1
|
Zhou J, Tison K, Zhou H, Bai L, Acharyya RK, McEachern D, Metwally H, Wang Y, Pitter M, Choi JE, Vatan L, Liao P, Yu J, Lin H, Jiang L, Wei S, Gao X, Grove S, Parolia A, Cieslik M, Kryczek I, Green MD, Lin JX, Chinnaiyan AM, Leonard WJ, Wang S, Zou W. STAT5 and STAT3 balance shapes dendritic cell function and tumour immunity. Nature 2025:10.1038/s41586-025-09000-3. [PMID: 40369063 DOI: 10.1038/s41586-025-09000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Immune checkpoint blockade (ICB) has transformed cancer therapy1,2. The efficacy of immunotherapy depends on dendritic cell-mediated tumour antigen presentation, T cell priming and activation3,4. However, the relationship between the key transcription factors in dendritic cells and ICB efficacy remains unknown. Here we found that ICB reprograms the interplay between the STAT3 and STAT5 transcriptional pathways in dendritic cells, thereby activating T cell immunity and enabling ICB efficacy. Mechanistically, STAT3 restrained the JAK2 and STAT5 transcriptional pathway, determining the fate of dendritic cell function. As STAT3 is often activated in the tumour microenvironment5, we developed two distinct PROTAC (proteolysis-targeting chimera) degraders of STAT3, SD-36 and SD-2301. STAT3 degraders effectively degraded STAT3 in dendritic cells and reprogrammed the dendritic cell-transcriptional network towards immunogenicity. Furthermore, STAT3 degrader monotherapy was efficacious in treatment of advanced tumours and ICB-resistant tumours without toxicity in mice. Thus, the crosstalk between STAT3 and STAT5 transcriptional pathways determines the dendritic cell phenotype in the tumour microenvironment and STAT3 degraders hold promise for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Kole Tison
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Haibin Zhou
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Longchuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ranjan Kumar Acharyya
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Donna McEachern
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hoda Metwally
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yu Wang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Pitter
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jae Eun Choi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Long Jiang
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xue Gao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Michael D Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shaomeng Wang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA.
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Yang Y, Wang TY, Li Q, Lu J, Ren Y, Weiner AB, Fry J, Liu Q, Yum C, Wang R, Guo Q, Wan Y, Ji Z, Dong X, Lotan TL, Schaeffer EM, Yang R, Cao Q. Androgen receptor-regulated lncRNA PRCAT71 promotes AR signaling through the interaction with KHSRP in prostate cancer. SCIENCE ADVANCES 2025; 11:eadk6989. [PMID: 40203114 PMCID: PMC11980854 DOI: 10.1126/sciadv.adk6989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Mounting evidence indicates that long noncoding RNAs (lncRNAs) play vital roles in tumorigenesis and progression of cancers. However, the functions and regulatory mechanisms of lncRNAs in prostate cancer (PCa) are still largely unknown. In this study, we found an lncRNA, PCa-associated transcript 71 (PRCAT71), highly expressed in metastatic and primary PCa compared to benign prostate tissues. Silencing PRCAT71 inhibited cancerous properties of PCa cells and androgen receptor (AR) signaling. Mechanistically, PRCAT71 acts as a scaffold to recruit K homology (KH)-type splicing regulatory protein (KHSRP) to AR messenger RNA (mRNA) and stabilize AR mRNA, leading to activated AR signaling. KHSRP plays a critical role in PCa progression. PRCAT71 is transcriptionally regulated by AR-driven enhancers, forming a positive regulatory loop between AR and PRCAT71 in PCa. Our study demonstrates a coordinated regulation of AR mRNA by lncRNA PRCAT71 and RNA binding protein KHSRP and provides insight that the PRCAT71-KHSRP-AR axis is a promising therapeutic target for treating PCa.
Collapse
Affiliation(s)
- Yongyong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ting-You Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qianru Li
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jiawen Lu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yanan Ren
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam B. Weiner
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua Fry
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Liu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chaehyun Yum
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rui Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qingxiang Guo
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yu Wan
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Zhe Ji
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Edward M. Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Cao
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
3
|
García-Heredia A, Guerra-Núñez L, Martín-Climent P, Rojas E, López-Domínguez R, Alcántara-Domínguez C, Alenda C, Valor LM. Transcriptomics and epigenomics datasets of primary brain cancers in formalin-fixed paraffin embedded format. Sci Data 2025; 12:273. [PMID: 39955294 PMCID: PMC11830079 DOI: 10.1038/s41597-025-04597-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
The access of public omics-based datasets is of paramount importance in brain cancer research as allows the proposal and validation of both biomarkers and therapeutic targets in gliomas, especially in the most prevalent and aggressive glioblastomas. Taking profit of current advances in next generation sequencing and DNA methylation profiling, we have created datasets from approximately 150 formalin-fixed paraffin embedded (FFPE) tumours. These datasets enable for the first time integrative transcriptional and epigenetics studies in a context that consider the degradation and fixation-derived chemical alterations of the most extended archiving format in hospitals, and provide an independent cohort from current public databases for further validation of putative novel biomarkers. Alongside with the most profusely known glioblastomas, astrocytomas and oligodendrogliomas, we have also included for comparison purposes few examples of rare tumours that are often neglected in brain cancer research. Taken together, we provide a valuable tool to explore combined gene expression and DNA methylation patterns in the study of gliomas and glioneuronal tumours.
Collapse
Affiliation(s)
- Anabel García-Heredia
- Laboratorio de Investigación, Centro de Diagnóstico, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Luna Guerra-Núñez
- Laboratorio de Investigación, Centro de Diagnóstico, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Paula Martín-Climent
- Laboratorio de Investigación, Centro de Diagnóstico, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Estefanía Rojas
- Departamento de Patología, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Raúl López-Domínguez
- Centro Pfizer - Universidad de Granada - Junta de Andalucía de Genómica e Investigación Oncológica (GENYO), 18016, Granada, Spain
| | - Clara Alcántara-Domínguez
- Centro Pfizer - Universidad de Granada - Junta de Andalucía de Genómica e Investigación Oncológica (GENYO), 18016, Granada, Spain
| | - Cristina Alenda
- Departamento de Patología, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Luis M Valor
- Laboratorio de Investigación, Centro de Diagnóstico, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), 03202, Elche, Spain.
| |
Collapse
|
4
|
Göcz B, Rumpler É, Szentkirályi-Tóth S, Skrapits K, Takács S, Sárvári M, Farkas I, Póliska S, Hrabovszky E. Laser-capture microdissection for spatial transcriptomics of immunohistochemically detected neurons. J Biol Chem 2025; 301:108150. [PMID: 39736395 PMCID: PMC11910328 DOI: 10.1016/j.jbc.2024.108150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/29/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025] Open
Abstract
We developed a versatile 'IHC/LCM-Seq' method for spatial transcriptomics of immunohistochemically detected neurons collected with laser-capture microdissection (LCM). IHC/LCM-Seq uses aluminon and polyvinyl sulfonic acid for inventive RNA-preserving strategies to maintain RNA integrity in free-floating sections of 4% formaldehyde-fixed brains. To validate IHC/LCM-Seq, we first immunostained and harvested striatal cholinergic interneurons with LCM. RNA preparations were subjected to random primer-based cDNA library preparation and bulk sequencing on the NextSeq Illumina platform. IHC/LCM-Seq detected ∼16,000 transcripts, reaching the sensitivity of a reference 'LCM-Seq method' developed for fluorescently tagged neurons microdissected from lightly formaldehyde-fixed and slide-mounted brain sections of transgenic mice. We successfully used the new IHC/LCM-Seq approach to provide unprecedented insight into the transcriptome of immunohistochemically detected gonadotropin-releasing hormone (GnRH) neurons regulating reproduction. The ∼13,000 to 14,000 transcripts identified in GnRH neurons of adult male rats and mice encoded 28 proteins implicated previously in human infertility, 35 neuropeptides, 34 nuclear receptors, and 164 G protein-coupled receptors. Functional experiments using slice electrophysiology established that the heavy Ntsr2 expression conveys a strong excitatory action of neurotensin on GnRH neurons. As an unexpected species difference, we found that GnRH neurons exclusively expressed estrogen receptor-β in rats and against the current consensus, the alpha estrogen receptor isoform in mice. The IHC/LCM-Seq technique we are reporting is a highly sensitive and accurate bulk sequencing approach to characterize the transcriptome landscape of immunohistochemically labeled neurons, including neuroendocrine GnRH cells. This method is readily applicable to any species, opening new perspectives also for future studies of the post mortem human brain.
Collapse
Affiliation(s)
- Balázs Göcz
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| | - Soma Szentkirályi-Tóth
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
5
|
McAfee JL, Alban TJ, Makarov V, Rupani A, Parthasarathy PB, Tu Z, Ronen S, Billings SD, Diaz CM, Chan TA, Ko JS. Genomic Landscape of Superficial Malignant Peripheral Nerve Sheath Tumor. J Transl Med 2025; 105:102183. [PMID: 39532239 DOI: 10.1016/j.labinv.2024.102183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Superficial malignant peripheral nerve sheath tumors (SF-MPNSTs) are rare cancers and can be difficult to distinguish from spindle cell (SCM) or desmoplastic (DM) melanomas. Their biology is poorly understood. We performed whole-exome sequencing and RNA sequencing (RNA-seq) on SF-MPNST (n = 8) and compared them with cases of SCM (n = 7), DM (n = 8), and deep MPNST (D-MPNST, n = 8). Immunohistochemical staining for H3K27me3 and PRAME was also performed. SF-MPNST demonstrated intermediate features between D-MPNST and melanoma. Patients were younger than those with melanoma and older than those with D-MPNST; the outcome was worse and better, respectively. SF-MPNST tumor mutational burden (TMB) was higher than D-MPNST and lower than melanoma; differences were significant only between SF-MPNST and SCM (P = .0454) and between D-MPNST and SCM (P = .001, Dunn's Kruskal-Wallis post hoc test). Despite having an overlapping mutational profile in some common cancer-associated genes, the COSMIC mutational signatures clustered DM and SCM together with UV light exposure signatures (SBS7a, 7b), and SF- and D-MPNST together with defective DNA base excision repair (SBS30, 36). RNA-seq revealed differentially expressed genes between SF-MPNST and SCM (1670 genes), DM (831 genes), and D-MPNST (614 genes), some of which hold promise for development as immunohistochemical markers (SOX8 and PLCH1) or aids (MLPH, CALB2, SOX11, and TBX4). H3K27me3 immunoreactivity was diffusely lost in most D-MPNSTs (7/8, 88%) but showed variable and patchy loss in SF-MPNSTs (2/8, 25%). PRAME was entirely negative in the majority (0+ in 20/31, 65%), including 11/15 melanomas, and showed no significant difference between groups (P = .105, Kruskal-Wallis test). Expression of immune cell transcripts was upregulated in melanomas relative to MPNSTs. Next-generation sequencing revealed multiple differential features between SF- MPNST, D-MPNST, SCM, and DM, including tumor mutation burden, mutational signatures, and differentially expressed genes. These findings help advance our understanding of disease pathogenesis and improve diagnostic modalities.
Collapse
Affiliation(s)
- John L McAfee
- Department of Pathology, Cleveland Clinic Pathology and Laboratory Medicine Institute, Cleveland, Ohio
| | - Tyler J Alban
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amit Rupani
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Prerana B Parthasarathy
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Zheng Tu
- Department of Pathology, Cleveland Clinic Pathology and Laboratory Medicine Institute, Cleveland, Ohio
| | - Shira Ronen
- Department of Pathology, Cleveland Clinic Pathology and Laboratory Medicine Institute, Cleveland, Ohio
| | - Steven D Billings
- Department of Pathology, Cleveland Clinic Pathology and Laboratory Medicine Institute, Cleveland, Ohio
| | - C Marcela Diaz
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; National Center for Regenerative Medicine, Cleveland, Ohio
| | - Jennifer S Ko
- Department of Pathology, Cleveland Clinic Pathology and Laboratory Medicine Institute, Cleveland, Ohio; Center for Immunotherapy and Precision Immuno-Oncology and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
6
|
Gibbs DL, Cioffi G, Aguilar B, Waite KA, Pan E, Mandel J, Umemura Y, Luo J, Rubin JB, Pot D, Barnholtz-Sloan J. Robust Cluster Prediction Across Data Types Validates Association of Sex and Therapy Response in GBM. Cancers (Basel) 2025; 17:445. [PMID: 39941811 PMCID: PMC11815886 DOI: 10.3390/cancers17030445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Previous studies have described sex-specific patient subtyping in glioblastoma. The cluster labels associated with these "legacy data" were used to train a predictive model capable of recapitulating this clustering in contemporary contexts. METHODS We used robust ensemble machine learning to train a model using gene microarray data to perform multi-platform predictions including RNA-seq and potentially scRNA-seq. RESULTS The engineered feature set was composed of many previously reported genes that are associated with patient prognosis. Interestingly, these well-known genes formed a predictive signature only for female patients, and the application of the predictive signature to male patients produced unexpected results. CONCLUSIONS This work demonstrates how annotated "legacy data" can be used to build robust predictive models capable of multi-target predictions across multiple platforms.
