
Antonio Carlos Tallon-Cobos, Konstantinos Vazaios, Piotr Waranecki, Marliek van Hoesel, Annelisa M. Cornel, Benjamin Schwalm, Norman Mack, Ella de Boed, Jasper van der Lugt, Stefan Nierkens, Marcel Kool, Eelco W. Hoving, Dennis S. Metselaar, Esther Hulleman
View: Text | PDF

CAR-T-cells targeting the glycoprotein GD2 show potent anti-tumor efficacy in high-risk ependymoma models

  • Text
  • PDF
Abstract

Authors

Antonio Carlos Tallon-Cobos, Konstantinos Vazaios, Piotr Waranecki, Marliek van Hoesel, Annelisa M. Cornel, Benjamin Schwalm, Norman Mack, Ella de Boed, Jasper van der Lugt, Stefan Nierkens, Marcel Kool, Eelco W. Hoving, Dennis S. Metselaar, Esther Hulleman

×

Estrogen Receptor signaling drives immune evasion and immunotherapy resistance in HR+ breast cancer
José Ángel Palomeque, Gabriel Serra-Mir, Sandra Blasco-Benito, Helena Brunel, Pau Torren-Duran, Iván Pérez-Núñez, Chiara Cannatá, Laura Comerma, Silvia Menendez, Sonia Servitja, Tamara Martos, Maria Castro, Rodrigo L. Borges, Joanna I. Lopez-Velazco, Sara Manzano, Santiago Duro-Sánchez, Joaquin Arribas, Maria M. Caffarel, Ander Urruticoechea, Jose A. Seoane, Lluis Morey, Joan Albanell, Toni Celià-Terrassa
José Ángel Palomeque, Gabriel Serra-Mir, Sandra Blasco-Benito, Helena Brunel, Pau Torren-Duran, Iván Pérez-Núñez, Chiara Cannatá, Laura Comerma, Silvia Menendez, Sonia Servitja, Tamara Martos, Maria Castro, Rodrigo L. Borges, Joanna I. Lopez-Velazco, Sara Manzano, Santiago Duro-Sánchez, Joaquin Arribas, Maria M. Caffarel, Ander Urruticoechea, Jose A. Seoane, Lluis Morey, Joan Albanell, Toni Celià-Terrassa
View: Text | PDF

Estrogen Receptor signaling drives immune evasion and immunotherapy resistance in HR+ breast cancer

  • Text
  • PDF
Abstract

Hormone Receptor positive (HR+) breast cancers (BC) are typically “immune-cold” poorly immune infiltrated tumors that do not respond to immune-checkpoint blockade (ICB) therapies. Using clinical data, we report that estrogen receptor (ERα) signaling associates with immunosuppressive pathways and lack of response to ICB in HR+ patients. In this study, we validate ER-mediated immunosuppression by engineering and modulating ER in preclinical models in vitro, in vivo and ex vivo. Mechanistically, we found that ERα hijacks LCOR, a nuclear receptor corepressor, thereby preventing LCOR’s function in the induction of tumor immunogenicity and immune infiltration, which is normally observed in the absence of ERα, such as in ER-negative BC. In HR+BC, we demonstrate that the molecular disruption of LCOR and ERα interaction using anti-ER therapies or using a mutant of the LCOR nuclear-receptor binding domain (LSKLL into LSKAA) that does not interact with ERα, restores LCOR’s immunogenic functions. Remarkably, the LCOR-ERα disruption converts HR+BC immune-cold tumors into immune-hot tumors responsive to ICB by increased antigen presentation machinery (APM) expression, immune infiltration, T cell recognition and mediated killing. In conclusion, ERα inhibition and the disruption of LCOR to ERα represent a novel therapeutic strategy and an opportunity to elicit immunotherapeutic benefit in HR+BC patients.

Authors

José Ángel Palomeque, Gabriel Serra-Mir, Sandra Blasco-Benito, Helena Brunel, Pau Torren-Duran, Iván Pérez-Núñez, Chiara Cannatá, Laura Comerma, Silvia Menendez, Sonia Servitja, Tamara Martos, Maria Castro, Rodrigo L. Borges, Joanna I. Lopez-Velazco, Sara Manzano, Santiago Duro-Sánchez, Joaquin Arribas, Maria M. Caffarel, Ander Urruticoechea, Jose A. Seoane, Lluis Morey, Joan Albanell, Toni Celià-Terrassa