Collapse
Affiliation(s)
- David L. Gibbs
- Thorsson-Shmulevich Lab, Institute of Systems Biology, Seattle, WA 98109, USA
| | - Gino Cioffi
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
| | - Boris Aguilar
- Thorsson-Shmulevich Lab, Institute of Systems Biology, Seattle, WA 98109, USA
| | - Kristin A. Waite
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
| | - Edward Pan
- Global Oncology Research & Development, Daiichi-Sankyo, Inc., Basking Ridge, NJ 07920, USA
| | - Jacob Mandel
- Department of Neurology and Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yoshie Umemura
- IVY Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jingqin Luo
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center Biostatistics and Qualitative Research Shared Resource, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua B. Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Pot
- General Dynamics Information Technology, Falls Church, VA 22042, USA
| | - Jill Barnholtz-Sloan
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Gallardo-Gómez M. Analysis of the Pattern of RNA Expression in the Skin of TR-Deficient Mice By RNA-seq. Methods Mol Biol 2025; 2876:151-162. [PMID: 39579314 DOI: 10.1007/978-1-0716-4252-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
RNA-sequencing (RNA-seq) has become the method of choice for whole transcriptome analysis, as it enables profiling of a wide range of RNA molecules and the always evolving bioinformatic pipelines allow the extraction of diverse biological information from a single sample. However, there is not a gold-standard RNA-seq protocol nor an optimal bioinformatic pipeline defined, which can challenge new users. The aim of this chapter is to describe the basic RNA-sequencing pipeline, from RNA extraction and library preparation to the bioinformatic preprocessing and downstream analyses. The steps are oriented to the transcriptome analysis of the skin of TR-deficient mice, but the protocol can be easily translated to other tissue types or organisms.
Collapse
Affiliation(s)
- María Gallardo-Gómez
- Translational Oncology Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
8
|
Heath EI, Chen W, Choi JE, Dobson K, Smith M, Maj T, Kryczek I, Zou W, Chinnaiyan AM, Qiao Y. Phase II trial of multi-tyrosine kinase inhibitor ESK981 in combination with PD-1 inhibitor nivolumab in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2024; 42:675-684. [PMID: 39503807 DOI: 10.1007/s10637-024-01482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/23/2024] [Indexed: 12/08/2024]
Abstract
Increasing the response rates of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer (mCRPC) presents a significant challenge. ESK981 is a multi-tyrosine kinase and PIKfyve lipid kinase inhibitor that augments immunotherapeutic responses. In this phase II study, ESK981 was combined with the PD-1 blocking monoclonal antibody nivolumab to test for potentially improved response rates in patients with mCRPC who have progressed on androgen receptor (AR)-targeted agents and chemotherapy. Eligible patients received ESK981 orally once daily for five consecutive days, followed by a two-day break. Patients were also treated with nivolumab intravenously on Day 1 of each 28-day cycle. The primary endpoints were a 50% reduction in prostate-specific antigen (PSA50), and safety. Secondary endpoints included radiographic progression free survival (rPFS) and overall survival (OS). Additional investigations included whole exome sequencing in patients. Ten patients were enrolled. The maximum PSA decline from baseline of 14% was achieved in only one patient. Grade 3 treatment-related adverse events (AEs) included fatigue, anemia, and lymphopenia. There were no Grade 4 events. The median rPFS was 3.7 months (95% CI, 1.6-8.4). The median OS was 9.6 months (95% CI, 1.8-22.4). The study was terminated due to futility after 10 patients. Whole exome sequencing identified AR amplification in 63% of patients (5/8). ESK981 + nivolumab showed no antitumor activity in patients with AR-positive (AR+) mCRPC. Further evaluation of ESK981 combined with the PD-1 inhibitor nivolumab in AR + mCRPC patients is not warranted. (Trial registration: ClinicalTrials.gov NCT04159896. Registration date: November 12, 2019.).
Collapse
Affiliation(s)
- Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Wei Chen
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jae E Choi
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kimberlee Dobson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Melanie Smith
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Li J, Tang HT, Liu Q, Li CH, Chen WY, Yu ZW, Wang L, Lin L, Zhao JL, Zhao CY, Chen LQ, Tian D. Case report: A case of giant malignant solitary fibrous tumor of the pleura with Doege-Potter's syndrome and review of the literature. Front Oncol 2024; 14:1437535. [PMID: 39678506 PMCID: PMC11638045 DOI: 10.3389/fonc.2024.1437535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
The solitary fibrous tumor of the pleura (SFTP) is a rare intrathoracic neoplasm that commonly originates from the subpleural mesenchymal cells of the visceral pleura and accounts for less than 5% of all pleural tumors. We reported a case of a 54-year-old man with a two-week history of hypoglycemia, a six-month history of productive cough and fatigue, and chronic right chest pain. Radiological techniques revealed a giant intra-thoracic mass with hypervascularization, and pathological staining was carried out to make a definitive diagnosis of SFTP. Interventional embolization was conducted to block the main feeding vessels before the surgery, and an anterolateral thoracotomy combined with a transverse sternotomy was performed to achieve a complete resection, which demonstrates significant potential for further application in patients with unilateral giant SFTP. The postoperative course was uneventful, with no signs of hypoglycemia observed during the follow-up. Additionally, we reviewed and prospected the research progress on SFTP. The aim of this study is to enhance clinicians' understanding of SFTP through our case and to provide a detailed review of the current research.
Collapse
Affiliation(s)
- Jie Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hong-Tao Tang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Integrated Care Management Center, West China Hospital, Sichuan University, Chengdu, China
| | - Cai-Han Li
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wei-Yang Chen
- Department of Thoracic Surgery, The First Hospital of China Medical University, China Medical University, Shenyang, China
| | - Zeng-Wei Yu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jin-Lan Zhao
- Anesthesia Surgery Center, West China Hospital, Sichuan University/West China School of Nursing, Shenyang, China
| | - Chun-Yan Zhao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Long-Qi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Heath EI, Chen W, Heilbrun L, Choi JE, Dobson K, Smith M, Maj T, Vaishampayan U, Kryczek I, Zou W, Chinnaiyan AM, Qiao Y. Phase II trial of multi-kinase inhibitor ESK981 in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2024; 42:566-574. [PMID: 39227508 PMCID: PMC11756588 DOI: 10.1007/s10637-024-01463-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
ESK981 is a potent tyrosine kinase and PIKfyve lipid kinase inhibitor. This phase II trial evaluated the efficacy of ESK981 as a single agent in patients with androgen receptor-positive (AR +) metastatic castration-resistant prostate cancer (mCRPC). Eligible patients had mCRPC with progression on AR-targeted agents and without prior chemotherapy treatment. Each patient received 160 mg ESK981 once daily for 5 days per week for 4 weeks per cycle (except for an adverse event (AE) occurrence). The primary endpoints were a 50% reduction in prostate-specific antigen (PSA50), and safety. Secondary endpoints included the time and the duration of PSA response, PSA progression rates, PSA progression free survival (PFS) and overall survival (OS). Exploratory investigations included whole exome sequencing in patients before treatment, and morphological evaluation of biopsy samples pre- and post-treatment. PSA was evaluated in 13 patients. Only one patient (7.7% two-sided 95% Wilson CI (0.4%, 33.3%)) experienced a reduction in their PSA levels by 50% or more. The most common grade 3 treatment-related AEs were cardiac disorders, diarrhea, hypertension, alanine transaminase and aspartate transaminase elevations. No grade 4-5 events occurred. Median PFS was 1.8 months, and median OS was 12.1 months. Peripheral immune cells showed increased T cell activation and cytokine production in two patients who received 12-weeks of ESK981. Although relatively well tolerated, ESK981 alone showed no anti-tumor activity in patients with AR + mCRPC and its further evaluation as a single agent in AR + mCRPC is not warranted. (Trial registration: ClinicalTrials.gov, NCT03456804. Registration date: March 7, 2018).
Collapse
Affiliation(s)
- Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Wei Chen
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lance Heilbrun
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jae E Choi
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kimberlee Dobson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Melanie Smith
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Nguyen CB, Reimers MA, Perera C, Abida W, Chou J, Feng FY, Antonarakis ES, McKay RR, Pachynski RK, Zhang J, Reichert ZR, Palmbos PL, Caram ME, Vaishampayan UN, Heath EI, Hopkins AC, Cieslik MP, Wu YM, Robinson D, Baladandayuthapani V, Chinnaiyan AM, Alva AS. Evaluating Immune Checkpoint Blockade in Metastatic Castration-Resistant Prostate Cancers with Deleterious CDK12 Alterations in the Phase 2 IMPACT Trial. Clin Cancer Res 2024; 30:3200-3210. [PMID: 38787530 PMCID: PMC11293970 DOI: 10.1158/1078-0432.ccr-24-0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/08/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE CDK12 inactivation in metastatic castration-resistant prostate cancer (mCRPC) may predict immunotherapy responses. This phase 2 trial evaluated the efficacy of immune checkpoint inhibitor (ICI) therapy in patients with CDK12-altered mCRPC. PATIENTS AND METHODS Eligible patients had mCRPC with deleterious CDK12 alterations and any prior therapies except ICI. Cohort A received ipilimumab (1 mg/kg) with nivolumab (3 mg/kg) every 3 weeks for up to four cycles, followed by nivolumab 480 mg every 4 weeks. Cohort C received nivolumab alone 480 mg every 4 weeks. Patients with CDK12-altered nonprostate tumors were enrolled in cohort B and not reported. The primary endpoint was a 50% reduction in PSA (PSA50). Key secondary endpoints included PSA progression-free survival, overall survival, objective response rate, and safety. RESULTS PSA was evaluable in 23 patients in cohort A and 14 in cohort C. Median lines of prior therapy were two in cohorts A and C, including any prior novel hormonal agent (74% and 79%) and chemotherapy (57% and 36%). The PSA50 rate was 9% [95% confidence interval (CI), 1%-28%] in cohort A with two responders; neither had microsatellite instability or a tumor mutational burden >10 mutations/megabase. No PSA50 responses occurred in cohort C. Median PSA progression-free survival was 7.0 months (95% CI, 3.6-11.4) in cohort A and 4.5 months (95% CI, 3.4-13.8) in cohort C. Median overall survival was 9.0 months (95% CI, 6.2-12.3) in cohort A and 13.8 months (95% CI, 3.6-not reached) in cohort C. CONCLUSIONS There was minimal activity with ICI therapy in patients with CDK12-altered mCRPC.
Collapse
Affiliation(s)
- Charles B. Nguyen
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| | | | - Chamila Perera
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonathan Chou
- Diller Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA
| | - Felix Y. Feng
- Diller Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA
| | | | - Rana R. McKay
- Moores Cancer Center, University of California San Diego, San Diego, CA
| | | | | | | | - Phillip L. Palmbos
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| | - Megan E.V. Caram
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| | | | | | - Alexander C. Hopkins
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Marcin P. Cieslik
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Dan Robinson
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Ajjai S. Alva
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
12
|
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JCY, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, Chinnaiyan AM. PIKfyve controls dendritic cell function and tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582543. [PMID: 38464258 PMCID: PMC10925294 DOI: 10.1101/2024.02.28.582543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The modern armamentarium for cancer treatment includes immunotherapy and targeted therapy, such as protein kinase inhibitors. However, the mechanisms that allow cancer-targeting drugs to effectively mobilize dendritic cells (DCs) and affect immunotherapy are poorly understood. Here, we report that among shared gene targets of clinically relevant protein kinase inhibitors, high PIKFYVE expression was least predictive of complete response in patients who received immune checkpoint blockade (ICB). In immune cells, high PIKFYVE expression in DCs was associated with worse response to ICB. Genetic and pharmacological studies demonstrated that PIKfyve ablation enhanced DC function via selectively altering the alternate/non-canonical NF-κB pathway. Both loss of Pikfyve in DCs and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively controls DCs, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
Collapse
Affiliation(s)
- Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Gurkan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yi Bao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mahnoor Gondal
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sahr Yazdani
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - JiaJia Zhou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Gaopeng Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rui Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael D. Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JCY, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, Chinnaiyan AM. PIKfyve, expressed by CD11c-positive cells, controls tumor immunity. Nat Commun 2024; 15:5487. [PMID: 38942798 PMCID: PMC11213953 DOI: 10.1038/s41467-024-48931-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 05/15/2024] [Indexed: 06/30/2024] Open
Abstract
Cancer treatment continues to shift from utilizing traditional therapies to targeted ones, such as protein kinase inhibitors and immunotherapy. Mobilizing dendritic cells (DC) and other myeloid cells with antigen presenting and cancer cell killing capacities is an attractive but not fully exploited approach. Here, we show that PIKFYVE is a shared gene target of clinically relevant protein kinase inhibitors and high expression of this gene in DCs is associated with poor patient response to immune checkpoint blockade (ICB) therapy. Genetic and pharmacological studies demonstrate that PIKfyve ablation enhances the function of CD11c+ cells (predominantly dendritic cells) via selectively altering the non-canonical NF-κB pathway. Both loss of Pikfyve in CD11c+ cells and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively regulates the function of CD11c+ cells, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
Collapse
Affiliation(s)
- Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Yuanyuan Qiao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Gurkan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi Bao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mahnoor Gondal
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sahr Yazdani
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - JiaJia Zhou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Gaopeng Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rui Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael D Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA.
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Ura H, Niida Y. Comparison of RNA-Sequencing Methods for Degraded RNA. Int J Mol Sci 2024; 25:6143. [PMID: 38892331 PMCID: PMC11172666 DOI: 10.3390/ijms25116143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
RNA sequencing (RNA-Seq) is a powerful technique and is increasingly being used in clinical research and drug development. Currently, several RNA-Seq methods have been developed. However, the relative advantage of each method for degraded RNA and low-input RNA, such as RNA samples collected in the field of clinical setting, has remained unknown. The Standard method of RNA-Seq captures mRNA by poly(A) capturing using Oligo dT beads, which is not suitable for degraded RNA. Here, we used three commercially available RNA-Seq library preparation kits (SMART-Seq, xGen Broad-range, and RamDA-Seq) using random primer instead of Oligo dT beads. To evaluate the performance of these methods, we compared the correlation, the number of detected expressing genes, and the expression levels with the Standard RNA-Seq method. Although the performance of RamDA-Seq was similar to that of Standard RNA-Seq, the performance for low-input RNA and degraded RNA has decreased. The performance of SMART-Seq was better than xGen and RamDA-Seq in low-input RNA and degraded RNA. Furthermore, the depletion of ribosomal RNA (rRNA) improved the performance of SMART-Seq and xGen due to increased expression levels. SMART-Seq with rRNA depletion has relative advantages for RNA-Seq using low-input and degraded RNA.
Collapse
Affiliation(s)
- Hiroki Ura
- Center for Clinical Genomics, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku 920-0923, Japan;
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0923, Japan
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku 920-0923, Japan;
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0923, Japan
| |
Collapse
|
15
|
Kandarpa M, Robinson D, Wu YM, Qin T, Pettit K, Li Q, Luker G, Sartor M, Chinnaiyan A, Talpaz M. Broad Next-Generation Integrated Sequencing of Myelofibrosis Identifies Disease-Specific and Age-Related Genomic Alterations. Clin Cancer Res 2024; 30:1972-1983. [PMID: 38386293 PMCID: PMC11061602 DOI: 10.1158/1078-0432.ccr-23-0372] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/18/2023] [Accepted: 02/20/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE Myeloproliferative neoplasms (MPN) are characterized by the overproduction of differentiated myeloid cells. Mutations in JAK2, CALR, and MPL are considered drivers of Bcr-Abl-ve MPN, including essential thrombocythemia (ET), polycythemia vera (PV), prefibrotic primary myelofibrosis (prePMF), and overt myelofibrosis (MF). However, how these driver mutations lead to phenotypically distinct and/or overlapping diseases is unclear. EXPERIMENTAL DESIGN To compare the genetic landscape of MF to ET/PV/PrePMF, we sequenced 1,711 genes for mutations along with whole transcriptome RNA sequencing of 137 patients with MPN. RESULTS In addition to driver mutations, 234 and 74 genes were found to be mutated in overt MF (N = 106) and ET/PV/PrePMF (N = 31), respectively. Overt MF had more mutations compared with ET/PV/prePMF (5 vs. 4 per subject, P = 0.006). Genes frequently mutated in MF included high-risk genes (ASXL1, SRSF2, EZH2, IDH1/2, and U2AF1) and Ras pathway genes. Mutations in NRAS, KRAS, SRSF2, EZH2, IDH2, and NF1 were exclusive to MF. Advancing age, higher DIPSS, and poor overall survival (OS) correlated with increased variants in MF. Ras mutations were associated with higher leukocytes and platelets and poor OS. The comparison of gene expression showed upregulation of proliferation and inflammatory pathways in MF. Notably, ADGRL4, DNASE1L3, PLEKHGB4, HSPG2, MAMDC2, and DPYSL3 were differentially expressed in hematopoietic stem and differentiated cells. CONCLUSIONS Our results illustrate that evolution of MF from ET/PV/PrePMF likely advances with age, accumulation of mutations, and activation of proliferative pathways. The genes and pathways identified by integrated genomics approach provide insight into disease transformation and progression and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Malathi Kandarpa
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Dan Robinson
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Kristen Pettit
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Qing Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Gary Luker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Maureen Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Arul Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Moshe Talpaz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Mehra R, Nallandhighal S, Cotta B, Knuth Z, Su F, Kasputis A, Zhang Y, Wang R, Cao X, Udager AM, Dhanasekaran SM, Cieslik MP, Morgan TM, Salami SS. Discovery and Validation of a 15-Gene Prognostic Signature for Clear Cell Renal Cell Carcinoma. JCO Precis Oncol 2024; 8:e2300565. [PMID: 38810179 PMCID: PMC11569832 DOI: 10.1200/po.23.00565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/11/2024] [Accepted: 03/15/2024] [Indexed: 05/31/2024] Open
Abstract
PURPOSE Develop and validate gene expression-based biomarker associated with recurrent disease to facilitate risk stratification of clear cell renal cell carcinoma (ccRCC). MATERIALS AND METHODS We retrospectively identified 110 patients who underwent radical nephrectomy for ccRCC (discovery cohort). Patients who recurred were matched on the basis of grade/stage to patients without recurrence. Capture whole-transcriptome sequencing was performed on RNA isolated from archival tissue using the Illumina platform. We developed a gene-expression signature to predict recurrence-free survival/disease-free survival (DFS) using a 15-fold lasso and elastic-net regularized linear Cox model. We derived the 31-gene cell cycle progression (mxCCP) score using RNA-seq data for each patient. Kaplan-Meier (KM) curves and multivariable Cox proportional hazard testing were used to validate the independent prognostic impact of the gene-expression signature on DFS, disease-specific survival (DSS), and overall survival (OS) in two validation data sets (combined n = 761). RESULTS After quality control, the discovery cohort comprised 50 patients with recurrence and 41 patients without, with a median follow-up of 26 and 36 months, respectively. We developed a 15-gene (15G) signature, which was independently associated with worse DFS and DSS (DFS: hazard ratio [HR], 11.08 [95% CI, 4.9 to 25.1]; DSS: HR, 9.67 [95% CI, 3.4 to 27.7]) in a multivariable model adjusting for clinicopathologic parameters (including stage, size, grade, and necrosis [SSIGN] score and Memorial Sloan Kettering Cancer Center nomogram) and mxCCP score. The 15G signature was also independently associated with worse DFS and DSS in both validation data sets (Validation A [n = 382], DFS: HR, 2.6 [95% CI, 1.6 to 4.3]; DSS: HR, 3 [95% CI, 1.4 to 6.1] and Validation B (n = 379), DFS: HR, 2.1 [95% CI, 1.2 to 3.6]; OS: HR, 3 [95% CI, 1.6 to 5.7]) adjusting for clinicopathologic variables and mxCCP score. CONCLUSION We developed and validated a novel 15G prognostic signature to improve risk stratification of patients with ccRCC. Pending further validation, this signature has the potential to facilitate optimal treatment allocation.
Collapse
Affiliation(s)
- Rohit Mehra
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | | | - Brittney Cotta
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Zayne Knuth
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Fengyun Su
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Amy Kasputis
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rui Wang
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Aaron M. Udager
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Saravana M Dhanasekaran
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Marcin P. Cieslik
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Todd M. Morgan
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Simpa S. Salami
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Kotlov N, Shaposhnikov K, Tazearslan C, Chasse M, Baisangurov A, Podsvirova S, Fernandez D, Abdou M, Kaneunyenye L, Morgan K, Cheremushkin I, Zemskiy P, Chelushkin M, Sorokina M, Belova E, Khorkova S, Lozinsky Y, Nuzhdina K, Vasileva E, Kravchenko D, Suryamohan K, Nomie K, Curran J, Fowler N, Bagaev A. Procrustes is a machine-learning approach that removes cross-platform batch effects from clinical RNA sequencing data. Commun Biol 2024; 7:392. [PMID: 38555407 PMCID: PMC10981711 DOI: 10.1038/s42003-024-06020-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 03/06/2024] [Indexed: 04/02/2024] Open
Abstract
With the increased use of gene expression profiling for personalized oncology, optimized RNA sequencing (RNA-seq) protocols and algorithms are necessary to provide comparable expression measurements between exome capture (EC)-based and poly-A RNA-seq. Here, we developed and optimized an EC-based protocol for processing formalin-fixed, paraffin-embedded samples and a machine-learning algorithm, Procrustes, to overcome batch effects across RNA-seq data obtained using different sample preparation protocols like EC-based or poly-A RNA-seq protocols. Applying Procrustes to samples processed using EC and poly-A RNA-seq protocols showed the expression of 61% of genes (N = 20,062) to correlate across both protocols (concordance correlation coefficient > 0.8, versus 26% before transformation by Procrustes), including 84% of cancer-specific and cancer microenvironment-related genes (versus 36% before applying Procrustes; N = 1,438). Benchmarking analyses also showed Procrustes to outperform other batch correction methods. Finally, we showed that Procrustes can project RNA-seq data for a single sample to a larger cohort of RNA-seq data. Future application of Procrustes will enable direct gene expression analysis for single tumor samples to support gene expression-based treatment decisions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mary Abdou
- BostonGene, Corp., Waltham, MA, 02453, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Karagianni K, Bibi A, Madé A, Acharya S, Parkkonen M, Barbalata T, Srivastava PK, de Gonzalo-Calvo D, Emanueli C, Martelli F, Devaux Y, Dafou D, Nossent AY, on behalf of EU-CardioRNA COST Action CA17129. Recommendations for detection, validation, and evaluation of RNA editing events in cardiovascular and neurological/neurodegenerative diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102085. [PMID: 38192612 PMCID: PMC10772297 DOI: 10.1016/j.omtn.2023.102085] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
RNA editing, a common and potentially highly functional form of RNA modification, encompasses two different RNA modifications, namely adenosine to inosine (A-to-I) and cytidine to uridine (C-to-U) editing. As inosines are interpreted as guanosines by the cellular machinery, both A-to-I and C-to-U editing change the nucleotide sequence of the RNA. Editing events in coding sequences have the potential to change the amino acid sequence of proteins, whereas editing events in noncoding RNAs can, for example, affect microRNA target binding. With advancing RNA sequencing technology, more RNA editing events are being discovered, studied, and reported. However, RNA editing events are still often overlooked or discarded as sequence read quality defects. With this position paper, we aim to provide guidelines and recommendations for the detection, validation, and follow-up experiments to study RNA editing, taking examples from the fields of cardiovascular and brain disease. We discuss all steps, from sample collection, storage, and preparation, to different strategies for RNA sequencing and editing-sensitive data analysis strategies, to validation and follow-up experiments, as well as potential pitfalls and gaps in the available technologies. This paper may be used as an experimental guideline for RNA editing studies in any disease context.
Collapse
Affiliation(s)
- Korina Karagianni
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Alisia Madé
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milan, Italy
| | - Shubhra Acharya
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-alzette, Luxembourg
| | - Mikko Parkkonen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Teodora Barbalata
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B. P. Hasdeu Street, 050568 Bucharest, Romania
| | | | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | | | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milan, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Dimitra Dafou
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - A. Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Department of Nutrition, Exercise and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| | - on behalf of EU-CardioRNA COST Action CA17129
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-alzette, Luxembourg
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B. P. Hasdeu Street, 050568 Bucharest, Romania
- National Heart & Lung Institute, Imperial College London, London, UK
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Department of Nutrition, Exercise and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Abdulfatah E, Al-Obaidy KI, Robinson D, Wu YM, Heider A, Idrees MT, Ulbright TM, Kunju LP, Wu A. Molecular characterization of large cell calcifying sertoli cell tumors: A multi-institutional study of 6 benign and 2 malignant tumors. Hum Pathol 2024; 144:15-21. [PMID: 38154678 DOI: 10.1016/j.humpath.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
Large cell calcifying Sertoli cell tumors (LCCSCTs) are rare testicular tumors, representing <1 % of all testicular neoplasms. Almost 40 % of patients with LCCSCTs will present in the context of the inherited tumor predisposition syndrome, the Carney complex. While most LCCSCTs are benign, 10-20 % have malignant behavior. The aim of our study was to analyze LCCSCTs for novel molecular alterations in addition to PRKAR1A mutations and to identify potential drivers for malignant progression. Eight LCCSCTs diagnosed at two institutions were included. Two patients had the Carney complex confirmed on subsequent genetic testing, and two tumors had several adverse pathological findings. One patient presented with metastatic disease at the time of initial diagnosis. Targeted next-generation sequencing detected PRKAR1A alterations in all cases, with heterozygous PRKAR1A mutations in 5 tumors, germline Carney-complex-associated PRKAR1A mutation in 2 patients, and PRKAR1A fusion in 1 tumor. Additionally, sequencing the metastatic case identified CDKN1B and TERT promoter gene mutations. All tumors showed a low tumoral mutational burden and unremarkable copy number alterations except for frequent LOH of 17q24 encompassing the PRKAR1A locus. RNA expression analysis showed increased expression of several markers including novel PRUNE2, and usual markers like inhibin and calretinin. Our study showed that while LCCSCTs have been reported in the setting of cancer predisposition syndromes, the majority of these tumors occur sporadically. PRKAR1A alterations were present in all cases and appear to be the major driver in LCCSCTs. It remains to be determined whether malignant progression may be caused by additional driver mutations.
Collapse
Affiliation(s)
- Eman Abdulfatah
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | | | - Dan Robinson
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Amer Heider
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | - Lakshmi Pryia Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Angela Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Hu Z, Kovach AE, Yellapantula V, Ostrow D, Doan A, Ji J, Schmidt RJ, Gu Z, Bhojwani D, Raca G. Transcriptome Sequencing Allows Comprehensive Genomic Characterization of Pediatric B-Acute Lymphoblastic Leukemia in an Academic Clinical Laboratory. J Mol Diagn 2024; 26:49-60. [PMID: 37981088 PMCID: PMC10773144 DOI: 10.1016/j.jmoldx.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 11/21/2023] Open
Abstract
Studies have shown the power of transcriptome sequencing [RNA sequencing (RNA-Seq)] in identifying known and novel oncogenic drivers and molecular subtypes of B-acute lymphoblastic leukemia (B-ALL). The current study investigated whether the clinically validated RNA-Seq assay, coupled with a custom analysis pipeline, could be used for a comprehensive B-ALL classification. Following comprehensive clinical testing, RNA-Seq was performed on 76 retrospective B-ALL cases, 28 of which had known and 48 had undetermined subtype. Subtypes were accurately identified in all 28 known cases, and in 38 of 48 unknown cases (79%). The subtypes of the unknown cases included the following: PAX5alt (n = 12), DUX4-rearranged (n = 6), Philadelphia chromosome-like (n = 5), low hyperdiploid (n = 4), ETV6::RUNX1-like (n = 3), MEF2D-rearranged (n = 2), PAX5 P80R (n = 2), ZEB2/CEBP (n = 1), NUTM1-rearranged (n = 1), ZNF384-rearranged (n = 1), and TCF3::PBX1 (n = 1). In 15 of 38 cases (39%), classification based on expression profile was corroborated by detection of subtype-defining oncogenic drivers missed by clinical testing. RNA-Seq analysis also detected large copy number abnormalities, oncogenic hot-spot sequence variants, and intragenic IKZF1 deletions. This pilot study confirms the feasibility of implementing an RNA-Seq workflow for clinical diagnosis of molecular subtypes in pediatric B-ALL, reinforcing that RNA-Seq represents a promising global genomic assay for this heterogeneous leukemia.
Collapse
Affiliation(s)
- Zunsong Hu
- Department of Computational and Quantitative Medicine and Systems Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Alexandra E Kovach
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Venkata Yellapantula
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Dejerianne Ostrow
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Andrew Doan
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Jianling Ji
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Ryan J Schmidt
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Zhaohui Gu
- Department of Computational and Quantitative Medicine and Systems Biology, Beckman Research Institute of City of Hope, Duarte, California.
| | - Deepa Bhojwani
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Gordana Raca
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California.
| |
Collapse
|
21
|
Bao Y, Qiao Y, Choi JE, Zhang Y, Mannan R, Cheng C, He T, Zheng Y, Yu J, Gondal M, Cruz G, Grove S, Cao X, Su F, Wang R, Chang Y, Kryczek I, Cieslik M, Green MD, Zou W, Chinnaiyan AM. Targeting the lipid kinase PIKfyve upregulates surface expression of MHC class I to augment cancer immunotherapy. Proc Natl Acad Sci U S A 2023; 120:e2314416120. [PMID: 38011559 PMCID: PMC10710078 DOI: 10.1073/pnas.2314416120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
Despite the remarkable clinical success of immunotherapies in a subset of cancer patients, many fail to respond to treatment and exhibit resistance. Here, we found that genetic or pharmacologic inhibition of the lipid kinase PIKfyve, a regulator of autophagic flux and lysosomal biogenesis, upregulated surface expression of major histocompatibility complex class I (MHC-I) in cancer cells via impairing autophagic flux, resulting in enhanced cancer cell killing mediated by CD8+ T cells. Genetic depletion or pharmacologic inhibition of PIKfyve elevated tumor-specific MHC-I surface expression, increased intratumoral functional CD8+ T cells, and slowed tumor progression in multiple syngeneic mouse models. Importantly, enhanced antitumor responses by Pikfyve-depletion were CD8+ T cell- and MHC-I-dependent, as CD8+ T cell depletion or B2m knockout rescued tumor growth. Furthermore, PIKfyve inhibition improved response to immune checkpoint blockade (ICB), adoptive cell therapy, and a therapeutic vaccine. High expression of PIKFYVE was also predictive of poor response to ICB and prognostic of poor survival in ICB-treated cohorts. Collectively, our findings show that targeting PIKfyve enhances immunotherapies by elevating surface expression of MHC-I in cancer cells, and PIKfyve inhibitors have potential as agents to increase immunotherapy response in cancer patients.
Collapse
Affiliation(s)
- Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
| | - Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Mahnoor Gondal
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yu Chang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109
| | - Michael D. Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI48109
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI48109
| | - Weiping Zou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Urology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
22
|
Romanens L, Chaskar P, Marcone R, Ryser S, Tille JC, Genolet R, Heimgartner-Hu K, Heimgartner K, Moore JS, Liaudet N, Kaya G, Pittet MJ, Dietrich PY, Delorenzi M, Speiser DE, Harari A, Tsantoulis P, Labidi-Galy SI. Clonal expansion of intra-epithelial T cells in breast cancer revealed by spatial transcriptomics. Int J Cancer 2023; 153:1568-1578. [PMID: 37306359 DOI: 10.1002/ijc.34620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 06/13/2023]
Abstract
The spatial distribution of tumor-infiltrating lymphocytes (TIL) predicts breast cancer outcome and response to systemic therapy, highlighting the importance of an intact tissue structure for characterizing tumors. Here, we present ST-FFPE, a spatial transcriptomics method for the analysis of formalin-fixed paraffin-embedded samples, which opens the possibility of interrogating archival tissue. The method involves extraction, exome capture and sequencing of RNA from different tumor compartments microdissected by laser-capture, and can be used to study the cellular composition of tumor microenvironment. Focusing on triple-negative breast cancer (TNBC), we characterized T cells, B cells, dendritic cells, fibroblasts and endothelial cells in both stromal and intra-epithelial compartments. We found a highly variable spatial distribution of immune cell subsets among tumors. This analysis revealed that the immune repertoires of intra-epithelial T and B cells were consistently less diverse and more clonal than those of stromal T and B cells. T-cell receptor (TCR) sequencing confirmed a reduced diversity and higher clonality of intra-epithelial T cells relative to the corresponding stromal T cells. Analysis of the top 10 dominant clonotypes in the two compartments showed a majority of shared but also some unique clonotypes both in stromal and intra-epithelial T cells. Hyperexpanded clonotypes were more abundant among intra-epithelial than stromal T cells. These findings validate the ST-FFPE method and suggest an accumulation of antigen-specific T cells within tumor core. Because ST-FFPE is applicable for analysis of previously collected tissue samples, it could be useful for rapid assessment of intratumoral cellular heterogeneity in multiple disease and treatment settings.
Collapse
Affiliation(s)
- Lou Romanens
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
| | - Prasad Chaskar
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Rachel Marcone
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Stephan Ryser
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
| | - Jean-Christophe Tille
- Department of Diagnosis, Division of Clinical Pathology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Raphael Genolet
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Ketty Heimgartner-Hu
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
| | - Killian Heimgartner
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
| | - Jonathan S Moore
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
| | - Nicolas Liaudet
- Bioimaging Core Facility, Faculty of Medicine, University of Geneva, Genève, Switzerland
| | - Gürkan Kaya
- Department of Diagnosis, Division of Clinical Pathology, Hôpitaux Universitaires de Genève, Genève, Switzerland
- Department of Medicine, Division of Dermatology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Mikael J Pittet
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Genève, Switzerland
- Ludwig Institute for Cancer Research, Lausanne, Switzerland
- AGORA Cancer Center, Lausanne, Switzerland
| | - Pierre-Yves Dietrich
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Mauro Delorenzi
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Daniel E Speiser
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Swiss Cancer Center Leman, Lausanne, Switzerland
- AGORA Cancer Center, Lausanne, Switzerland
| | - Petros Tsantoulis
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Sana Intidhar Labidi-Galy
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Genève, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| |
Collapse
|
23
|
Song K, Elboudwarej E, Zhao X, Zhuo L, Pan D, Liu J, Brachmann C, Patterson SD, Yoon OK, Zavodovskaya M. RNA-seq RNAaccess identified as the preferred method for gene expression analysis of low quality FFPE samples. PLoS One 2023; 18:e0293400. [PMID: 37883360 PMCID: PMC10602291 DOI: 10.1371/journal.pone.0293400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical tumor tissues that are preserved as formalin-fixed paraffin-embedded (FFPE) samples result in extensive cross-linking, fragmentation, and chemical modification of RNA, posing significant challenges for RNA-seq-based gene expression profiling. This study sought to define an optimal RNA-seq protocol for FFPE samples. We employed a common RNA extraction method and then compared RNA-seq library preparation protocols including RNAaccess, RiboZero and PolyA in terms of sequencing quality and concordance of gene expression using FFPE and case-matched fresh-frozen (FF) triple-negative breast cancer (TNBC) tissues. We found that RNAaccess, a method based on exome capture, produced the most concordant results. Applying RNAaccess to FFPE gastric cancer tissues, we established a minimum RNA DV200 requirement of 10% and a RNA input amount of 10ng that generated highly reproducible gene expression data. Lastly, we demonstrated that RNAaccess and NanoString platforms produced highly concordant expression profiles from FFPE samples for shared genes; however, RNA-seq may be preferred for clinical biomarker discovery work because of the broader coverage of the transcriptome. Taken together, these results support the selection of RNA-seq RNAaccess method for gene expression profiling of FFPE samples. The minimum requirements for RNA quality and input established here may allow for inclusion of clinical FFPE samples of sub-optimal quality in gene expression analyses and ultimately increasing the statistical power of such analyses.
Collapse
Affiliation(s)
- Kai Song
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Emon Elboudwarej
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Xi Zhao
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Luting Zhuo
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - David Pan
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Jinfeng Liu
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Carrie Brachmann
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Scott D. Patterson
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Oh Kyu Yoon
- Gilead Sciences, Inc., Foster City, California, United States of America
| | | |
Collapse
|
24
|
Rumpler É, Göcz B, Skrapits K, Sárvári M, Takács S, Farkas I, Póliska S, Papp M, Solymosi N, Hrabovszky E. Development of a versatile LCM-Seq method for spatial transcriptomics of fluorescently tagged cholinergic neuron populations. J Biol Chem 2023; 299:105121. [PMID: 37536628 PMCID: PMC10477691 DOI: 10.1016/j.jbc.2023.105121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/29/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023] Open
Abstract
Single-cell transcriptomics are powerful tools to define neuronal cell types based on co-expressed gene clusters. Limited RNA input in these technologies necessarily compromises transcriptome coverage and accuracy of differential expression analysis. We propose that bulk RNA-Seq of neuronal pools defined by spatial position offers an alternative strategy to overcome these technical limitations. We report a laser-capture microdissection (LCM)-Seq method which allows deep transcriptome profiling of fluorescently tagged neuron populations isolated with LCM from histological sections of transgenic mice. Mild formaldehyde fixation of ZsGreen marker protein, LCM sampling of ∼300 pooled neurons, followed by RNA isolation, library preparation and RNA-Seq with methods optimized for nanogram amounts of moderately degraded RNA enabled us to detect ∼15,000 different transcripts in fluorescently labeled cholinergic neuron populations. The LCM-Seq approach showed excellent accuracy in quantitative studies, allowing us to detect 2891 transcripts expressed differentially between the spatially defined and clinically relevant cholinergic neuron populations of the dorsal caudate-putamen and medial septum. In summary, the LCM-Seq method we report in this study is a versatile, sensitive, and accurate bulk sequencing approach to study the transcriptome profile and differential gene expression of fluorescently tagged neuronal populations isolated from transgenic mice with high spatial precision.
Collapse
Affiliation(s)
- Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary.
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Márton Papp
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
25
|
Mannan R, Wang X, Bawa PS, Zhang Y, Skala SL, Chinnaiyan AK, Dagar A, Wang L, Zelenka-Wang SB, McMurry LM, Daniel N, Cao X, Sangoi AR, Gupta S, Vaishampayan UN, Hafez KS, Morgan TM, Spratt DE, Tretiakova MS, Argani P, Chinnaiyan AM, Dhanasekaran SM, Mehra R. Characterization of Intercalated Cell Markers KIT and LINC01187 in Chromophobe Renal Cell Carcinoma and Other Renal Neoplasms. Int J Surg Pathol 2023; 31:1027-1040. [PMID: 36250542 DOI: 10.1177/10668969221125793] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Introduction. Chromophobe renal cell carcinoma (chromophobe RCC) is the third major subcategory of renal tumors after clear cell RCC and papillary RCC, accounting for approximately 5% of all RCC subtypes. Other oncocytic neoplasms seen commonly in surgical pathology practice include the eosinophilic variant of chromophobe RCC, renal oncocytoma, and low-grade oncocytic unclassified RCC. Methods. In our recent next-generation sequencing based study, we nominated a lineage-specific novel biomarker LINC01187 (long intergenic non-protein coding RNA 1187) which was found to be enriched in chromophobe RCC. Like KIT (cluster of differentiation 117; CD117), a clinically utilized chromophobe RCC related biomarker, LINC01187 is expressed in intercalated cells of the nephron. In this follow-up study, we performed KIT immunohistochemistry and LINC01187 RNA in situ hybridization (RNA-ISH) on a cohort of chromophobe RCC and other renal neoplasms, characterized the expression patterns, and quantified the expression signals of the two biomarkers in both primary and metastatic settings. Results. LINC01187, in comparison to KIT, exhibits stronger and more uniform expression within tumors while maintaining temporal and spatial consistency. LINC01187 also is devoid of intra-tumoral heterogeneous expression pattern, a phenomenon commonly noted with KIT. Conclusions. LINC01187 expression can augment the currently utilized KIT assay and help facilitate easy microscopic analyses in routine surgical pathology practice.
Collapse
Affiliation(s)
- Rahul Mannan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Xiaoming Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Pushpinder S Bawa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Stephanie L Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Aniket Dagar
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Lisha Wang
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Sylvia B Zelenka-Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Lisa M McMurry
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Nikita Daniel
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, Ann Arbor, MI, USA
| | - Ankur R Sangoi
- Department of Pathology, El Camino Hospital, Mountain View, CA, USA
| | - Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ulka N Vaishampayan
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Department of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Khaled S Hafez
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Todd M Morgan
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, Ann Arbor, MI, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Saravana M Dhanasekaran
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Michuda J, Breschi A, Kapilivsky J, Manghnani K, McCarter C, Hockenberry AJ, Mineo B, Igartua C, Dudley JT, Stumpe MC, Beaubier N, Shirazi M, Jones R, Morency E, Blackwell K, Guinney J, Beauchamp KA, Taxter T. Validation of a Transcriptome-Based Assay for Classifying Cancers of Unknown Primary Origin. Mol Diagn Ther 2023; 27:499-511. [PMID: 37099070 PMCID: PMC10300170 DOI: 10.1007/s40291-023-00650-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2023] [Indexed: 04/27/2023]
Abstract
INTRODUCTION Cancers assume a variety of distinct histologies, and may originate from a myriad of sites including solid organs, hematopoietic cells, and connective tissue. Clinical decision-making based on consensus guidelines such as the National Comprehensive Cancer Network (NCCN) is often predicated on a specific histologic and anatomic diagnosis, supported by clinical features and pathologist interpretation of morphology and immunohistochemical (IHC) staining patterns. However, in patients with nonspecific morphologic and IHC findings-in addition to ambiguous clinical presentations such as recurrence versus new primary-a definitive diagnosis may not be possible, resulting in the patient being categorized as having a cancer of unknown primary (CUP). Therapeutic options and clinical outcomes are poor for patients with CUP, with a median survival of 8-11 months. METHODS Here, we describe and validate the Tempus Tumor Origin (Tempus TO) assay, an RNA-sequencing-based machine learning classifier capable of discriminating between 68 clinically relevant cancer subtypes. Model accuracy was assessed using primary and/or metastatic samples with known subtype. RESULTS We show that the Tempus TO model is 91% accurate when assessed on both a retrospectively held out cohort and a set of samples sequenced after model freeze that collectively contained 9210 total samples with known diagnoses. When evaluated on a cohort of CUPs, the model recapitulated established associations between genomic alterations and cancer subtype. DISCUSSION Combining diagnostic prediction tests (e.g., Tempus TO) with sequencing-based variant reporting (e.g., Tempus xT) may expand therapeutic options for patients with cancers of unknown primary or uncertain histology.
Collapse
|
27
|
Oreper D, Klaeger S, Jhunjhunwala S, Delamarre L. The peptide woods are lovely, dark and deep: Hunting for novel cancer antigens. Semin Immunol 2023; 67:101758. [PMID: 37027981 DOI: 10.1016/j.smim.2023.101758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
Harnessing the patient's immune system to control a tumor is a proven avenue for cancer therapy. T cell therapies as well as therapeutic vaccines, which target specific antigens of interest, are being explored as treatments in conjunction with immune checkpoint blockade. For these therapies, selecting the best suited antigens is crucial. Most of the focus has thus far been on neoantigens that arise from tumor-specific somatic mutations. Although there is clear evidence that T-cell responses against mutated neoantigens are protective, the large majority of these mutations are not immunogenic. In addition, most somatic mutations are unique to each individual patient and their targeting requires the development of individualized approaches. Therefore, novel antigen types are needed to broaden the scope of such treatments. We review high throughput approaches for discovering novel tumor antigens and some of the key challenges associated with their detection, and discuss considerations when selecting tumor antigens to target in the clinic.
Collapse
Affiliation(s)
- Daniel Oreper
- Genentech, 1 DNA way, South San Francisco, 94080 CA, USA.
| | - Susan Klaeger
- Genentech, 1 DNA way, South San Francisco, 94080 CA, USA.
| | | | | |
Collapse
|
28
|
Crudele F, Bianchi N, Terrazzan A, Ancona P, Frassoldati A, Gasparini P, D'Adamo AP, Papaioannou D, Garzon R, Wójcicka A, Gaj P, Jażdżewski K, Palatini J, Volinia S. Circular RNAs Could Encode Unique Proteins and Affect Cancer Pathways. BIOLOGY 2023; 12:biology12040493. [PMID: 37106694 PMCID: PMC10135897 DOI: 10.3390/biology12040493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023]
Abstract
circRNAs constitute a novel class of RNA, generally considered as non-coding RNAs; nonetheless, their coding potential has been under scrutiny. In this work, we systematically explored the predicted proteins of more than 160,000 circRNAs detected by exome capture RNA-sequencing and collected in the MiOncoCirc pan-cancer compendium, including normal and cancer samples from different types of tissues. For the functional evaluation, we compared their primary structure and domain composition with those derived from the same linear mRNAs. Among the 4362 circRNAs potentially encoding proteins with a unique primary structure and 1179 encoding proteins with a novel domain composition, 183 were differentially expressed in cancer. In particular, eight were associated with prognosis in acute myeloid leukemia. The functional classification of the dysregulated circRNA-encoded polypeptides showed an enrichment in the heme and cancer signaling, DNA-binding, and phosphorylation processes, and disclosed the roles of some circRNA-based effectors in cancer.
Collapse
Affiliation(s)
- Francesca Crudele
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Genetics Unit, Institute for Maternal and Child Health, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Burlo Garofolo, 34137 Trieste, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Laboratory for Advanced Therapy Technologies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Antonio Frassoldati
- Department of Oncology, Azienda Ospedaliero-Universitaria St. Anna di Ferrara, 44124 Ferrara, Italy
| | - Paolo Gasparini
- Genetics Unit, Institute for Maternal and Child Health, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Burlo Garofolo, 34137 Trieste, Italy
| | - Adamo P D'Adamo
- Genetics Unit, Institute for Maternal and Child Health, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Burlo Garofolo, 34137 Trieste, Italy
| | - Dimitrios Papaioannou
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Ramiro Garzon
- Division of Hematology and Hematological Malignancies, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Paweł Gaj
- Warsaw Genomics INC, 01-682 Warszawa, Poland
| | - Krystian Jażdżewski
- Human Cancer Genetics, Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Jeffrey Palatini
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Laboratory for Advanced Therapy Technologies (LTTA), University of Ferrara, 44121 Ferrara, Italy
- CNBCh, Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| |
Collapse
|
29
|
Li H, Zhang Y, Bing J, Han J, Hu J, Zhao H, Sun X. Intron-capture RNA-seq reveals the landscape of intronic RNAs in Arabidopsis. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 196:75-88. [PMID: 36701993 DOI: 10.1016/j.plaphy.2023.01.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/19/2023] [Indexed: 06/17/2023]
Abstract
Intronic RNAs have been overlooked for a long time: They are functional, but treated as "junk." In this work, we designed a new sequencing strategy to investigate intronic RNAs. By using intron-capture RNA-seq, we systematically analyzed the intronic RNAs in Arabidopsis by zooming into the intronic regions an order of magnitude deeper than in previous work. Our key findings include: (1) Intron-capture RNA-seq is a much more efficient approach to analyze intronic RNAs than total RNA-seq and mRNA-seq. (2) We identified three types of intronic RNAs, and found that the GC pattern differs significantly between the introns with and without intronic RNAs. (3) We detected many hidden elements in introns, including circular RNAs, splice junctions, and transcripts that have previously been overlooked. (4) The expression of these intronic RNAs varies during the time course of pathogen infection, which indicates that an unknown mechanism may exist for these RNAs. (5) We also demonstrated that most of intronic RNAs are detectable in both Arabidopsis and rice, suggesting that these non-coding molecules are conserved. Taken together, this work proposes an efficient strategy to analyze intronic RNAs, and provides an unprecedented view of this essential component in biological pathways.
Collapse
Affiliation(s)
- Han Li
- Agricultural Big-Data Research Center, College of Information Science and Engineering, Shandong Agricultural University, Taian, China
| | - Yimai Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Jianhao Bing
- Agricultural Big-Data Research Center, College of Information Science and Engineering, Shandong Agricultural University, Taian, China
| | - Jinyu Han
- Agricultural Big-Data Research Center, College of Information Science and Engineering, Shandong Agricultural University, Taian, China
| | - Jiming Hu
- Agricultural Big-Data Research Center, College of Information Science and Engineering, Shandong Agricultural University, Taian, China
| | - Hongwei Zhao
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China.
| | - Xiaoyong Sun
- Agricultural Big-Data Research Center, College of Information Science and Engineering, Shandong Agricultural University, Taian, China.
| |
Collapse
|
30
|
Decruyenaere P, Verniers K, Poma-Soto F, Van Dorpe J, Offner F, Vandesompele J. RNA Extraction Method Impacts Quality Metrics and Sequencing Results in Formalin-Fixed, Paraffin-Embedded Tissue Samples. J Transl Med 2023; 103:100027. [PMID: 37039153 DOI: 10.1016/j.labinv.2022.100027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/19/2022] [Accepted: 11/03/2022] [Indexed: 01/11/2023] Open
Abstract
Archived formalin-fixed, paraffin-embedded (FFPE) tissue samples are being increasingly used in molecular cancer research. Compared with fresh-frozen tissue, the nucleic acid analysis of FFPE tissue is technically more challenging. This study aimed to compare the impact of 3 different RNA extraction methods on yield, quality, and sequencing-based gene expression results in FFPE samples. RNA extraction was performed in 16 FFPE tumor specimens from patients with diffuse large B-cell lymphoma and in reference FFPE material from microsatellite-stable and microsatellite-instable cell lines (3 replicates each) using 2 silica-based procedures (A, miRNeasy FFPE; C, iCatcher FFPE Tissue RNA) and 1 isotachophoresis-based procedure (B, Ionic FFPE to Pure RNA). The RNA yield; RNA integrity, as reflected by the distribution value 200; and RNA purity, as reflected by the 260/280 and the 260/230 nm absorbance ratios, were determined. The RNA was sequenced on the NovaSeq 6000 instrument using the TruSeq RNA Exome and SMARTer Stranded Total RNA-Seq Pico v3 library preparations kits. Our results highlight the impact of RNA extraction methodology on both preanalytical and sequencing-based gene expression results. Overall, methods B and C outperformed method A because these showed significantly higher fractions of uniquely mapped reads, an increased number of detectable genes, a lower fraction of duplicated reads, and better representation of the B-cell receptor repertoire. Differences among the extraction methods were generally more explicit for the total RNA sequencing method than for the exome-capture sequencing method. Importantly, the predicative value of quality metrics varies among extraction kits, and caution should be applied when comparing and interpreting results obtained using different methods.
Collapse
|
31
|
Liu Y, Bhagwate A, Winham SJ, Stephens MT, Harker BW, McDonough SJ, Stallings-Mann ML, Heinzen EP, Vierkant RA, Hoskin TL, Frost MH, Carter JM, Pfrender ME, Littlepage L, Radisky DC, Cunningham JM, Degnim AC, Wang C. Quality control recommendations for RNASeq using FFPE samples based on pre-sequencing lab metrics and post-sequencing bioinformatics metrics. BMC Med Genomics 2022; 15:195. [PMID: 36114500 PMCID: PMC9479231 DOI: 10.1186/s12920-022-01355-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background Formalin-fixed, paraffin-embedded (FFPE) tissues have many advantages for identification of risk biomarkers, including wide availability and potential for extended follow-up endpoints. However, RNA derived from archival FFPE samples has limited quality. Here we identified parameters that determine which FFPE samples have the potential for successful RNA extraction, library preparation, and generation of usable RNAseq data. Methods We optimized library preparation protocols designed for use with FFPE samples using seven FFPE and Fresh Frozen replicate pairs, and tested optimized protocols using a study set of 130 FFPE biopsies from women with benign breast disease. Metrics from RNA extraction and preparation procedures were collected and compared with bioinformatics sequencing summary statistics. Finally, a decision tree model was built to learn the relationship between pre-sequencing lab metrics and qc pass/fail status as determined by bioinformatics metrics. Results Samples that failed bioinformatics qc tended to have low median sample-wise correlation within the cohort (Spearman correlation < 0.75), low number of reads mapped to gene regions (< 25 million), or low number of detectable genes (11,400 # of detected genes with TPM > 4). The median RNA concentration and pre-capture library Qubit values for qc failed samples were 18.9 ng/ul and 2.08 ng/ul respectively, which were significantly lower than those of qc pass samples (40.8 ng/ul and 5.82 ng/ul). We built a decision tree model based on input RNA concentration, input library qubit values, and achieved an F score of 0.848 in predicting QC status (pass/fail) of FFPE samples. Conclusions We provide a bioinformatics quality control recommendation for FFPE samples from breast tissue by evaluating bioinformatic and sample metrics. Our results suggest a minimum concentration of 25 ng/ul FFPE-extracted RNA for library preparation and 1.7 ng/ul pre-capture library output to achieve adequate RNA-seq data for downstream bioinformatics analysis.
Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01355-0.
Collapse
|
32
|
Wilbur HC, Robinson DR, Wu YM, Kumar-Sinha C, Chinnaiyan AM, Chugh R. Identification of Novel PGR-NR4A3 Fusion in Extraskeletal Myxoid Chondrosarcoma and Resultant Patient Benefit From Tamoxifen Therapy. JCO Precis Oncol 2022; 6:e2200039. [PMID: 36103645 PMCID: PMC9489176 DOI: 10.1200/po.22.00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/24/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- H. Catherine Wilbur
- Department of Medicine, Division of Hematology and Oncology, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Dan R. Robinson
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Department of Pathology and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Department of Pathology and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Department of Pathology and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Department of Pathology and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Rashmi Chugh
- Department of Medicine, Division of Hematology and Oncology, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
33
|
Lu W, Zhou Q, Chen Y. Impact of RNA degradation on next-generation sequencing transcriptome data. Genomics 2022; 114:110429. [PMID: 35810931 DOI: 10.1016/j.ygeno.2022.110429] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/16/2022] [Accepted: 07/06/2022] [Indexed: 11/04/2022]
Abstract
RNA sequencing is an innovative technology to study transcriptomes in both biological and clinical research. However, clinical specimens from patients undergoing surgical operations have a major challenge due to sample degradation. This study replicated the process of RNA degradation by maintaining cells at room temperature to achieve none, slight, middle, and high levels of RNA degradation with decreasing RNA integrity numbers (RIN) of approximately 9.8, 6.7, 4.4, and 2.5, respectively. Next, the differential expression of mRNA and long non-coding RNA (lncRNA) was analyzed in the four degradation groups along with pathway enrichment analysis. The results showed that the similarity of lncRNAs exhibited significant differences even for a slight level of RNA degradation compared with the non-degraded RNA sample. Also, the RNA degradation process was found to be universal, global, and random; the differentially expressed genes increased with an increase in degradation but the pathway enrichment phenomenon was not significantly observed.
Collapse
Affiliation(s)
- Wenxiang Lu
- State Key Lab of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Qin Zhou
- Department of Obstetrics and Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan 215300, China
| | - Yi Chen
- Department of Obstetrics and Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan 215300, China.
| |
Collapse
|
34
|
The genetic heterogeneity and drug resistance mechanisms of relapsed refractory multiple myeloma. Nat Commun 2022; 13:3750. [PMID: 35768438 PMCID: PMC9243087 DOI: 10.1038/s41467-022-31430-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022] Open
Abstract
Multiple myeloma is the second most common hematological malignancy. Despite significant advances in treatment, relapse is common and carries a poor prognosis. Thus, it is critical to elucidate the genetic factors contributing to disease progression and drug resistance. Here, we carry out integrative clinical sequencing of 511 relapsed, refractory multiple myeloma (RRMM) patients to define the disease’s molecular alterations landscape. The NF-κB and RAS/MAPK pathways are more commonly altered than previously reported, with a prevalence of 45–65% each. In the RAS/MAPK pathway, there is a long tail of variants associated with the RASopathies. By comparing our RRMM cases with untreated patients, we identify a diverse set of alterations conferring resistance to three main classes of targeted therapy in 22% of our cohort. Activating mutations in IL6ST are also enriched in RRMM. Taken together, our study serves as a resource for future investigations of RRMM biology and potentially informs clinical management. The genetic factors involved in disease progression and drug resistance in multiple myeloma (MM) are varied and complex. Here, genomic and transcriptomic profiling of 511 relapsed and refractory MM patients reveals genetic alterations in several oncogenic pathways contributing to progression and resistance to MM therapies.
Collapse
|
35
|
Swanson GM, Estill MS, Krawetz SA. The transcript integrity index (TII) provides a standard measure of sperm RNA. Syst Biol Reprod Med 2022; 68:258-271. [PMID: 35658756 DOI: 10.1080/19396368.2022.2071133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Standardizing RNA quality is key to interpreting RNA-seq data as a compromised sample can mask the underlying biology. The challenge remains when evaluating RNA quality in samples with high RNA fragmentation. For example, programmed fragmentation and cytoplasmic expulsion, integral to sperm maturation, is a prime example of the complexities of interpreting RNA-seq data, given that fragmentation can be random and\or targeted. To meet this challenge, we developed an algorithm that accurately measures RNA quality in samples with high fragmentation, such as spermatozoa. The integrity of 1,000 previously identified abundant sperm transcripts were independently visualized and evaluated using the Transcript Integrity Index (TII) algorithm to identify intact transcripts. Full-length transcripts from visual and the TII algorithm were evaluated for testis preference in humans using the GTEx tissues database. Samples were then filtered by the Interquartile Range (IQR), identifying those in which the greatest number of transcripts failed to pass the visual or TII thresholds. Transcript lists were overlapped, forming the set of intact transcripts used as TII standards. Each sample was re-evaluated as a function of this TII set of intact transcripts, with poor quality samples identified as those failing in the largest number of transcripts. While ontologically enriched in roles related to spermatogenesis and/or fertilization, samples did not segregate based on birth outcome. The TII algorithm proved an effective means to identify samples of similar quality from sperm, a cell type enriched in biologically fragmented RNAs. The algorithm should facilitate other studies using samples compromised by high levels of RNA fragmentation, such as Formalin-Fixed Paraffin-Embedded samples. Requisite to assessing male health, TII provides a solution to the long-sought-after standard that identifies samples of similar quality.
Collapse
Affiliation(s)
- Grace M Swanson
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Molly S Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen A Krawetz
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
36
|
Wang D, Rolfe PA, Foernzler D, O'Rourke D, Zhao S, Scheuenpflug J, Feng Z. Comparison of Two Illumina Whole Transcriptome RNA Sequencing Library Preparation Methods Using Human Cancer FFPE Specimens. Technol Cancer Res Treat 2022; 21:15330338221076304. [PMID: 35138205 PMCID: PMC8832632 DOI: 10.1177/15330338221076304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Objective: RNA extraction and library preparation from formalin-fixed, paraffin-embedded (FFPE) samples are crucial pre-analytical steps towards achieving optimal downstream RNA sequencing (RNASeq) results. In this study, we assessed 2 Illumina library preparation methods for RNA-Seq analysis using archived FFPE samples from human cancer indications at 2 independent vendors. Methods: Twenty-five FFPE samples from 5 indications (non-small cell lung cancer, colorectal cancer, renal carcinoma, breast cancer, and hepatocellular carcinoma) were included, covering a wide range of sample storage durations (3-25 years-old), sample qualities, and specimen types (resection vs core needle biopsy). Each sample was processed independently by both vendors. Total RNA was isolated using the Qiagen miRNeasy FFPE kit followed by library construction using either TruSeq Stranded Total RNA library preparation kit with Ribo-Zero Gold, or TruSeq RNA Access library preparation kit. Libraries were normalized to 20 pM and sequenced on an Illumina HiSeq 2500 using V3 chemistry in paired-end mode with a read length of 2 × 50 bp. The data were processed through a standard RNASeq pipeline to produce counts and transcripts per millions for each gene in each sample to compare 2 library kits at 2 different vendors. Results: Our data showed that TruSeq RNA Access libraries yield over 80% exonic reads across different quality samples, indicating higher selectivity of the exome pull down by the capture approach compared to the random priming of the TruSeq Stranded Total kit. The overall QC data for FFPE RNA extraction, library preparation, and sequencing generated by the 2 vendors are comparable, and downstream gene expression quantification results show high concordance as well. With the TruSeq Stranded Total kit, the mean Spearman correlation between vendors was 0.87 and the mean Pearson correlation was 0.76. With the TruSeq RNA Access kit, the mean Spearman correlation between vendors was 0.89 and the mean Pearson correlation was 0.73. Interestingly, examination of the cross-vendor correlations compared to various common QC statistics suggested that library concentration is better correlated with consistency between vendors than is the RNA quantity. Conclusions: Our analyses provide evidence to guide selection of sequencing methods for FFPE samples in which the sample quality may be severely compromised.
Collapse
Affiliation(s)
- Danyi Wang
- 189697Global Clinical Biomarkers and Companion Diagnostics, Translational Medicine, Global Development, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - P Alexander Rolfe
- 2792Immunology and Immuno-Oncology Bioinformatics, Translational Medicine, Global Development, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Dorothee Foernzler
- 189697Global Clinical Biomarkers and Companion Diagnostics, Translational Medicine, Global Development, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Dennis O'Rourke
- 189697Global Clinical Biomarkers and Companion Diagnostics, Translational Medicine, Global Development, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Sheng Zhao
- Oncology Bioinformatics, Translational Medicine, Global Development, Merck KGaA, Darmstadt, Germany
| | - Juergen Scheuenpflug
- Global Clinical Biomarkers and Companion Diagnostics, Translational Medicine, Global Development, Merck KGaA, Darmstadt, Germany
| | - Zheng Feng
- 189697Global Clinical Biomarkers and Companion Diagnostics, Translational Medicine, Global Development, EMD Serono Research and Development Institute, Billerica, MA, USA
| |
Collapse
|
37
|
Potemkin N, Cawood SMF, Treece J, Guévremont D, Rand CJ, McLean C, Stanton JAL, Williams JM. A method for simultaneous detection of small and long RNA biotypes by ribodepleted RNA-Seq. Sci Rep 2022; 12:621. [PMID: 35022475 PMCID: PMC8755727 DOI: 10.1038/s41598-021-04209-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/24/2021] [Indexed: 11/09/2022] Open
Abstract
RNA sequencing offers unprecedented access to the transcriptome. Key to this is the identification and quantification of many different species of RNA from the same sample at the same time. In this study we describe a novel protocol for simultaneous detection of coding and non-coding transcripts using modifications to the Ion Total RNA-Seq kit v2 protocol, with integration of QIASeq FastSelect rRNA removal kit. We report highly consistent sequencing libraries can be produced from both frozen high integrity mouse hippocampal tissue and the more challenging post-mortem human tissue. Removal of rRNA using FastSelect was extremely efficient, resulting in less than 1.5% rRNA content in the final library. We identified > 30,000 unique transcripts from all samples, including protein-coding genes and many species of non-coding RNA, in biologically-relevant proportions. Furthermore, the normalized sequencing read count for select genes significantly negatively correlated with Ct values from qRT-PCR analysis from the same samples. These results indicate that this protocol accurately and consistently identifies and quantifies a wide variety of transcripts simultaneously. The highly efficient rRNA depletion, coupled with minimized sample handling and without complicated and high-loss size selection protocols, makes this protocol useful to researchers wishing to investigate whole transcriptomes.
Collapse
Affiliation(s)
- Nikita Potemkin
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Sophie M F Cawood
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Jackson Treece
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Diane Guévremont
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Christy J Rand
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Catriona McLean
- Victorian Brain Bank, The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Anatomical Pathology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Jo-Ann L Stanton
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand.
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
38
|
Weckbach LT, Schweizer L, Kraechan A, Bieber S, Ishikawa-Ankerhold H, Hausleiter J, Massberg S, Straub T, Klingel K, Grabmaier U, Zwiebel M, Mann M, Schulz C. Association of Complement and MAPK Activation With SARS-CoV-2-Associated Myocardial Inflammation. JAMA Cardiol 2021; 7:286-297. [PMID: 34910083 PMCID: PMC8674808 DOI: 10.1001/jamacardio.2021.5133] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Question What is the cardiac phenotype of patients with SARS-CoV-2 infection compared with viral and immune-mediated myocarditis and noninflammatory cardiomyopathy? Findings In this case series of 19 patients undergoing endomyocardial biopsies, cardiac specimens of patients with SARS-CoV-2 infection had a higher abundance of complement-associated factors and serine/threonine protein kinases, with mitogen-activated protein kinase–associated pathways having the highest abundance. Similarities in the cardiac immune signature were found among those with SARS-CoV-2 infection and viral myocarditis. Meaning In this study, the exploratory data, which characterized myocardial inflammation by deep phenotyping, have implications for the development of treatment strategies to reduce SARS-CoV-2–mediated tissue injury; these findings require confirmation in a prospective and extended cohort of patients. Importance Myocardial injury is a common feature of patients with SARS-CoV-2 infection. However, the cardiac inflammatory processes associated with SARS-CoV-2 infection are not completely understood. Objective To investigate the inflammatory cardiac phenotype associated with SARS-CoV-2 infection compared with viral myocarditis, immune-mediated myocarditis, and noninflammatory cardiomyopathy by integrating histologic, transcriptomic, and proteomic profiling. Design, Setting, and Participants This case series was a cooperative study between the Ludwig Maximilian University Hospital Munich and the Cardiopathology Referral Center at the University of Tübingen in Germany. A cohort of 19 patients with suspected myocarditis was examined; of those, 5 patients were hospitalized with SARS-CoV-2 infection between March and May 2020. Cardiac tissue specimens from those 5 patients were compared with specimens from 5 patients with immune-mediated myocarditis, 4 patients with non–SARS-CoV-2 viral myocarditis, and 5 patients with noninflammatory cardiomyopathy, collected from January to August 2019. Exposures Endomyocardial biopsy. Main Outcomes and Measures The inflammatory cardiac phenotypes were measured by immunohistologic analysis, RNA exome capture sequencing, and mass spectrometry–based proteomic analysis of endomyocardial biopsy specimens. Results Among 19 participants, the median age was 58 years (range, 37-76 years), and 15 individuals (79%) were male. Data on race and ethnicity were not collected. The abundance of CD163+ macrophages was generally higher in the cardiac tissue of patients with myocarditis, whereas lymphocyte counts were lower in the tissue of patients with SARS-CoV-2 infection vs patients with non–SARS-CoV-2 virus-associated and immune-mediated myocarditis. Among those with SARS-CoV-2 infection, components of the complement cascade, including C1q subunits (transcriptomic analysis: 2.5-fold to 3.6-fold increase; proteomic analysis: 2.0-fold to 3.4-fold increase) and serine/cysteine proteinase inhibitor clade G member 1 (transcriptomic analysis: 1.7-fold increase; proteomic analysis: 2.6-fold increase), belonged to the most commonly upregulated transcripts and differentially abundant proteins. In cardiac macrophages, the abundance of C1q was highest in SARS-CoV-2 infection. Assessment of important signaling cascades identified an upregulation of the serine/threonine mitogen-activated protein kinase pathways. Conclusions and Relevance This case series found that the cardiac immune signature varied in inflammatory conditions with different etiologic characteristics. Future studies are needed to examine the role of these immune pathways in myocardial inflammation.
Collapse
Affiliation(s)
- Ludwig T Weckbach
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig Maximilian University Munich, Planegg-Martinsried, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Lisa Schweizer
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Planegg-Martinsried, Germany
| | - Angelina Kraechan
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig Maximilian University Munich, Planegg-Martinsried, Germany
| | - Stephanie Bieber
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany
| | | | - Jörg Hausleiter
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Ludwig Maximilian University Munich, Planegg-Martinsried, Germany
| | - Karin Klingel
- Cardiopathology Department, Institute for Pathology and Neuropathology, Tübingen University Hospital, Tübingen, Germany
| | - Ulrich Grabmaier
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Maximilian Zwiebel
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Planegg-Martinsried, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Planegg-Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University Hospital Munich, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | | |
Collapse
|
39
|
Genome-wide spatial expression profiling in formalin-fixed tissues. CELL GENOMICS 2021; 1:100065. [PMID: 36776149 PMCID: PMC9903805 DOI: 10.1016/j.xgen.2021.100065] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 04/27/2021] [Accepted: 08/30/2021] [Indexed: 12/31/2022]
Abstract
Formalin-fixed paraffin embedding (FFPE) is the most widespread long-term tissue preservation approach. Here, we report a procedure to perform genome-wide spatial analysis of mRNA in FFPE-fixed tissue sections, using well-established, commercially available methods for imaging and spatial barcoding using slides spotted with barcoded oligo(dT) probes to capture the 3' end of mRNA molecules in tissue sections. We applied this method for expression profiling and cell type mapping in coronal sections from the mouse brain to demonstrate the method's capability to delineate anatomical regions from a molecular perspective. We also profiled the spatial composition of transcriptomic signatures in two ovarian carcinosarcoma samples, exemplifying the method's potential to elucidate molecular mechanisms in heterogeneous clinical samples. Finally, we demonstrate the applicability of the assay to characterize human lung and kidney organoids and a human lung biopsy specimen infected with SARS-CoV-2. We anticipate that genome-wide spatial gene expression profiling in FFPE biospecimens will be used for retrospective analysis of biobank samples, which will facilitate longitudinal studies of biological processes and biomarker discovery.
Collapse
|
40
|
Jang JS, Holicky E, Lau J, McDonough S, Mutawe M, Koster MJ, Warrington KJ, Cuninngham JM. Application of the 3' mRNA-Seq using unique molecular identifiers in highly degraded RNA derived from formalin-fixed, paraffin-embedded tissue. BMC Genomics 2021; 22:759. [PMID: 34689749 PMCID: PMC8543821 DOI: 10.1186/s12864-021-08068-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/10/2021] [Indexed: 11/11/2022] Open
Abstract
Background Archival formalin-fixed, paraffin-embedded (FFPE) tissue samples with clinical and histological data are a singularly valuable resource for developing new molecular biomarkers. However, transcriptome analysis remains challenging with standard mRNA-seq methods as FFPE derived-RNA samples are often highly modified and fragmented. The recently developed 3′ mRNA-seq method sequences the 3′ region of mRNA using unique molecular identifiers (UMI), thus generating gene expression data with minimal PCR bias. In this study, we evaluated the performance of 3′ mRNA-Seq using Lexogen QuantSeq 3′ mRNA-Seq Library Prep Kit FWD with UMI, comparing with TruSeq Stranded mRNA-Seq and RNA Exome Capture kit. The fresh-frozen (FF) and FFPE tissues yielded nucleotide sizes range from 13 to > 70% of DV200 values; input amounts ranged from 1 ng to 100 ng for validation. Results The total mapped reads of QuantSeq 3′ mRNA-Seq to the reference genome ranged from 99 to 74% across all samples. After PCR bias correction, 3 to 56% of total sequenced reads were retained. QuantSeq 3′ mRNA-Seq data showed highly reproducible data across replicates in Universal Human Reference RNA (UHR, R > 0.94) at input amounts from 1 ng to 100 ng, and FF and FFPE paired samples (R = 0.92) at 10 ng. Severely degraded FFPE RNA with ≤30% of DV200 value showed good concordance (R > 0.87) with 100 ng input. A moderate correlation was observed when directly comparing QuantSeq 3′ mRNA-Seq data with TruSeq Stranded mRNA-Seq (R = 0.78) and RNA Exome Capture data (R > 0.67). Conclusion In this study, QuantSeq 3′ mRNA-Seq with PCR bias correction using UMI is shown to be a suitable method for gene quantification in both FF and FFPE RNAs. 3′ mRNA-Seq with UMI may be applied to severely degraded RNA from FFPE tissues generating high-quality sequencing data. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08068-1.
Collapse
Affiliation(s)
- Jin Sung Jang
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA. .,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Eileen Holicky
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA
| | - Julie Lau
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA
| | - Samantha McDonough
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mark Mutawe
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew J Koster
- Department of Internal Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth J Warrington
- Department of Internal Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Julie M Cuninngham
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Stabile Research Building, 200 First Street SW, Rochester, MN, 55905, USA. .,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
41
|
Skaftason A, Qu Y, Abdulla M, Nordlund J, Berglund M, Ednersson SB, Andersson PO, Enblad G, Amini RM, Rosenquist R, Mansouri L. Transcriptome sequencing of archived lymphoma specimens is feasible and clinically relevant using exome capture technology. Genes Chromosomes Cancer 2021; 61:27-36. [PMID: 34647650 DOI: 10.1002/gcc.23002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 11/06/2022] Open
Abstract
Formalin-fixed, paraffin-embedded (FFPE) specimens are an underutilized resource in medical research, particularly in the setting of transcriptome sequencing, as RNA from these samples is often degraded. We took advantage of an exome capture-based RNA-sequencing protocol to explore global gene expression in paired fresh-frozen (FF) and FFPE samples from 16 diffuse large B-cell lymphoma (DLBCL) patients. While FFPE samples generated fewer mapped reads compared to their FF counterparts, these reads captured the same library complexity and had a similar number of genes expressed on average. Furthermore, gene expression demonstrated a high correlation when comparing housekeeping genes only or across the entire transcriptome (r = 0.99 for both comparisons). Differences in gene expression were primarily seen in lowly expressed genes and genes with small or large coding sequences. Using cell-of-origin classifiers and clinically relevant gene expression signatures for DLBCL, FF, and FFPE samples from the same biopsy paired nearly perfectly in clustering analysis. This was further confirmed in a validation cohort of 50 FFPE DLBCL samples. In summary, we found the biological differences between tumors to be far greater than artifacts created as a result of degraded RNA. We conclude that exome capture transcriptome sequencing data from archival samples can confidently be used for cell-of-origin classification of DLBCL samples.
Collapse
Affiliation(s)
- Aron Skaftason
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ying Qu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Maysaa Abdulla
- Clinical and Experimental Pathology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jessica Nordlund
- Department of Medical Sciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mattias Berglund
- Experimental and Clinical Oncology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Susanne Bram Ednersson
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Ola Andersson
- Department of Medicine, Section of Hematology, South Älvsborg Hospital, Borås, Sweden.,Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gunilla Enblad
- Experimental and Clinical Oncology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rose-Marie Amini
- Clinical and Experimental Pathology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Larry Mansouri
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Sun L, McNulty SN, Evenson MJ, Zhu X, Robinson J, Mann PR, Duncavage EJ, Pfeifer JD. Clinical Implications of a Targeted RNA-Sequencing Panel in the Detection of Gene Fusions in Solid Tumors. J Mol Diagn 2021; 23:1749-1760. [PMID: 34562614 DOI: 10.1016/j.jmoldx.2021.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/09/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
The detection of recurrent gene fusions can help confirm diagnoses in solid tumors, particularly when the morphology and staining are unusual or nonspecific, and can guide therapeutic decisions. Although fluorescence in situ hybridization and PCR are often used to identify fusions, the rearrangement must be suspected, with only a few prioritized probes run. We hypothesized that the Illumina TruSight RNA Fusion Panel, which detects fusions of 507 genes and their partners, would uncover fusions with greater sensitivity than other approaches, leading to changes in diagnosis, prognosis, or therapy. Targeted RNA sequencing was performed on formalin-fixed, paraffin-embedded sarcoma and carcinoma cases in which fluorescence in situ hybridization, RT-PCR, or DNA-based sequencing was conducted during the diagnostic workup. Of 153 cases, 138 (90%) were sequenced with adequate quality control metrics. A total of 101 of 138 (73%) cases were concordant by RNA sequencing and the prior test method. RNA sequencing identified an additional 30 cases (22%) with fusions that were not detected by conventional methods. In seven cases (5%), the additional fusion information provided by RNA sequencing would have altered the diagnosis and management. A total of 19 novel fusion pairs (not previously described in the literature) were discovered (14%). Overall, the findings show that a targeted RNA-sequencing method can detect gene fusions in formalin-fixed, paraffin-embedded specimens with high sensitivity.
Collapse
Affiliation(s)
- Lulu Sun
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri.
| | - Samantha N McNulty
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael J Evenson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaopei Zhu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua Robinson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Patrick R Mann
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - John D Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
43
|
Doddapaneni H, Cregeen SJ, Sucgang R, Meng Q, Qin X, Avadhanula V, Chao H, Menon V, Nicholson E, Henke D, Piedra FA, Rajan A, Momin Z, Kottapalli K, Hoffman KL, Sedlazeck FJ, Metcalf G, Piedra PA, Muzny DM, Petrosino JF, Gibbs RA. Oligonucleotide capture sequencing of the SARS-CoV-2 genome and subgenomic fragments from COVID-19 individuals. PLoS One 2021; 16:e0244468. [PMID: 34432798 PMCID: PMC8386831 DOI: 10.1371/journal.pone.0244468] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
The newly emerged and rapidly spreading SARS-CoV-2 causes coronavirus disease 2019 (COVID-19). To facilitate a deeper understanding of the viral biology we developed a capture sequencing methodology to generate SARS-CoV-2 genomic and transcriptome sequences from infected patients. We utilized an oligonucleotide probe-set representing the full-length genome to obtain both genomic and transcriptome (subgenomic open reading frames [ORFs]) sequences from 45 SARS-CoV-2 clinical samples with varying viral titers. For samples with higher viral loads (cycle threshold value under 33, based on the CDC qPCR assay) complete genomes were generated. Analysis of junction reads revealed regions of differential transcriptional activity among samples. Mixed allelic frequencies along the 20kb ORF1ab gene in one sample, suggested the presence of a defective viral RNA species subpopulation maintained in mixture with functional RNA in one sample. The associated workflow is straightforward, and hybridization-based capture offers an effective and scalable approach for sequencing SARS-CoV-2 from patient samples.
Collapse
Affiliation(s)
- Harsha Doddapaneni
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sara Javornik Cregeen
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard Sucgang
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Qingchang Meng
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiang Qin
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hsu Chao
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Vipin Menon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Erin Nicholson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - David Henke
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zeineen Momin
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kavya Kottapalli
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kristi L. Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fritz J. Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ginger Metcalf
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna M. Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph F. Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
44
|
Shohdy KS, Bareja R, Sigouros M, Wilkes DC, Dorsaint P, Manohar J, Bockelman D, Xiang JZ, Kim R, Ohara K, Eng K, Mosquera JM, Elemento O, Sboner A, Alonso A, Faltas BM. Functional comparison of exome capture-based methods for transcriptomic profiling of formalin-fixed paraffin-embedded tumors. NPJ Genom Med 2021; 6:66. [PMID: 34385467 PMCID: PMC8360986 DOI: 10.1038/s41525-021-00231-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/26/2021] [Indexed: 11/08/2022] Open
Abstract
The availability of fresh frozen (FF) tissue is a barrier for implementing RNA sequencing (RNA-seq) in the clinic. The majority of clinical samples are stored as formalin-fixed, paraffin-embedded (FFPE) tissues. Exome capture platforms have been developed for RNA-seq from FFPE samples. However, these methods have not been systematically compared. We performed transcriptomic analysis of 32 FFPE tumor samples from 11 patients using three exome capture-based methods: Agilent SureSelect V6, TWIST NGS Exome, and IDT XGen Exome Research Panel. We compared these methods to the TruSeq RNA-seq of fresh frozen (FF-TruSeq) tumor samples from the same patients. We assessed the recovery of clinically relevant biological features. The Spearman's correlation coefficients between the global expression profiles of the three capture-based methods from FFPE and matched FF-TruSeq were high (rho = 0.72-0.9, p < 0.05). A significant correlation between the expression of key immune genes between individual capture-based methods and FF-TruSeq (rho = 0.76-0.88, p < 0.05) was observed. All exome capture-based methods reliably detected outlier expression of actionable gene transcripts, including ERBB2, MET, NTRK1, and PPARG. In urothelial cancer samples, the Agilent assay was associated with the highest molecular subtype concordance with FF-TruSeq (Cohen's k = 0.7, p < 0.01). The Agilent and IDT assays detected all the clinically relevant fusions that were initially identified in FF-TruSeq. All FFPE exome capture-based methods had comparable performance and concordance with FF-TruSeq. Our findings will enable the implementation of RNA-seq in the clinic to guide precision oncology approaches.
Collapse
Affiliation(s)
- Kyrillus S Shohdy
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Clinical Oncology, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Michael Sigouros
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David C Wilkes
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Princesca Dorsaint
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Jyothi Manohar
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Bockelman
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jenny Z Xiang
- Genomic Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Rob Kim
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kentaro Ohara
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kenneth Eng
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrea Sboner
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alicia Alonso
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Bishoy M Faltas
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
45
|
Abstract
Technological innovation and rapid reduction in sequencing costs have enabled the genomic profiling of hundreds of cancer-associated genes as a component of routine cancer care. Tumour genomic profiling can refine cancer subtype classification, identify which patients are most likely to benefit from systemic therapies and screen for germline variants that influence heritable cancer risk. Here, we discuss ongoing efforts to enhance the clinical utility of tumour genomic profiling by integrating tumour and germline analyses, characterizing allelic context and identifying mutational signatures that influence therapy response. We also discuss the potential clinical utility of more comprehensive whole-genome and whole-transcriptome sequencing and ultra-sensitive cell-free DNA profiling platforms, which allow for minimally invasive, serial analyses of tumour-derived DNA in blood.
Collapse
Affiliation(s)
- Debyani Chakravarty
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
46
|
Damavandi F, Wang W, Shen WZ, Cetinel S, Jordan T, Jovel J, Montemagno C, Wong GKS. Enrichment of low abundance DNA/RNA by oligonucleotide-clicked iron oxide nanoparticles. Sci Rep 2021; 11:13053. [PMID: 34158543 PMCID: PMC8219684 DOI: 10.1038/s41598-021-92376-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023] Open
Abstract
Detection of low abundance target DNA/RNA for clinical or research purposes is challenging because the target sequences can be hidden under a large background of human genomic or non-human metagenomic sequences. We describe a probe-based capture method to enrich for target sequences with DNA-clicked iron oxide nanoparticles. Our method was tested against commercial capture assays using streptavidin beads, on a set of probes derived from a common genotype of the hepatitis C virus. We showed that our method is more specific and sensitive, most likely due to the combination of an inert silica coating and a high density of DNA probes clicked to the nanoparticles. This facilitates target capture below the limits of detection for TaqMan qPCR, and we believe that this method has the potential to transform management of infectious diseases.
Collapse
Affiliation(s)
- Fereshte Damavandi
- Ingenuity Lab, 1-070C, 11421 Saskatchewan Drive NW, Edmonton, AB, T6G 2M9, Canada.,Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| | - Weiwei Wang
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Geneis Inc., Bldg A, 5 Guangshun North Street, Beijing, China
| | - Wei-Zheng Shen
- Ingenuity Lab, 1-070C, 11421 Saskatchewan Drive NW, Edmonton, AB, T6G 2M9, Canada.,Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| | - Sibel Cetinel
- Ingenuity Lab, 1-070C, 11421 Saskatchewan Drive NW, Edmonton, AB, T6G 2M9, Canada.,Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada.,Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, 34956, Turkey
| | - Tracy Jordan
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Juan Jovel
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Carlo Montemagno
- Ingenuity Lab, 1-070C, 11421 Saskatchewan Drive NW, Edmonton, AB, T6G 2M9, Canada.,Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| | - Gane Ka-Shu Wong
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada.
| |
Collapse
|
47
|
Haile S, Corbett RD, LeBlanc VG, Wei L, Pleasance S, Bilobram S, Nip KM, Brown K, Trinh E, Smith J, Trinh DL, Bala M, Chuah E, Coope RJN, Moore RA, Mungall AJ, Mungall KL, Zhao Y, Hirst M, Aparicio S, Birol I, Jones SJM, Marra MA. A Scalable Strand-Specific Protocol Enabling Full-Length Total RNA Sequencing From Single Cells. Front Genet 2021; 12:665888. [PMID: 34149808 PMCID: PMC8209500 DOI: 10.3389/fgene.2021.665888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
RNA sequencing (RNAseq) has been widely used to generate bulk gene expression measurements collected from pools of cells. Only relatively recently have single-cell RNAseq (scRNAseq) methods provided opportunities for gene expression analyses at the single-cell level, allowing researchers to study heterogeneous mixtures of cells at unprecedented resolution. Tumors tend to be composed of heterogeneous cellular mixtures and are frequently the subjects of such analyses. Extensive method developments have led to several protocols for scRNAseq but, owing to the small amounts of RNA in single cells, technical constraints have required compromises. For example, the majority of scRNAseq methods are limited to sequencing only the 3' or 5' termini of transcripts. Other protocols that facilitate full-length transcript profiling tend to capture only polyadenylated mRNAs and are generally limited to processing only 96 cells at a time. Here, we address these limitations and present a novel protocol that allows for the high-throughput sequencing of full-length, total RNA at single-cell resolution. We demonstrate that our method produced strand-specific sequencing data for both polyadenylated and non-polyadenylated transcripts, enabled the profiling of transcript regions beyond only transcript termini, and yielded data rich enough to allow identification of cell types from heterogeneous biological samples.
Collapse
Affiliation(s)
- Simon Haile
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Richard D Corbett
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Veronique G LeBlanc
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Lisa Wei
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Stephen Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Steve Bilobram
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Ka Ming Nip
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Kirstin Brown
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Eva Trinh
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Jillian Smith
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Diane L Trinh
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Miruna Bala
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Robin J N Coope
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Martin Hirst
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Samuel Aparicio
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
48
|
Overman M, Javle M, Davis RE, Vats P, Kumar-Sinha C, Xiao L, Mettu NB, Parra ER, Benson AB, Lopez CD, Munugalavadla V, Patel P, Tao L, Neelapu S, Maitra A. Randomized phase II study of the Bruton tyrosine kinase inhibitor acalabrutinib, alone or with pembrolizumab in patients with advanced pancreatic cancer. J Immunother Cancer 2021; 8:jitc-2020-000587. [PMID: 32114502 PMCID: PMC7057435 DOI: 10.1136/jitc-2020-000587] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/18/2022] Open
Abstract
Background The immunosuppressive desmoplastic stroma of pancreatic cancer represents a major hurdle to developing an effective immune response. Preclinical studies in pancreatic cancer have demonstrated promising anti-tumor activity with Bruton tyrosine kinase (BTK) inhibition combined with programmed cell death receptor-1 (PD-1) blockade. Methods This was a phase II, multicenter, open-label, randomized (1:1) clinical trial evaluating the BTK inhibitor acalabrutinib, alone (monotherapy) or in combination with the anti-PD-1 antibody pembrolizumab (combination therapy). Eligible patients were adults with histologically confirmed metastatic or locally advanced unresectable pancreatic ductal adenocarcinoma with an Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≤1 who had received at least one prior systemic therapy. Oral acalabrutinib 100 mg twice daily was administered with or without intravenous pembrolizumab 200 mg on day 1 of each 3-week cycle. Peripheral blood was analyzed for changes in immune markers, and tumors from exceptional responders were molecularly analyzed. Results A total of 77 patients were enrolled (37 monotherapy; 40 combination therapy) with a median age of 64 years; 77% had an ECOG PS of 1. The median number of prior therapies was 3 (range 1–6). Grade 3–4 treatment-related adverse events were seen in 14.3% of patients in the monotherapy arm and 15.8% of those in the combination therapy arm. The overall response rate and disease control rate were 0% and 14.3% with monotherapy and 7.9% and 21.1% with combination therapy, respectively. Median progression-free survival was 1.4 months in both arms. Peripheral blood flow analysis demonstrated consistent reductions in granulocytic (CD15+) myeloid-derived suppressor cells (MDSCs) over time. Two exceptional responders were found to be microsatellite stable with low tumor mutation burden, low neoantigen load and no defects in the homologous DNA repair pathway. Conclusions The combination of acalabrutinib and pembrolizumab was well tolerated, but limited clinical activity was seen with either acalabrutinib monotherapy or combination therapy. Peripheral reductions in MDSCs were seen. Efforts to understand and target the pancreatic tumor microenvironment should continue. Trial registration number NCT02362048.
Collapse
Affiliation(s)
- Michael Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard E Davis
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pankaj Vats
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lianchun Xiao
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Niharika B Mettu
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Al B Benson
- Department of Medicine, Hematology Oncology Division, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, Illinois, USA
| | - Charles D Lopez
- Department of Oncology, School of Medicine, Oregon Health and Science University Foundation, Portland, Oregon, USA
| | | | - Priti Patel
- Acerta Pharma LLC, Redwood City, California, USA
| | - Lin Tao
- Acerta Pharma LLC, Redwood City, California, USA
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
49
|
Wagner MJ, Lyons YA, Siedel JH, Dood R, Nagaraja AS, Haemmerle M, Mangala LS, Chanana P, Lazar AJ, Wang WL, Ravi V, Holland EC, Sood AK. Combined VEGFR and MAPK pathway inhibition in angiosarcoma. Sci Rep 2021; 11:9362. [PMID: 33931674 PMCID: PMC8087824 DOI: 10.1038/s41598-021-88703-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Angiosarcoma is an aggressive malignancy of endothelial cells that carries a high mortality rate. Cytotoxic chemotherapy can elicit clinical responses, but the duration of response is limited. Sequencing reveals multiple mutations in angiogenesis pathways in angiosarcomas, particularly in vascular endothelial growth factor (VEGFR) and mitogen-activated protein kinase (MAPK) signaling. We aimed to determine the biological relevance of these pathways in angiosarcoma. Tissue microarray consisting of clinical formalin-fixed paraffin embedded tissue archival samples were stained for phospho- extracellular signal-regulated kinase (p-ERK) with immunohistochemistry. Angiosarcoma cell lines were treated with the mitogen-activated protein kinase kinase (MEK) inhibitor trametinib, pan-VEGFR inhibitor cediranib, or combined trametinib and cediranib and viability was assessed. Reverse phase protein array (RPPA) was performed to assess multiple oncogenic protein pathways. SVR angiosarcoma cells were grown in vivo and gene expression effects of treatment were assessed with whole exome RNA sequencing. MAPK signaling was found active in over half of clinical angiosarcoma samples. Inhibition of MAPK signaling with the MEK inhibitor trametinib decreased the viability of angiosarcoma cells. Combined inhibition of the VEGF and MAPK pathways with cediranib and trametinib had an additive effect in in vitro models, and a combinatorial effect in an in vivo model. Combined treatment led to smaller tumors than treatment with either agent alone. RNA-seq demonstrated distinct expression signatures between the trametinib treated tumors and those treated with both trametinib and cediranib. These results indicate a clinical study of combined VEGFR and MEK inhibition in angiosarcoma is warranted.
Collapse
Affiliation(s)
- Michael J Wagner
- Division of Medical Oncology, University of Washington, 825 Eastlake Ave E, Seattle, WA, 98109, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.
| | - Yasmin A Lyons
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| | - Jean H Siedel
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| | - Robert Dood
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| | - Archana S Nagaraja
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| | - Monika Haemmerle
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
- Section for Experimental Pathology, Medical Faculty, Institute of Pathology, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| | - Pritha Chanana
- Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Alexander J Lazar
- Department of Pathology, UT MD Anderson Cancer Center, Houston, USA
- Department of Genomic Medicine, UT MD Anderson Cancer Center, Houston, USA
| | - Wei-Lien Wang
- Department of Pathology, UT MD Anderson Cancer Center, Houston, USA
| | - Vinod Ravi
- Sarcoma Medical Oncology, UT MD Anderson Cancer Center, Houston, USA
| | - Eric C Holland
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine and Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
50
|
Li Z, Jin Y, Zou Q, Shi X, Wu Q, Lin Z, He Q, Huang G, Qi S. Integrated genomic and transcriptomic analysis suggests KRT18 mutation and MTAP are key genetic alterations related to the prognosis between astrocytoma and glioblastoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:713. [PMID: 33987411 PMCID: PMC8106028 DOI: 10.21037/atm-21-1317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Astrocytoma and glioblastoma (GBM) are the two main subtypes of glioma, with the 2016 World Health Organization Classification of Tumors of the Central Nervous System (CNS WHO) classifying them into different grades. GBM is the most malignant among all CNS tumors with a 5-year survival rate of less than 5%. Although the prognosis of patients with astrocytoma is better than that of GBM in general, patients with anaplastic astrocytoma (AA) and isocitrate dehydrogenase (IDH) wild type have a similar prognosis as GBM and entail a high risk of progression. Exploring the molecular driving force behind the malignant phenotype of astrocytoma and GBM will help explain the diversity of glioma and discover new drug targets. Methods We enrolled 12 patients with astrocytoma and 12 patients with GBM and performed whole-exome sequencing (WES) and RNA sequencing analysis on tumor samples from the patients. Results We found that the somatic mutation of KRT18, which is associated with cell apoptosis and adhesion by interacting with receptor 1-associated protein (TRADD) and pinin, was significantly enriched in astrocytoma, but rare in GBM. Copy number loss of MTAP, which is closely related to a poor prognosis of glioma, was found to be significantly enriched in GBM. In addition, different somatic copy number alteration (SCNA), gene expression, and immune cell infiltration patterns between astrocytoma and GBM were found. Conclusions This study revealed the distinct characteristics of astrocytoma and GBM at the DNA and RNA level. Somatic mutation of KRT18 and copy number loss of MTAP, two key genetic alterative genes in astrocytoma and GBM, have the potential to become therapeutic targets in glioma.
Collapse
Affiliation(s)
- Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yinghui Jin
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingping Zou
- GenomiCare Biotechnology (Shanghai) Co., Ltd., Shanghai, China
| | - Xiaofeng Shi
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, China
| | - Qianchao Wu
- GenomiCare Biotechnology (Shanghai) Co., Ltd., Shanghai, China
| | - Zhiying Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qun He
- GenomiCare Biotechnology (Shanghai) Co., Ltd., Shanghai, China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|