×

Pancreatic volume and immune biomarkers predict checkpoint inhibitor-associated autoimmune diabetes in humans
Linda Wu, John M. Wentworth, Christopher Liddle, Nicole Fewings, Matteo Carlino, David A. Brown, Roderick Clifton-Bligh, Georgina V. Long, Richard A. Scolyer, Nicholas Norris, Sarah C. Sasson, Venessa H.M. Tsang, Alexander M. Menzies, Jenny E. Gunton
Linda Wu, John M. Wentworth, Christopher Liddle, Nicole Fewings, Matteo Carlino, David A. Brown, Roderick Clifton-Bligh, Georgina V. Long, Richard A. Scolyer, Nicholas Norris, Sarah C. Sasson, Venessa H.M. Tsang, Alexander M. Menzies, Jenny E. Gunton
View: Text | PDF

Pancreatic volume and immune biomarkers predict checkpoint inhibitor-associated autoimmune diabetes in humans

  • Text
  • PDF
Abstract

BACKGROUND. Checkpoint inhibitor-associated autoimmune diabetes (CIADM) is a rare but life-altering complication of immune checkpoint inhibitor (ICI) therapy. Biomarkers that predict type 1 diabetes (T1D) are unreliable for CIADM. AIM. To identify biomarkers for prediction of CIADM. METHODS. From our prospective biobank, 14 CIADM patients who had metastatic melanoma treated with anti-PD-1 ± anti-CTLA4 were identified. Controls were selected from the same biobank, matched 2:1. Pre-treatment, on-ICI and post-CIADM serum and peripheral blood mononuclear cells (PBMCs) were analysed. Serum was analysed for T1D autoantibodies, C-peptide, glucose and cytokines. PBMCs were profiled using flow cytometry. Pancreatic volume was measured using CT volumetry. RESUTLS. Before treatment, CIADM patients had smaller pancreatic volume (27% reduction, p=0.044) and higher anti-GAD antibody titres (median 2.9 versus 0, p=0.01). They had significantly higher baseline proportions of Th17 helper cells (p=0.03), higher CD4+ central memory cells (p=0.04) and lower naïve CD4+ cells (p=0.01). With ICI treatment, greater declines in pancreatic volume were seen in CIADM patients (p<0.0001). Activated CD4+ subsets increased significantly in CIADM and controls with immune-related adverse effects (IRAE) but not controls without IRAE. Using only pre-treatment results, pancreatic volume, anti-GAD antibody titre and baseline immune flow profile were highly predictive of CIADM development, with an area under the curve (AUC) of >0.96. CONCLUSIONS. People who develop CIADM are immunologically predisposed and have antecedent pancreatic and immunological changes that accurately predict disease with excellent sensitivity. These biomarkers could be used to guide ICI use, particularly when planning treatment for low-risk tumours. FUNDING. JEG is supported by NHMRC Investigator grant 2033228. AMM by NHMRC Investigator grant 2009476 and GVL by NHMRC Investigator grant 2007839.

Authors

Linda Wu, John M. Wentworth, Christopher Liddle, Nicole Fewings, Matteo Carlino, David A. Brown, Roderick Clifton-Bligh, Georgina V. Long, Richard A. Scolyer, Nicholas Norris, Sarah C. Sasson, Venessa H.M. Tsang, Alexander M. Menzies, Jenny E. Gunton

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 138
  • 139
  • Next →
  • ← Previous
  • 1
  • 2
  • Next →
Attack of the WNT inducing macrophages
Luke Boulter and colleagues reveal that WNT ligand-secreting macrophages promote cholangiocarcinoma proliferation….
Published February 17, 2015
Scientific Show StopperOncology

Metabolic reprogramming in prostate cancer
Subhamoy Dasgupta and colleagues reveal that steroid receptor coactivator 2 mediates metabolic reprogramming that supports growth and survival of prostate cancer cells….
Published February 9, 2015
Scientific Show StopperOncology

NOTCH keeps bladder cancer at bay
Antonio Maraver, Pablo Fernandez-Marcos, and colleagues demonstrate the NOTCH serves as a tumor suppressor in the bladder…
Published January 9, 2015
Scientific Show StopperOncology

T cell signals substitute for EBV oncoprotein
Shi-Dong Ma and colleagues develop a human cord blood-based model of EBV-induced B cell lymphoma to evaluate the requirement of EBV oncoprotein LMP1 in pathogenesis…
Published December 8, 2014
Scientific Show StopperOncology
  • ← Previous
  • 1
  • 2
  • Next →
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts