1
|
Faupel-Badger J, Kohaar I, Bahl M, Chan AT, Campbell JD, Ding L, De Marzo AM, Maitra A, Merrick DT, Hawk ET, Wistuba II, Ghobrial IM, Lippman SM, Lu KH, Lawler M, Kay NE, Tlsty TD, Rebbeck TR, Srivastava S. Defining precancer: a grand challenge for the cancer community. Nat Rev Cancer 2024; 24:792-809. [PMID: 39354069 DOI: 10.1038/s41568-024-00744-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
The term 'precancer' typically refers to an early stage of neoplastic development that is distinguishable from normal tissue owing to molecular and phenotypic alterations, resulting in abnormal cells that are at least partially self-sustaining and function outside of normal cellular cues that constrain cell proliferation and survival. Although such cells are often histologically distinct from both the corresponding normal and invasive cancer cells of the same tissue origin, defining precancer remains a challenge for both the research and clinical communities. Once sufficient molecular and phenotypic changes have occurred in the precancer, the tissue is identified as a 'cancer' by a histopathologist. While even diagnosing cancer can at times be challenging, the determination of invasive cancer is generally less ambiguous and suggests a high likelihood of and potential for metastatic disease. The 'hallmarks of cancer' set out the fundamental organizing principles of malignant transformation but exactly how many of these hallmarks and in what configuration they define precancer has not been clearly and consistently determined. In this Expert Recommendation, we provide a starting point for a conceptual framework for defining precancer, which is based on molecular, pathological, clinical and epidemiological criteria, with the goal of advancing our understanding of the initial changes that occur and opportunities to intervene at the earliest possible time point.
Collapse
Affiliation(s)
| | - Indu Kohaar
- Division of Cancer Prevention, National Cancer Institute, NIH, Rockville, MD, USA
| | - Manisha Bahl
- Division of Breast Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joshua D Campbell
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Li Ding
- Department of Medicine and Genetics, McDonnell Genome Institute, and Siteman Cancer Center, Washington University in St Louis, Saint Louis, MO, USA
| | - Angelo M De Marzo
- Department of Pathology, Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel T Merrick
- Division of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest T Hawk
- Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott M Lippman
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Karen H Lu
- Department of Gynecological Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mark Lawler
- Patrick G Johnson Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Thea D Tlsty
- Department of Medicine and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Timothy R Rebbeck
- Dana-Farber Cancer Institute and Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Sudhir Srivastava
- Division of Cancer Prevention, National Cancer Institute, NIH, Rockville, MD, USA.
| |
Collapse
|
2
|
Faraji F, Ramirez SI, Clubb L, Sato K, Burghi V, Hoang TS, Officer A, Anguiano Quiroz PY, Galloway WM, Mikulski Z, Medetgul-Ernar K, Marangoni P, Jones KB, Molinolo AA, Kim K, Sakaguchi K, Califano JA, Smith Q, Goren A, Klein OD, Tamayo P, Gutkind JS. YAP-Driven Oral Epithelial Stem Cell Malignant Reprogramming at Single Cell Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550427. [PMID: 37546810 PMCID: PMC10402053 DOI: 10.1101/2023.07.24.550427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo, however, remains elusive. Here we employ spatiotemporally controlled oncogene activation and tumor suppressor inhibition together with multiomics to unveil the processes underlying oral epithelial progenitor cell reprogramming into tumor initiating cells (TIC) at single cell resolution. TIC displayed a distinct stem-like state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal (pEMT) invasive gene programs. YAP-mediated TIC programs included the activation of oncogenic transcriptional networks and mTOR signaling, and the recruitment of myeloid cells to the invasive front contributing to tumor infiltration. TIC transcriptional programs are conserved in human head and neck cancer and associated with poor patient survival. These findings illuminate processes underlying cancer initiation at single cell resolution, and identify candidate targets for early cancer detection and prevention.
Collapse
|
3
|
Bychkovsky BL, Myers S, Warren LEG, De Placido P, Parsons HA. Ductal Carcinoma In Situ. Hematol Oncol Clin North Am 2024; 38:831-849. [PMID: 38960507 DOI: 10.1016/j.hoc.2024.05.014] [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: 07/05/2024]
Abstract
In breast cancer (BC) pathogenesis models, normal cells acquire somatic mutations and there is a stepwise progression from high-risk lesions and ductal carcinoma in situ to invasive cancer. The precancer biology of mammary tissue warrants better characterization to understand how different BC subtypes emerge. Primary methods for BC prevention or risk reduction include lifestyle changes, surgery, and chemoprevention. Surgical intervention for BC prevention involves risk-reducing prophylactic mastectomy, typically performed either synchronously with the treatment of a primary tumor or as a bilateral procedure in high-risk women. Chemoprevention with endocrine therapy carries adherence-limiting toxicity.
Collapse
Affiliation(s)
- Brittany L Bychkovsky
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sara Myers
- Harvard Medical School, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | - Laura E G Warren
- Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pietro De Placido
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Heather A Parsons
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
Shakerian N, Darzi-Eslam E, Afsharnoori F, Bana N, Noorabad Ghahroodi F, Tarin M, Mard-Soltani M, Khalesi B, Hashemi ZS, Khalili S. Therapeutic and diagnostic applications of exosomes in colorectal cancer. Med Oncol 2024; 41:203. [PMID: 39031221 DOI: 10.1007/s12032-024-02440-3] [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: 05/01/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024]
Abstract
Exosomes play a key role in colorectal cancer (CRC) related processes. This review explores the various functions of exosomes in CRC and their potential as diagnostic markers, therapeutic targets, and drug delivery vehicles. Exosomal long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) significantly influence CRC progression. Specific exosomal lncRNAs are linked to drug resistance and tumor growth, respectively, highlighting their therapeutic potential. Similarly, miRNAs like miR-21, miR-10b, and miR-92a-3p, carried by exosomes, contribute to chemotherapy resistance by altering signaling pathways and gene expression in CRC cells. The review also discusses exosomes' utility in CRC diagnosis. Exosomes from cancer cells have distinct molecular signatures compared to healthy cells, making them reliable biomarkers. Specific exosomal lncRNAs (e.g., CRNDE-h) and miRNAs (e.g., miR-17-92a) have shown effectiveness in early CRC detection and monitoring of treatment responses. Furthermore, exosomes show promise as vehicles for targeted drug delivery. The potential of mesenchymal stem cell (MSC)-derived exosomes in CRC treatment is also noted, with their role varying from promoting to inhibiting tumor progression. The application of multi-omics approaches to exosome research is highlighted, emphasizing the potential for discovering novel CRC biomarkers through comprehensive genomic, transcriptomic, proteomic, and metabolomic analyses. The review also explores the emerging field of exosome-based vaccines, which utilize exosomes' natural properties to elicit strong immune responses. In conclusion, exosomes represent a promising frontier in CRC research, offering new avenues for diagnosis, treatment, and prevention. Their unique properties and versatile functions underscore the need for continued investigation into their clinical applications and underlying mechanisms.
Collapse
Affiliation(s)
- Neda Shakerian
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Elham Darzi-Eslam
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Afsharnoori
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nikoo Bana
- Kish International Campus, University of Teheran, Tehran, Iran
| | - Faezeh Noorabad Ghahroodi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Education and Extension Organization, Razi Vaccine and Serum Research Institute, Agricultural Research, Karaj, 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
5
|
Teschendorff AE. Computational single-cell methods for predicting cancer risk. Biochem Soc Trans 2024; 52:1503-1514. [PMID: 38856037 DOI: 10.1042/bst20231488] [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: 01/29/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024]
Abstract
Despite recent biotechnological breakthroughs, cancer risk prediction remains a formidable computational and experimental challenge. Addressing it is critical in order to improve prevention, early detection and survival rates. Here, I briefly summarize some key emerging theoretical and computational challenges as well as recent computational advances that promise to help realize the goals of cancer-risk prediction. The focus is on computational strategies based on single-cell data, in particular on bottom-up network modeling approaches that aim to estimate cancer stemness and dedifferentiation at single-cell resolution from a systems-biological perspective. I will describe two promising methods, a tissue and cell-lineage independent one based on the concept of diffusion network entropy, and a tissue and cell-lineage specific one that uses transcription factor regulons. Application of these tools to single-cell and single-nucleus RNA-seq data from stages prior to invasive cancer reveal that they can successfully delineate the heterogeneous inter-cellular cancer-risk landscape, identifying those cells that are more likely to turn cancerous. Bottom-up systems biological modeling of single-cell omic data is a novel computational analysis paradigm that promises to facilitate the development of preventive, early detection and cancer-risk prediction strategies.
Collapse
Affiliation(s)
- Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
6
|
Emens LA, Romero PJ, Anderson AC, Bruno TC, Capitini CM, Collyar D, Gulley JL, Hwu P, Posey AD, Silk AW, Wargo JA. Challenges and opportunities in cancer immunotherapy: a Society for Immunotherapy of Cancer (SITC) strategic vision. J Immunother Cancer 2024; 12:e009063. [PMID: 38901879 PMCID: PMC11191773 DOI: 10.1136/jitc-2024-009063] [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] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Ana Carrizosa Anderson
- The Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tullia C Bruno
- Department of Immunology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian M Capitini
- Department of Pediatrics and Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Deborah Collyar
- Patient Advocates in Research (PAIR), Danville, California, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Avery D Posey
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
7
|
Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D, Wu C. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Target Ther 2024; 9:149. [PMID: 38890350 PMCID: PMC11189549 DOI: 10.1038/s41392-024-01848-7] [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: 01/01/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 06/20/2024] Open
Abstract
Tumorigenesis is a multistep process, with oncogenic mutations in a normal cell conferring clonal advantage as the initial event. However, despite pervasive somatic mutations and clonal expansion in normal tissues, their transformation into cancer remains a rare event, indicating the presence of additional driver events for progression to an irreversible, highly heterogeneous, and invasive lesion. Recently, researchers are emphasizing the mechanisms of environmental tumor risk factors and epigenetic alterations that are profoundly influencing early clonal expansion and malignant evolution, independently of inducing mutations. Additionally, clonal evolution in tumorigenesis reflects a multifaceted interplay between cell-intrinsic identities and various cell-extrinsic factors that exert selective pressures to either restrain uncontrolled proliferation or allow specific clones to progress into tumors. However, the mechanisms by which driver events induce both intrinsic cellular competency and remodel environmental stress to facilitate malignant transformation are not fully understood. In this review, we summarize the genetic, epigenetic, and external driver events, and their effects on the co-evolution of the transformed cells and their ecosystem during tumor initiation and early malignant evolution. A deeper understanding of the earliest molecular events holds promise for translational applications, predicting individuals at high-risk of tumor and developing strategies to intercept malignant transformation.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyi Xiao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yonglin Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyu Wang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Lingxuan Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Changping Laboratory, 100021, Beijing, China
| | - Yanrong Shen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, 100006, Beijing, China.
| |
Collapse
|
8
|
Zhu T, Tong H, Du Z, Beck S, Teschendorff AE. An improved epigenetic counter to track mitotic age in normal and precancerous tissues. Nat Commun 2024; 15:4211. [PMID: 38760334 PMCID: PMC11101651 DOI: 10.1038/s41467-024-48649-8] [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: 09/24/2023] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
The cumulative number of stem cell divisions in a tissue, known as mitotic age, is thought to be a major determinant of cancer-risk. Somatic mutational and DNA methylation (DNAm) clocks are promising tools to molecularly track mitotic age, yet their relationship is underexplored and their potential for cancer risk prediction in normal tissues remains to be demonstrated. Here we build and validate an improved pan-tissue DNAm counter of total mitotic age called stemTOC. We demonstrate that stemTOC's mitotic age proxy increases with the tumor cell-of-origin fraction in each of 15 cancer-types, in precancerous lesions, and in normal tissues exposed to major cancer risk factors. Extensive benchmarking against 6 other mitotic counters shows that stemTOC compares favorably, specially in the preinvasive and normal-tissue contexts. By cross-correlating stemTOC to two clock-like somatic mutational signatures, we confirm the mitotic-like nature of only one of these. Our data points towards DNAm as a promising molecular substrate for detecting mitotic-age increases in normal tissues and precancerous lesions, and hence for developing cancer-risk prediction strategies.
Collapse
Affiliation(s)
- Tianyu Zhu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Huige Tong
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Zhaozhen Du
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Stephan Beck
- Medical Genomics Group, UCL Cancer Institute, University College London, 72 Huntley Street, WC1E 6BT, London, UK
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
9
|
Teschendorff AE. On epigenetic stochasticity, entropy and cancer risk. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230054. [PMID: 38432318 PMCID: PMC10909509 DOI: 10.1098/rstb.2023.0054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/26/2023] [Indexed: 03/05/2024] Open
Abstract
Epigenetic changes are known to accrue in normal cells as a result of ageing and cumulative exposure to cancer risk factors. Increasing evidence points towards age-related epigenetic changes being acquired in a quasi-stochastic manner, and that they may play a causal role in cancer development. Here, I describe the quasi-stochastic nature of DNA methylation (DNAm) changes in ageing cells as well as in normal cells at risk of neoplastic transformation, discussing the implications of this stochasticity for developing cancer risk prediction strategies, and in particular, how it may require a conceptual paradigm shift in how we select cancer risk markers. I also describe the mounting evidence that a significant proportion of DNAm changes in ageing and cancer development are related to cell proliferation, reflecting tissue-turnover and the opportunity this offers for predicting cancer risk via the development of epigenetic mitotic-like clocks. Finally, I describe how age-associated DNAm changes may be causally implicated in cancer development via an irreversible suppression of tissue-specific transcription factors that increases epigenetic and transcriptomic entropy, promoting a more plastic yet aberrant cancer stem-cell state. This article is part of a discussion meeting issue 'Causes and consequences of stochastic processes in development and disease'.
Collapse
Affiliation(s)
- Andrew E. Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, People's Republic of China
| |
Collapse
|
10
|
Luo R, Liu J, Wen J, Zhou X. Single-cell Landscape of Malignant Transition: Unraveling Cancer Cell-of-Origin and Heterogeneous Tissue Microenvironment. RESEARCH SQUARE 2024:rs.3.rs-4085185. [PMID: 38645221 PMCID: PMC11030487 DOI: 10.21203/rs.3.rs-4085185/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Understanding disease progression and sophisticated tumor ecosystems is imperative for investigating tumorigenesis mechanisms and developing novel prevention strategies. Here, we dissected heterogeneous microenvironments during malignant transitions by leveraging data from 1396 samples spanning 13 major tissues. Within transitional stem-like subpopulations highly enriched in precancers and cancers, we identified 30 recurring cellular states strongly linked to malignancy, including hypoxia and epithelial senescence, revealing a high degree of plasticity in epithelial stem cells. By characterizing dynamics in stem-cell crosstalk with the microenvironment along the pseudotime axis, we found differential roles of ANXA1 at different stages of tumor development. In precancerous stages, reduced ANXA1 levels promoted monocyte differentiation toward M1 macrophages and inflammatory responses, whereas during malignant progression, upregulated ANXA1 fostered M2 macrophage polarization and cancer-associated fibroblast transformation by increasing TGF-β production. Our spatiotemporal analysis further provided insights into mechanisms responsible for immunosuppression and a potential target to control evolution of precancer and mitigate the risk for cancer development.
Collapse
Affiliation(s)
| | - Jiajia Liu
- The University of Texas Health Science Center at Houston
| | - Jianguo Wen
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston
| | - Xiaobo Zhou
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston
| |
Collapse
|
11
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [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: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
12
|
Buradagunta CS, Arsang-Jang S, Massat B, Thapa B, Patek V, D'Souza A, Auer P, Urrutia R, Janz S, Dhakal B, Hari P, Dong J. Identification of novel loci for multiple myeloma when comparing with its precursor condition monoclonal gammopathy of unknown significance. Leukemia 2024; 38:383-385. [PMID: 37919604 PMCID: PMC11326531 DOI: 10.1038/s41375-023-02078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Affiliation(s)
| | - Shahram Arsang-Jang
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ben Massat
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bicky Thapa
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Victoria Patek
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anita D'Souza
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Paul Auer
- Division of Biostatistics, Institute for Health & Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Siegfried Janz
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Binod Dhakal
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Parameswaran Hari
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jing Dong
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
| |
Collapse
|
13
|
Allan Z, Liu DS, Lee MM, Tie J, Clemons NJ. A Practical Approach to Interpreting Circulating Tumor DNA in the Management of Gastrointestinal Cancers. Clin Chem 2024; 70:49-59. [PMID: 38175583 DOI: 10.1093/clinchem/hvad188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/19/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND There is accumulating evidence supporting the clinical use of circulating tumor DNA (ctDNA) in solid tumors, especially in different types of gastrointestinal cancer. As such, appraisal of the current and potential clinical utility of ctDNA is needed to guide clinicians in decision-making to facilitate its general applicability. CONTENT In this review, we firstly discuss considerations surrounding specimen collection, processing, storage, and analysis, which affect reporting and interpretation of results. Secondly, we evaluate a selection of studies on colorectal, esophago-gastric, and pancreatic cancer to determine the level of evidence for the use of ctDNA in disease screening, detection of molecular residual disease (MRD) and disease recurrence during surveillance, assessment of therapy response, and guiding targeted therapy. Lastly, we highlight current limitations in the clinical utility of ctDNA and future directions. SUMMARY Current evidence of ctDNA in gastrointestinal cancer is promising but varies depending on its specific clinical role and cancer type. Larger prospective trials are needed to validate different aspects of ctDNA clinical utility, and standardization of collection protocols, analytical assays, and reporting guidelines should be considered to facilitate its wider applicability.
Collapse
Affiliation(s)
- Zexi Allan
- Division of Cancer Research, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David S Liu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Upper Gastrointestinal Surgery Unit, Division of Surgery, Anaesthesia, and Procedural Medicine, Austin Health, Heidelberg, Victoria, Australia
| | - Margaret M Lee
- Division of Personalised Oncology, the Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jeanne Tie
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Division of Personalised Oncology, the Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Nicholas J Clemons
- Division of Cancer Research, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Hanna GJ, Villa A, Nandi SP, Shi R, ONeill A, Liu M, Quinn CT, Treister NS, Sroussi HY, Vacharotayangul P, Goguen LA, Annino DJ, Rettig EM, Jo VY, Wong KS, Lizotte P, Paweletz CP, Uppaluri R, Haddad RI, Cohen EEW, Alexandrov LB, William WN, Lippman SM, Woo SB. Nivolumab for Patients With High-Risk Oral Leukoplakia: A Nonrandomized Controlled Trial. JAMA Oncol 2024; 10:32-41. [PMID: 37971722 PMCID: PMC10654930 DOI: 10.1001/jamaoncol.2023.4853] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/07/2023] [Indexed: 11/19/2023]
Abstract
Importance Proliferative verrucous leukoplakia (PVL) is an aggressive oral precancerous disease characterized by a high risk of transformation to invasive oral squamous cell carcinoma (OSCC), and no therapies have been shown to affect its natural history. A recent study of the PVL immune landscape revealed a cytotoxic T-cell-rich microenvironment, providing strong rationale to investigate immune checkpoint therapy. Objective To determine the safety and clinical activity of anti-programmed cell death 1 protein (PD-1) therapy to treat high-risk PVL. Design, Setting, and Participants This nonrandomized, open-label, phase 2 clinical trial was conducted from January 2019 to December 2021 at a single academic medical center; median (range) follow-up was 21.1 (5.4-43.6) months. Participants were a population-based sample of patients with PVL (multifocal, contiguous, or a single lesion ≥4 cm with any degree of dysplasia). Intervention Patients underwent pretreatment biopsy (1-3 sites) and then received 4 doses of nivolumab (480 mg intravenously) every 28 days, followed by rebiopsy and intraoral photographs at each visit. Main Outcomes and Measures The primary end point was the change in composite score (size and degree of dysplasia) from before to after treatment (major response [MR]: >80% decrease in score; partial response: 40%-80% decrease). Secondary analyses included immune-related adverse events, cancer-free survival (CFS), PD-1 ligand 1 (PD-L1) expression, 9p21.3 deletion, and other exploratory immunologic and genomic associations of response. Results A total of 33 patients were enrolled (median [range] age, 63 [32-80] years; 18 [55%] were female), including 8 (24%) with previously resected early-stage OSCC. Twelve patients (36%) (95% CI, 20.4%-54.8%) had a response by composite score (3 MRs [9%]), 4 had progressive disease (>10% composite score increase, or cancer). Nine patients (27%) developed OSCC during the trial, with a 2-year CFS of 73% (95% CI, 53%-86%). Two patients (6%) discontinued because of toxic effects; 7 (21%) experienced grade 3 to 4 immune-related adverse events. PD-L1 combined positive scores were not associated with response or CFS. Of 20 whole-exome sequenced patients, all 6 patients who had progression to OSCC after nivolumab treatment exhibited 9p21.3 somatic copy-number loss on pretreatment biopsy, while only 4 of the 14 patients (29%) who did not develop OSCC had 9p21.3 loss. Conclusions and Relevance This immune checkpoint therapy precancer nonrandomized clinical trial met its prespecified response end point, suggesting potential clinical activity for nivolumab in high-risk PVL. Findings identified immunogenomic associations to inform future trials in this precancerous disease with unmet medical need that has been difficult to study. Trial Registration ClinicalTrials.gov Identifier: NCT03692325.
Collapse
Affiliation(s)
- Glenn J. Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alessandro Villa
- Miami Cancer Institute and Herbert Wertheim College of Medicine, Florida International University, Miami
| | - Shuvro P. Nandi
- Moores Cancer Center, UC San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, California
| | - Ruichao Shi
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anne ONeill
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mofei Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles T. Quinn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nathaniel S. Treister
- Division of Oral Medicine and Dentistry, Dana-Farber Cancer Institute and Brigham & Women’s Hospital, Boston, Massachusetts
| | - Herve Y. Sroussi
- Division of Oral Medicine and Dentistry, Dana-Farber Cancer Institute and Brigham & Women’s Hospital, Boston, Massachusetts
| | - Piamkamon Vacharotayangul
- Division of Oral Medicine and Dentistry, Dana-Farber Cancer Institute and Brigham & Women’s Hospital, Boston, Massachusetts
| | - Laura A. Goguen
- Division of Otolaryngology–Head and Neck Surgery, Brigham & Women’s Hospital and Head and Neck Surgical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Donald J. Annino
- Division of Otolaryngology–Head and Neck Surgery, Brigham & Women’s Hospital and Head and Neck Surgical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eleni M. Rettig
- Division of Otolaryngology–Head and Neck Surgery, Brigham & Women’s Hospital and Head and Neck Surgical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vickie Y. Jo
- Department of Pathology, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Kristine S. Wong
- Department of Pathology, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Patrick Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P. Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ravindra Uppaluri
- Division of Otolaryngology–Head and Neck Surgery, Brigham & Women’s Hospital and Head and Neck Surgical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert I. Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Ludmil B. Alexandrov
- Moores Cancer Center, UC San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, California
- Department of Bioengineering, UC San Diego, La Jolla, California
| | - William N. William
- Oncology Center, Hospital BP, a Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | | | - Sook-bin Woo
- Division of Oral Medicine and Dentistry, Dana-Farber Cancer Institute and Brigham & Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
15
|
Weeks LD, Ebert BL. Causes and consequences of clonal hematopoiesis. Blood 2023; 142:2235-2246. [PMID: 37931207 PMCID: PMC10862247 DOI: 10.1182/blood.2023022222] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
ABSTRACT Clonal hematopoiesis (CH) is described as the outsized contribution of expanded clones of hematopoietic stem and progenitor cells (HSPCs) to blood cell production. The prevalence of CH increases dramatically with age. CH can be caused by somatic mutations in individual genes or by gains and/or losses of larger chromosomal segments. CH is a premalignant state; the somatic mutations detected in CH are the initiating mutations for hematologic malignancies, and CH is a strong predictor of the development of blood cancers. Moreover, CH is associated with nonmalignant disorders and increased overall mortality. The somatic mutations that drive clonal expansion of HSPCs can alter the function of terminally differentiated blood cells, including the release of elevated levels of inflammatory cytokines. These cytokines may then contribute to a broad range of inflammatory disorders that increase in prevalence with age. Specific somatic mutations in the peripheral blood in coordination with blood count parameters can powerfully predict the development of hematologic malignancies and overall mortality in CH. In this review, we summarize the current understanding of CH nosology and origins. We provide an overview of available tools for risk stratification and discuss management strategies for patients with CH presenting to hematology clinics.
Collapse
Affiliation(s)
- Lachelle D. Weeks
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Early Detection and Interception of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Early Detection and Interception of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| |
Collapse
|
16
|
Sullivan BA, Qin X, Redding TS, Weiss D, Upchurch J, Sims KJ, Dominitz JA, Stone A, Ear B, Williams CD, Lieberman DA, Hauser ER. Colorectal Cancer Polygenic Risk Score Is Associated With Screening Colonoscopy Findings but Not Follow-Up Outcomes. GASTRO HEP ADVANCES 2023; 3:151-161. [PMID: 39129957 PMCID: PMC11307447 DOI: 10.1016/j.gastha.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Colorectal cancer (CRC) polygenic risk scores (PRS) may help personalize CRC prevention strategies. We investigated whether an existing PRS was associated with advanced neoplasia (AN) in a population undergoing screening and follow-up colonoscopy. Methods We evaluated 10-year outcomes in the Cooperative Studies Program #380 screening colonoscopy cohort, which includes a biorepository of selected individuals with baseline AN (defined as CRC or adenoma ≥10 mm or villous histology, or high-grade dysplasia) and matched individuals without AN. A PRS was constructed from 136 prespecified CRC-risk single nucleotide polymorphisms. Multivariate logistic regression was used to evaluate the PRS for associations with AN prevalence at baseline screening colonoscopy or incident AN in participants with at least one follow-up colonoscopy. Results The PRS was associated with AN risk at baseline screening colonoscopy (P = .004). Participants in the lowest PRS quintile had more than a 70% decreased risk of AN at baseline (odds ratio 0.29, 95% confidence interval 0.14-0.58; P < .001) compared to participants with a PRS in the middle quintile. Using a PRS cut-off of more than the first quintile to indicate need for colonoscopy as primary screening, the sensitivity for detecting AN at baseline is 91.8%. We did not observe a relationship between the PRS and incident AN during follow-up (P = .28). Conclusion A PRS could identify individuals at low risk for prevalent AN. Ongoing work will determine whether this PRS can identify a subset of individuals at sufficiently low risk who could safely delay or be reassured about noninvasive screening. Otherwise, more research is needed to augment these genetic tools to predict incident AN during long-term follow-up.
Collapse
Affiliation(s)
- Brian A. Sullivan
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
- Division of Gastroenterology, Duke University, Durham, North Carolina
| | - Xuejun Qin
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
- Division of Gastroenterology, Duke University, Durham, North Carolina
| | - Thomas S. Redding
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
| | - David Weiss
- Cooperative Studies Program Coordinating Center, Perry Point VA Medical Center, Perry Point, Maryland
| | - Julie Upchurch
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
| | - Kellie J. Sims
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
| | - Jason A. Dominitz
- Division of Gastroenterology, VA Puget Sound Health Care System, Seattle, Washington
- Division of Gastroenterology, University of Washington School of Medicine, Seattle, Washington
| | - Anjanette Stone
- Cooperative Studies Program Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Health System, Little Rock, Arkansas
| | - Belinda Ear
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
| | - Christina D. Williams
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
- Division of Gastroenterology, Duke University, Durham, North Carolina
| | - David A. Lieberman
- Division of Gastroenterology, VA Portland Health Care System, Portland, Oregon
- Division of Gastroenterology, Oregon Health & Science University, Portland, Oregon
| | - Elizabeth R. Hauser
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, North Carolina
- Division of Gastroenterology, Duke University, Durham, North Carolina
| |
Collapse
|
17
|
Chien H, Tsai C, Young C, Lee Y, Liao C, Yeh C, Chao A, Cho K, Chen C, Huang S. Single-nucleotide polymorphism at alcohol dehydrogenase 1B: A susceptible gene marker in oro-/hypopharyngeal cancers from genome-wide association study. Cancer Med 2023; 12:19174-19187. [PMID: 37706329 PMCID: PMC10557853 DOI: 10.1002/cam4.6506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/19/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023] Open
Abstract
INTRODUCTION In the era of precision preventive medicine, susceptible genetic markers for oro-/hypopharyngeal squamous cell carcinoma (OPSCC) have been investigated for genome-wide associations. MATERIALS AND METHODS A case-control study including 659 male head and neck squamous cell carcinoma (HNSCC) patients, including 331 oropharyngeal cancer, treated between March 1996 and December 2016 and 2400 normal controls was performed. A single-nucleotide polymorphism (SNP) array was used to determine genetic loci that increase susceptibility to OPSCC. RESULTS We analyzed the allele frequencies of 664,994 autosomal SNPs in 659 HNSCC cases; 7 SNPs scattered in loci of chromosomes 5, 7, 9, 11, and 19 were significant in genome-wide association analysis (Pc < 1.0669 × 10-7 ). In OPSCCs (n = 331), two clustered regions in chromosomes 4 and 6 were significantly different from the controls. We successfully identified a missense alteration of the SNP region in alcohol dehydrogenase 1B (ADH1B) (https://genome.ucsc.edu; hg38); the top correlated locus was rs1229984 (p = 1 × 10-11 ). Adjusted for environmental exposure, including smoking, alcohol, and areca quid, a region in chromosome 12, related to alcohol metabolism, was found to independently increase the susceptibility to OPSCC. The ADH1B rs1229984 AA genotype had better overall survival compared to the AG and GG genotypes (p = 0.042) in OPSCC. The GG genotype in rs1229984 was significantly associated with a younger age of onset than other genotypes (p = 0.001 and <0.001, respectively) in OPSCC patients who consumed alcohol. CONCLUSION ADH1B was an important genetic locus that significantly correlated with the development of OPSCCs and patient survival.
Collapse
Affiliation(s)
- Hui‐Tzu Chien
- Department of Nutrition and Health SciencesChang Gung University of Science and TechnologyTaoyuanTaiwan
- Geriatric and Long‐Term Care Research CenterChang Gung University of Science and TechnologyTaoyuanTaiwan
| | - Chia‐Lung Tsai
- Genomic Medicine Research Core LaboratoryChang Gung Memorial Hospital, Linkou BranchTaoyuanTaiwan
| | - Chi‐Kuan Young
- Department of Otolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital, Keelung BranchKeelungTaiwan
- Medical College, Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Yun‐Shien Lee
- Department of Nutrition and Health SciencesChang Gung University of Science and TechnologyTaoyuanTaiwan
- Department of BiotechnologyMing Chuan UniversityTaoyuanTaiwan
| | - Chun‐Ta Liao
- Department of Otolaryngology, Head and Neck SurgeryChang Gung Memorial HospitalLinkouTaiwan
| | - Chih‐Ching Yeh
- Master Program in Applied Molecular Epidemiology, College of Public HealthTaipei Medical UniversityTaipeiTaiwan
- School of Public Health, College of Public HealthTaipei Medical UniversityTaipeiTaiwan
- Cancer Center, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Angel Chao
- Department of Obstetrics and GynecologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Kai‐Lun Cho
- Department of Otolaryngology, Head and Neck SurgeryChang Gung Memorial HospitalLinkouTaiwan
| | - Ching‐Han Chen
- School of Medicine, Chang Gung Medical CollegeChang Gung UniversityTaoyuanTaiwan
| | - Shiang‐Fu Huang
- Department of Otolaryngology, Head and Neck SurgeryChang Gung Memorial HospitalLinkouTaiwan
- Graduate Institute of Clinical Medical SciencesChang Gung UniversityTaoyuanTaiwan
| |
Collapse
|
18
|
Rane JK, Frankell AM, Weeden CE, Swanton C. Clonal Evolution in Healthy and Premalignant Tissues: Implications for Early Cancer Interception Strategies. Cancer Prev Res (Phila) 2023; 16:369-378. [PMID: 36930945 PMCID: PMC7614725 DOI: 10.1158/1940-6207.capr-22-0469] [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: 01/13/2023] [Revised: 02/17/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
Histologically normal human tissues accumulate significant mutational burden with age. The extent and spectra of mutagenesis are comparable both in rapidly proliferating and post-mitotic tissues and in stem cells compared with their differentiated progeny. Some of these mutations provide increased fitness, giving rise to clones which, at times, can replace the entire surface area of tissues. Compared with cancer, somatic mutations in histologically normal tissues are primarily single-nucleotide variations. Interestingly though, the presence of these mutations and positive clonal selection in isolation remains a poor indicator of potential future cancer transformation in solid tissues. Common clonally expanded mutations in histologically normal tissues also do not always represent the most frequent early mutations in cancers of corresponding tissues, indicating differences in selection pressures. Preliminary evidence implies that stroma and immune system co-evolve with age, which may impact selection dynamics. In this review, we will explore the mutational landscape of histologically normal and premalignant human somatic tissues in detail and discuss cell-intrinsic and environmental factors that can determine the fate of positively selected mutations within them. Precisely pinpointing these determinants of cancer transformation would aid development of early cancer interventional and prevention strategies.
Collapse
Affiliation(s)
- Jayant K. Rane
- University College London Cancer Institute, London, UK
- Department of Clinical Oncology, University College London Hospitals, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Alexander M. Frankell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare E. Weeden
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals, London, UK
| |
Collapse
|
19
|
Bowen CM, Deng N, Reyes-Uribe L, Parra ER, Rocha P, Solis LM, Wistuba II, Sepeda VO, Vornik L, Perloff M, Szabo E, Umar A, Sinha KM, Brown PH, Vilar E. Naproxen chemoprevention induces proliferation of cytotoxic lymphocytes in Lynch Syndrome colorectal mucosa. Front Immunol 2023; 14:1162669. [PMID: 37207208 PMCID: PMC10189148 DOI: 10.3389/fimmu.2023.1162669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Background Recent clinical trial data from Lynch Syndrome (LS) carriers demonstrated that naproxen administered for 6-months is a safe primary chemoprevention that promotes activation of different resident immune cell types without increasing lymphoid cellularity. While intriguing, the precise immune cell types enriched by naproxen remained unanswered. Here, we have utilized cutting-edge technology to elucidate the immune cell types activated by naproxen in mucosal tissue of LS patients. Methods Normal colorectal mucosa samples (pre- and post-treatment) from a subset of patients enrolled in the randomized and placebo-controlled 'Naproxen Study' were obtained and subjected to a tissue microarray for image mass cytometry (IMC) analysis. IMC data was processed using tissue segmentation and functional markers to ascertain cell type abundance. Computational outputs were then used to quantitatively compare immune cell abundance in pre- and post-naproxen specimens. Results Using data-driven exploration, unsupervised clustering identified four populations of immune cell types with statistically significant changes between treatment and control groups. These four populations collectively describe a unique cell population of proliferating lymphocytes within mucosal samples from LS patients exposed to naproxen. Conclusions Our findings show that daily exposure of naproxen promotes T-cell proliferation in the colonic mucosa, which paves way for developing combination of immunoprevention strategies including naproxen for LS patients.
Collapse
Affiliation(s)
- Charles M. Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nan Deng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Edwin Roger Parra
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pedro Rocha
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M. Solis
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ignacio I. Wistuba
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Valerie O. Sepeda
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lana Vornik
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marjorie Perloff
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Krishna M. Sinha
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
20
|
Esai Selvan M, Onel K, Gnjatic S, Klein RJ, Gümüş ZH. Germline rare deleterious variant load alters cancer risk, age of onset and tumor characteristics. NPJ Precis Oncol 2023; 7:13. [PMID: 36707626 PMCID: PMC9883433 DOI: 10.1038/s41698-023-00354-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Recent studies show that rare, deleterious variants (RDVs) in certain genes are critical determinants of heritable cancer risk. To more comprehensively understand RDVs, we performed the largest-to-date germline variant calling analysis in a case-control setting for a multi-cancer association study from whole-exome sequencing data of 20,789 participants, split into discovery and validation cohorts. We confirm and extend known associations between cancer risk and germline RDVs in specific gene-sets, including DNA repair (OR = 1.50; p-value = 8.30e-07; 95% CI: 1.28-1.77), cancer predisposition (OR = 1.51; p-value = 4.58e-08; 95% CI: 1.30-1.75), and somatic cancer drivers (OR = 1.46; p-value = 4.04e-06; 95% CI: 1.24-1.72). Furthermore, personal RDV load in these gene-sets associated with increased risk, younger age of onset, increased M1 macrophages in tumor and, increased tumor mutational burden in specific cancers. Our findings can be used towards identifying high-risk individuals, who can then benefit from increased surveillance, earlier screening, and treatments that exploit their tumor characteristics, improving prognosis.
Collapse
Affiliation(s)
- Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kenan Onel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Clarkson BDS, Johnson RK, Bingel C, Lothaller C, Howe CL. Preservation of antigen-specific responses in cryopreserved CD4 + and CD8 + T cells expanded with IL-2 and IL-7. J Transl Autoimmun 2022; 5:100173. [PMID: 36467614 PMCID: PMC9713293 DOI: 10.1016/j.jtauto.2022.100173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
Objectives We sought to develop medium throughput standard operating procedures for screening cryopreserved human peripheral blood mononuclear cells (PBMCs) for CD4+ and CD8+ T cell responses to potential autoantigens. Methods Dendritic cells were loaded with a peptide cocktail from ubiquitous viruses or full-length viral protein antigens and cocultured with autologous T cells. We measured expression of surface activation markers on T cells by flow cytometry and cytometry by time of flight 24-72 h later. We tested responses among T cells freshly isolated from healthy control PBMCs, cryopreserved T cells, and T cells derived from a variety of T cell expansion protocols. We also compared the transcriptional profile of CD8+ T cells rested with interleukin (IL)7 for 48 h after 1) initial thawing, 2) expansion, and 3) secondary cryopreservation/thawing of expanded cells. To generate competent antigen presenting cells from PBMCs, we promoted differentiation of PBMCs into dendritic cells with granulocyte macrophage colony stimulating factor and IL-4. Results We observed robust dendritic cell differentiation from human PBMCs treated with 50 ng/mL GM-CSF and 20 ng/mL IL-4 in as little as 3 days. Dendritic cell purity was substantially increased by magnetically enriching for CD14+ monocytes prior to differentiation. We also measured antigen-dependent T cell activation in DC-T cell cocultures. However, polyclonal expansion of T cells with anti-CD3/antiCD28 abolished antigen-dependent upregulation of CD69 in our assay despite minimal transcriptional differences between rested CD8+ T cells before and after expansion. Furthermore, resting these expanded T cells in IL-2, IL-7 or IL-15 did not restore the antigen dependent responses. In contrast, T cells that were initially expanded with IL-2 + IL-7 rather than plate bound anti-CD3 + anti-CD28 retained responsiveness to antigen stimulation and these responses strongly correlated with responses measured at initial thawing. Significance While screening techniques for potential pathological autoantibodies have come a long way, comparable full-length protein target assays for screening patient T cells at medium throughput are noticeably lacking due to technical hurdles. Here we advance techniques that should have broad applicability to translational studies investigating cell mediated immunity in infectious or autoimmune diseases. Future studies are aimed at investigating possible CD8+ T cell autoantigens in MS and other CNS autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin DS. Clarkson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA,Corresponding author. Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN, 55905.
| | | | - Corinna Bingel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center, Heidelberg, Germany
| | | | - Charles L. Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
22
|
Solary E, Blanc P, Boutros M, Girvalaki C, Locatelli F, Medema RH, Nagy P, Tabernero J. UNCAN.eu, a European Initiative to UNderstand CANcer. Cancer Discov 2022; 12:2504-2508. [PMID: 36074491 DOI: 10.1158/2159-8290.cd-22-0970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
"UNCAN.eu" refers to a collective European effort seeking to enable a leap forward in our understanding of cancer. This initiative, which includes the creation of a European cancer research data hub, will pave the way to new advances in cancer care. Starting on September 1, 2022, a 15-month coordination and support action will generate a blueprint for UNCAN.eu. Here, we summarize the cancer research issues that the blueprint will propose to tackle at the European level.
Collapse
Affiliation(s)
- Eric Solary
- Université Paris-Saclay and INSERM, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Michael Boutros
- German Cancer Research Center (DKFZ) and Heidelberg University, Heidelberg, Germany
| | | | - Franco Locatelli
- Bambino Gesù Children's Hospital, University of the Sacred Heart, Rome, Italy
| | - Rene H Medema
- Oncode Institute and The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Péter Nagy
- National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
23
|
Olive GN, Yang IA, Marshall H, Bowman RV, Fong KM. More than meets the eye. Eur Respir J 2022; 60:60/3/2200763. [PMID: 36109046 DOI: 10.1183/13993003.00763-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Gerard N Olive
- Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia.,UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ian A Yang
- Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia.,UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Henry Marshall
- Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia.,UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Rayleen V Bowman
- Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia.,UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Kwun M Fong
- Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia .,UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
24
|
Agnoletto C, Volinia S. Mitochondria dysfunction in circulating tumor cells. Front Oncol 2022; 12:947479. [PMID: 35992829 PMCID: PMC9386562 DOI: 10.3389/fonc.2022.947479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 12/16/2022] Open
Abstract
Circulating tumor cells (CTCs) represent a subset of heterogeneous cells, which, once released from a tumor site, have the potential to give rise to metastasis in secondary sites. Recent research focused on the attempt to detect and characterize these rare cells in the circulation, and advancements in defining their molecular profile have been reported in diverse tumor species, with potential implications for clinical applications. Of note, metabolic alterations, involving mitochondria, have been implicated in the metastatic process, as key determinants in the transition of tumor cells to a mesenchymal or stemness-like phenotype, in drug resistance, and in induction of apoptosis. This review aimed to briefly analyse the most recent knowledge relative to mitochondria dysfunction in CTCs, and to envision implications of altered mitochondria in CTCs for a potential utility in clinics.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Rete Oncologica Veneta (ROV), Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Stefano Volinia
- Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Biological and Chemical Research Centre (CNBCh UW), University of Warsaw, Warsaw, Poland
- Center of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
25
|
Cancer Prevention Prioritized at AACR Annual Meeting and a New Working Group. Cancer Prev Res (Phila) 2022; 15:475-479. [DOI: 10.1158/1940-6207.capr-22-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Scientific advances in the late 19th century set the stage for progress in understanding and treating cancer, a disease that was previously considered almost hopeless. One hundred years later, cancer prevention is becoming an increasingly important focus for oncology research. New tools and ideas bring to the field some extremely promising molecular, organizational, social, and political approaches, which were a focus of the American Association for Cancer Research 2022 Annual Meeting and of the newly launched AACR Cancer Prevention Working Group (CPWG). We are moving toward precision prevention, better tools for early detection and for risk assessment, the use of a Precancer Atlas, unveiling of new biomarkers. Besides improving lifestyle, by avoiding risk factors such as tobacco use, excessive UV exposure, infectious agents, as well as poor dietary habits, lack of exercise, overweight, and obesity, many other factors can impact cancer risk, which is a warning to consider a multifaceted molecular but also social approach. Gender, ethnicity, geographic, and economic lines are associated with disparities in prevention, which we want to overcome. Here we summarize some challenges and priorities in cancer prevention emerging from the work of AACR and CPWG.
Collapse
|
26
|
Cao Y, Duan H, Su A, Xu L, Lai B. A pan-cancer analysis confirms PTPN11's potential as a prognostic and immunological biomarker. Aging (Albany NY) 2022; 14:5590-5610. [PMID: 35802774 PMCID: PMC9320542 DOI: 10.18632/aging.204171] [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] [Received: 03/29/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
Protein tyrosine phosphatase, non-receptor type 11 (PTPN11) is a multifunctional tyrosine phosphatase and has a significant part in many types of tumors. As of yet, neither the expression profile of PTPN11 nor its significance in pan-cancer diagnosis has been clarified. With the assistance of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we have comprehensively mapped the expression profiles, prognostic significance, genetic alteration, phosphorylation status, infiltration of immune cells, and functional properties of PTPN11 in 33 human tumors. There was an inconsistent expression of PTPN11 in different tumors, and the alteration of PTPN11 expression predicted the survival outcomes of cancer patients. A significant association was found between the genetic alteration levels of PTPN11 and some tumor predictions. Besides, the reduced PTPN11 phosphorylation levels were observed in breast cancer, clear cell RCC, head and neck carcinoma, and lung adenocarcinoma (LUAD). Furthermore, there was a significant association between PTPN11 expression and infiltration of cancer-associated fibroblasts and endothelial cells, along with tumor mutation burden, microsatellite instability, mismatch repair genes, and immunoregulators. Finally, pathway enrichment analysis demonstrated that PTPN11-associated terms and pathways were involved in malignancy. Taken together, PTPN11 may become a new biomarker and target for cancer therapy.
Collapse
Affiliation(s)
- Yapeng Cao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Haixia Duan
- Department of Reproduction Gynecology, Northwest Women and Children's Hospital, Xi'an, Shaanxi 710061, China
| | - Ailing Su
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Liran Xu
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| |
Collapse
|
27
|
Liu T, Zhao X, Lin Y, Luo Q, Zhang S, Xi Y, Chen Y, Lin L, Fan W, Yang J, Ma Y, Maity AK, Huang Y, Wang J, Chang J, Lin D, Teschendorff AE, Wu C. Computational identification of preneoplastic cells displaying high stemness and risk of cancer progression. Cancer Res 2022; 82:2520-2537. [PMID: 35536873 DOI: 10.1158/0008-5472.can-22-0668] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/05/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022]
Abstract
Evidence points towards the differentiation state of cells as a marker of cancer risk and progression. Measuring the differentiation state of single cells in a preneoplastic population could thus enable novel strategies for early detection and risk prediction. Recent maps of somatic mutagenesis in normal tissues from young healthy individuals have revealed cancer driver mutations, indicating that these do not correlate well with differentiation state and that other molecular events also contribute to cancer development. We hypothesized that the differentiation state of single cells can be measured by estimating the regulatory activity of the transcription factors (TFs) that control differentiation within that cell lineage. To this end, we present a novel computational method called CancerStemID that estimates a stemness index of cells from single-cell RNA-Seq data. CancerStemID is validated in two human esophageal squamous cell carcinoma (ESCC) cohorts, demonstrating how it can identify undifferentiated preneoplastic cells whose transcriptomic state is overrepresented in invasive cancer. Spatial transcriptomics and whole-genome bisulfite sequencing demonstrated that differentiation activity of tissue-specific TFs was decreased in cancer cells compared to the basal cell-of-origin layer and established that differentiation state correlated with differential DNA methylation at the promoters of these TFs, independently of underlying NOTCH1 and TP53 mutations. The findings were replicated in a mouse model of ESCC development, and the broad applicability of CancerStemID to other cancer-types was demonstrated. In summary, these data support an epigenetic stem-cell model of oncogenesis and highlight a novel computational strategy to identify stem-like preneoplastic cells that undergo positive selection.
Collapse
Affiliation(s)
- Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuan Zhao
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Lin
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University (PKU), Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiyi Xi
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yamei Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyi Fan
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Yang
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuling Ma
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alok K Maity
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanyi Huang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University (PKU), Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jiang Chang
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- CAMS Oxford Institute (COI), Chinese Academy of Medical Sciences, Beijing, China
- CAMS key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Brenner DR, Poirier A, Woods RR, Ellison LF, Billette JM, Demers AA, Zhang SX, Yao C, Finley C, Fitzgerald N, Saint-Jacques N, Shack L, Turner D, Holmes E. Projected estimates of cancer in Canada in 2022. CMAJ 2022; 194:E601-E607. [PMID: 35500919 PMCID: PMC9067380 DOI: 10.1503/cmaj.212097] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Regular cancer surveillance is crucial for understanding where progress is being made and where more must be done. We sought to provide an overview of the expected burden of cancer in Canada in 2022. METHODS We obtained data on new cancer incidence from the National Cancer Incidence Reporting System (1984-1991) and Canadian Cancer Registry (1992-2018). Mortality data (1984-2019) were obtained from the Canadian Vital Statistics - Death Database. We projected cancer incidence and mortality counts and rates to 2022 for 22 cancer types by sex and province or territory. Rates were age standardized to the 2011 Canadian standard population. RESULTS An estimated 233 900 new cancer cases and 85 100 cancer deaths are expected in Canada in 2022. We expect the most commonly diagnosed cancers to be lung overall (30 000), breast in females (28 600) and prostate in males (24 600). We also expect lung cancer to be the leading cause of cancer death, accounting for 24.3% of all cancer deaths, followed by colorectal (11.0%), pancreatic (6.7%) and breast cancers (6.5%). Incidence and mortality rates are generally expected to be higher in the eastern provinces of Canada than the western provinces. INTERPRETATION Although overall cancer rates are declining, the number of cases and deaths continues to climb, owing to population growth and the aging population. The projected high burden of lung cancer indicates a need for increased tobacco control and improvements in early detection and treatment. Success in breast and colorectal cancer screening and treatment likely account for the continued decline in their burden. The limited progress in early detection and new treatments for pancreatic cancer explains why it is expected to be the third leading cause of cancer death in Canada.
Collapse
Affiliation(s)
- Darren R Brenner
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont.
| | - Abbey Poirier
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Ryan R Woods
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Larry F Ellison
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Jean-Michel Billette
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Alain A Demers
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Shary Xinyu Zhang
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Chunhe Yao
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Christian Finley
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Natalie Fitzgerald
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Nathalie Saint-Jacques
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Lorraine Shack
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Donna Turner
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | - Elizabeth Holmes
- Departments of Oncology and Community Health Sciences (Brenner), Cumming School of Medicine, University of Calgary; Department of Cancer Epidemiology and Prevention Research (Brenner, Poirier), CancerControl Alberta, Alberta Health Services, Calgary, Alta.; Population Oncology (Woods), BC Cancer, Vancouver, BC; Centre for Population Health Data (Ellison, Billette, Zhang, Yao), Statistics Canada; Centre for Surveillance and Applied Research (Demers), Public Health Agency of Canada, Ottawa, Ont.; Departments of Surgery (Finley), McMaster University, St. Joseph's Health Care Centre, Hamilton, Ont.; Performance (Fitzgerald), Canadian Partnership Against Cancer, Toronto, Ont.; Nova Scotia Health Cancer Care Program (Saint-Jacques), Halifax, NS; Population Oncology (Shack), Cancer Care Manitoba, Winnipeg, Man.; Surveillance and Reporting (Turner), Cancer Care Alberta, Calgary, Alta.; Cancer Information and Policy Department (Holmes), Canadian Cancer Society, Toronto, Ont
| | | |
Collapse
|
29
|
Bouaoud J, Bossi P, Elkabets M, Schmitz S, van Kempen LC, Martinez P, Jagadeeshan S, Breuskin I, Puppels GJ, Hoffmann C, Hunter KD, Simon C, Machiels JP, Grégoire V, Bertolus C, Brakenhoff RH, Koljenović S, Saintigny P. Unmet Needs and Perspectives in Oral Cancer Prevention. Cancers (Basel) 2022; 14:cancers14071815. [PMID: 35406587 PMCID: PMC8997728 DOI: 10.3390/cancers14071815] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Oral potentially malignant disorders (OPMD) may precede oral squamous cell carcinoma (OSCC). Reported rates of malignant transformation of OPMD range from 3 to 50%. While some clinical, histological, and molecular factors have been associated with a high-risk OPMD, they are, to date, insufficiently accurate for treatment decision-making. Moreover, this range highlights differences in the clinical definition of OPMD, variation in follow-up periods, and molecular and biological heterogeneity of OPMD. Finally, while treatment of OPMD may improve outcome, standard therapy has been shown to be ineffective to prevent OSCC development in patients with OPMD. In this perspective paper, several experts discuss the main challenges in oral cancer prevention, in particular the need to (i) to define an OPMD classification system by integrating new pathological and molecular characteristics, aiming (ii) to better identify OPMD at high risk of malignant transformation, and (iii) to develop treatment strategies to eradicate OPMD or prevent malignant transformation.
Collapse
Affiliation(s)
- Jebrane Bouaoud
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France;
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France; (V.G.); (C.B.)
- Department of Maxillo-Facial Surgery, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
- Correspondence: (J.B.); (P.S.)
| | - Paolo Bossi
- Medical Oncology, ASST Spedali Civili Brescia, I-25064 Brescia, Italy;
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, I-25123 Brescia, Italy
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.E.); (S.J.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sandra Schmitz
- Department of Medical Oncology and Head and Neck Surgery, Institut Roi Albert II, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (S.S.); (J.-P.M.)
| | - Léon C. van Kempen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9712 CP Groningen, The Netherlands;
| | - Pierre Martinez
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France;
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France; (V.G.); (C.B.)
| | - Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.E.); (S.J.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ingrid Breuskin
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, F-94805 Villejuif, France;
| | - Gerwin J. Puppels
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Room Ee-1691, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands;
| | - Caroline Hoffmann
- INSERM U932 Research Unit, Department of Surgery, Institut Curie, PSL Research University, F-75006 Paris, France;
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
| | - Christian Simon
- Department of Otolaryngology and Head and Neck Surgery, Lausanne University Hospital, 1011 Lausanne, Switzerland;
| | - Jean-Pascal Machiels
- Department of Medical Oncology and Head and Neck Surgery, Institut Roi Albert II, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (S.S.); (J.-P.M.)
| | - Vincent Grégoire
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France; (V.G.); (C.B.)
- Radiation Oncology Department, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France
| | - Chloé Bertolus
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France; (V.G.); (C.B.)
- Department of Maxillo-Facial Surgery, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Ruud H. Brakenhoff
- Cancer Center Amsterdam, Section Head and Neck Cancer Biology & Immunology, Otolaryngology and Head and Neck Surgery, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands;
| | - Senada Koljenović
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Pierre Saintigny
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France;
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, F-69008 Lyon, France; (V.G.); (C.B.)
- Department of Medical Oncology, Centre Léon Bérard, Université Claude Bernard Lyon 1, University Lyon, 28 Promenade Léa et Napoléon Bullukian, F-69008 Lyon, France
- Correspondence: (J.B.); (P.S.)
| |
Collapse
|
30
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
31
|
Conserved MicroRNAs in Human Nasopharynx Tissue Samples from Swabs Are Differentially Expressed in Response to SARS-CoV-2. Genes (Basel) 2022; 13:genes13020348. [PMID: 35205390 PMCID: PMC8871708 DOI: 10.3390/genes13020348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
The use of high-throughput small RNA sequencing is well established as a technique to unveil the miRNAs in various tissues. The miRNA profiles are different between infected and non-infected tissues. We compare the SARS-CoV-2 positive and SARS-CoV-2 negative RNA samples extracted from human nasopharynx tissue samples to show different miRNA profiles. We explored differentially expressed miRNAs in response to SARS-CoV-2 in the RNA extracted from nasopharynx tissues of 10 SARS-CoV-2-positive and 10 SARS-CoV-2-negative patients. miRNAs were identified by small RNA sequencing, and the expression levels of selected miRNAs were validated by real-time RT-PCR. We identified 943 conserved miRNAs, likely generated through posttranscriptional modifications. The identified miRNAs were expressed in both RNA groups, NegS and PosS: miR-148a, miR-21, miR-34c, miR-34b, and miR-342. The most differentially expressed miRNA was miR-21, which is likely closely linked to the presence of SARS-CoV-2 in nasopharynx tissues. Our results contribute to further understanding the role of miRNAs in SARS-CoV-2 pathogenesis, which may be crucial for understanding disease symptom development in humans.
Collapse
|
32
|
Precision Medicine for Hepatocellular Carcinoma: Clinical Perspective. J Pers Med 2022; 12:jpm12020149. [PMID: 35207638 PMCID: PMC8879044 DOI: 10.3390/jpm12020149] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the major malignant diseases worldwide, characterized by growing incidence and high mortality rates despite apparent improvements in surveillance programs, diagnostic and treatment procedures, molecular therapies, and numerous research initiatives. Most HCCs occur in patients with liver cirrhosis, and the competing mortality risks from the tumor and the cirrhosis should be considered. Presently, previously identified risk factors, such as hepatitis virus infection, hepatic inflammation and fibrosis, and metabolic syndrome, may be used as chemoprevention targets. The application of precision medicine for HCC management challenges the one-size-fits-all concept; moreover, patients should no longer be treated entirely according to the histology of their tumor but based on molecular targets specific to their tumor biology. Next-generation sequencing emphasizes HCC molecular heterogeneity and aids our comprehension of possible vulnerabilities that can be exploited. Moreover, genetic sequencing as part of a precision medicine concept may work as a promising tool for postoperative cancer monitoring. The use of genetic and epigenetic markers to identify therapeutic vulnerability could change the diagnosis and treatment of HCC, which so far was based on Barcelona clinic liver cancer (BCLC) staging. In daily clinical practice, the shift from a stage-oriented to a therapeutic-oriented approach is needed to direct the choice of HCC treatment toward the potentially most effective option on an individual basis. The important factor in precision medicine is the promotion of patient management based on the individual approach, knowing that the final decision must be approved by a multidisciplinary expert team.
Collapse
|
33
|
Liang S, Willis J, Dou J, Mohanty V, Huang Y, Vilar E, Chen K. Sensei: how many samples to tell a change in cell type abundance? BMC Bioinformatics 2022; 23:2. [PMID: 34983369 PMCID: PMC8728970 DOI: 10.1186/s12859-021-04526-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022] Open
Abstract
Cellular heterogeneity underlies cancer evolution and metastasis. Advances in single-cell technologies such as single-cell RNA sequencing and mass cytometry have enabled interrogation of cell type-specific expression profiles and abundance across heterogeneous cancer samples obtained from clinical trials and preclinical studies. However, challenges remain in determining sample sizes needed for ascertaining changes in cell type abundances in a controlled study. To address this statistical challenge, we have developed a new approach, named Sensei, to determine the number of samples and the number of cells that are required to ascertain such changes between two groups of samples in single-cell studies. Sensei expands the t-test and models the cell abundances using a beta-binomial distribution. We evaluate the mathematical accuracy of Sensei and provide practical guidelines on over 20 cell types in over 30 cancer types based on knowledge acquired from the cancer cell atlas (TCGA) and prior single-cell studies. We provide a web application to enable user-friendly study design via https://kchen-lab.github.io/sensei/table_beta.html .
Collapse
Affiliation(s)
- Shaoheng Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Computer Science, Rice University, Houston, TX USA
| | - Jason Willis
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jinzhuang Dou
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Yuefan Huang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
34
|
Rosendahl Huber A, Van Hoeck A, Van Boxtel R. The Mutagenic Impact of Environmental Exposures in Human Cells and Cancer: Imprints Through Time. Front Genet 2021; 12:760039. [PMID: 34745228 PMCID: PMC8565797 DOI: 10.3389/fgene.2021.760039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
During life, the DNA of our cells is continuously exposed to external damaging processes. Despite the activity of various repair mechanisms, DNA damage eventually results in the accumulation of mutations in the genomes of our cells. Oncogenic mutations are at the root of carcinogenesis, and carcinogenic agents are often highly mutagenic. Over the past decade, whole genome sequencing data of healthy and tumor tissues have revealed how cells in our body gradually accumulate mutations because of exposure to various mutagenic processes. Dissection of mutation profiles based on the type and context specificities of the altered bases has revealed a variety of signatures that reflect past exposure to environmental mutagens, ranging from chemotherapeutic drugs to genotoxic gut bacteria. In this review, we discuss the latest knowledge on somatic mutation accumulation in human cells, and how environmental mutagenic factors further shape the mutation landscapes of tissues. In addition, not all carcinogenic agents induce mutations, which may point to alternative tumor-promoting mechanisms, such as altered clonal selection dynamics. In short, we provide an overview of how environmental factors induce mutations in the DNA of our healthy cells and how this contributes to carcinogenesis. A better understanding of how environmental mutagens shape the genomes of our cells can help to identify potential preventable causes of cancer.
Collapse
Affiliation(s)
- Axel Rosendahl Huber
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Arne Van Hoeck
- Oncode Institute, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Ruben Van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
35
|
An Epigenetic Perspective on Intra-Tumour Heterogeneity: Novel Insights and New Challenges from Multiple Fields. Cancers (Basel) 2021; 13:cancers13194969. [PMID: 34638453 PMCID: PMC8508087 DOI: 10.3390/cancers13194969] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Although research on cancer biology in recent decades has unveiled the main genetic perturbations driving the onset of tumorigenesis, we are still far from properly treating this disease without the occurrence of drug resistance and metastatic burden. This achievement is hampered by the onset of intra-tumour heterogeneity (ITH), which increases cancer cell fitness and plasticity, thereby fostering cell adaptation to foreign environments and stimuli. In this review, we discuss the contribution of the epigenetic factors in sustaining ITH and their interplay with the tumour microenvironment. We also highlight the recent technological advancements that are contributing to defining the epigenetic mechanisms governing tumour heterogeneity at the single-cell level. Abstract Cancer is a group of heterogeneous diseases that results from the occurrence of genetic alterations combined with epigenetic changes and environmental stimuli that increase cancer cell plasticity. Indeed, multiple cancer cell populations coexist within the same tumour, favouring cancer progression and metastatic dissemination as well as drug resistance, thereby representing a major obstacle for treatment. Epigenetic changes contribute to the onset of intra-tumour heterogeneity (ITH) as they facilitate cell adaptation to perturbation of the tumour microenvironment. Despite being its central role, the intrinsic multi-layered and reversible epigenetic pattern limits the possibility to uniquely determine its contribution to ITH. In this review, we first describe the major epigenetic mechanisms involved in tumourigenesis and then discuss how single-cell-based approaches contribute to dissecting the key role of epigenetic changes in tumour heterogeneity. Furthermore, we highlight the importance of dissecting the interplay between genetics, epigenetics, and tumour microenvironments to decipher the molecular mechanisms governing tumour progression and drug resistance.
Collapse
|
36
|
Maoz A, Merenstein C, Koga Y, Potter A, Gower AC, Liu G, Zhang S, Liu H, Stevenson C, Spira A, Reid ME, Campbell JD, Mazzilli SA, Lenburg ME, Beane J. Elevated T cell repertoire diversity is associated with progression of lung squamous cell premalignant lesions. J Immunother Cancer 2021; 9:jitc-2021-002647. [PMID: 34580161 PMCID: PMC8477334 DOI: 10.1136/jitc-2021-002647] [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] [Accepted: 08/03/2021] [Indexed: 11/21/2022] Open
Abstract
Objective The immune response to invasive carcinoma has been the focus of published work, but little is known about the adaptive immune response to bronchial premalignant lesions (PMLs), precursors of lung squamous cell carcinoma. This study was designed to characterize the T cell receptor (TCR) repertoire in PMLs and its association with clinical, pathological, and molecular features. Methods Endobronchial biopsies (n=295) and brushings (n=137) from high-risk subjects (n=50), undergoing lung cancer screening at approximately 1-year intervals via autofluorescence bronchoscopy and CT, were profiled by RNA-seq. We applied the TCR Repertoire Utilities for Solid Tissue/Tumor tool to the RNA-seq data to identify TCR CDR3 sequences across all samples. In the biopsies, we measured the correlation of TCR diversity with previously derived immune-associated PML transcriptional signatures and PML outcome. We also quantified the spatial and temporal distribution of shared and clonally expanded TCRs. Using the biopsies and brushes, the ratio of private (ie, found in one patient only) and public (ie, found in two or more patients) TCRs was quantified, and the CDR3 sequences were compared with those found in curated databases with known antigen specificities. Results We detected 39,303 unique TCR sequences across all samples. In PML biopsies, TCR diversity was negatively associated with a transcriptional signature of T cell mediated immune activation (p=4e-4) associated with PML outcome. Additionally, in lesions of the proliferative molecular subtype, TCR diversity was decreased in regressive versus progressive/persistent PMLs (p=0.045). Within each patient, TCRs were more likely to be shared between biopsies sampled at the same timepoint than biopsies sampled at the same anatomic location at different times. Clonally expanded TCRs, within a biopsied lesion, were more likely to be expanded at future time points than non-expanded clones. The majority of TCR sequences were found in a single sample, with only 3396 (8.6%) found in more than one sample and 1057 (2.7%) found in two or more patients (ie, public); however, when compared with a public database of CDR3 sequences, 4543 (11.6%) of TCRs were identified as public. TCRs with known antigen specificities were enriched among public TCRs (p<0.001). Conclusions Decreased TCR diversity may reflect nascent immune responses that contribute to PML elimination. Further studies are needed to explore the potential for immunoprevention of PMLs.
Collapse
Affiliation(s)
- Asaf Maoz
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,Boston Medical Center, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Carter Merenstein
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Yusuke Koga
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Austin Potter
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Adam C Gower
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Gang Liu
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Sherry Zhang
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Hanqiao Liu
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Avrum Spira
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,The Lung Cancer Initiative at Johnson and Johnson, Cambridge, Massachusetts, USA
| | - Mary E Reid
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Joshua D Campbell
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Sarah A Mazzilli
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Marc E Lenburg
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Jennifer Beane
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
37
|
Kadara H, Tran LM, Liu B, Vachani A, Li S, Sinjab A, Zhou XJ, Dubinett SM, Krysan K. Early Diagnosis and Screening for Lung Cancer. Cold Spring Harb Perspect Med 2021; 11:a037994. [PMID: 34001525 PMCID: PMC8415293 DOI: 10.1101/cshperspect.a037994] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cancer interception refers to actively blocking the cancer development process by preventing progression of premalignancy to invasive disease. The rate-limiting steps for effective lung cancer interception are the incomplete understanding of the earliest molecular events associated with lung carcinogenesis, the lack of preclinical models of pulmonary premalignancy, and the challenge of developing highly sensitive and specific methods for early detection. Recent advances in cancer interception are facilitated by developments in next-generation sequencing, computational methodologies, as well as the renewed emphasis in precision medicine and immuno-oncology. This review summarizes the current state of knowledge in the areas of molecular abnormalities in lung cancer continuum, preclinical human models of lung cancer pathogenesis, and the advances in early lung cancer diagnostics.
Collapse
Affiliation(s)
- Humam Kadara
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Linh M Tran
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Bin Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Anil Vachani
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, Pennsylvania 19104, USA
| | - Shuo Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xianghong J Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Steven M Dubinett
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, California 90024, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA
| | - Kostyantyn Krysan
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA
| |
Collapse
|
38
|
Kerr AR, Lodi G. Management of Oral Potentially Malignant Disorders. Oral Dis 2021; 27:2008-2025. [PMID: 34324758 DOI: 10.1111/odi.13980] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022]
Abstract
Patients with oral potentially malignant disorders (OPMDs), including oral leukoplakia and erythroplakia, proliferative verrucous leukoplakia, oral submucous fibrosis, and oral lichen planus/lichenoid lesions can be challenging to manage. A small proportion will undergo cancer development and determining a patient's cancer risk is key to making management decisions. Yet, our understanding of the natural history of OPMDs has not been fully elucidated, and a precision approach based on the integration of numerous predictive markers has not been validated by prospective studies. Evidence-based health promotion by clinicians and healthcare systems is not embraced universally. Medical and surgical interventions evaluated by rigorous research measuring important endpoints, such as cancer development, mortality, or survival are difficult and expensive to run. Most of these studies employ non-ideal surrogate endpoints and have deep methodologic flaws. Diagnostic criteria for enrolling research subjects are not uniform, and patients with the highest risk for cancer development comprise small proportions of those enrolled. Few studies explore quality of life and patient preferences. It is time to rethink how we approach the management of these patients, across each OPMD, and considering the healthcare infrastructure and cost effectiveness. Global networks with well-characterized patient populations with OPMDs and well-designed interventional trials using validated outcome measures are needed.
Collapse
Affiliation(s)
- A Ross Kerr
- Department of Oral & Maxillofacial Pathology, Radiology & Medicine.,New York University College of Dentistry, New York, NY, USA
| | - Giovanni Lodi
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milano, Italia
| |
Collapse
|
39
|
Liquid Biopsy Analysis in Clinical Practice: Focus on Lung Cancer. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2030021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. Despite the emergence of highly effective targeted therapies, up to 30% of advanced stage non-small cell lung cancer (NSCLC) patients do not undergo tissue molecular testing because of scarce tissue availability. Liquid biopsy, on the other hand, offers these patients a valuable opportunity to receive the best treatment options in a timely manner. Indeed, besides being much faster and less invasive than conventional tissue-based analysis, it can also yield specific information about the genetic make-up and evolution of patients’ tumors. However, several issues, including lack of standardized protocols for sample collection, processing, and interpretation, still need to be addressed before liquid biopsy can be fully incorporated into routine oncology practice. Here, we reviewed the most important challenges hindering the implementation of liquid biopsy in oncology practice, as well as the great advantages of this approach for the treatment of NSCLC patients.
Collapse
|
40
|
Jordan F, Huber S, Sommer S, Schenkirsch G, Frühwald MC, Trepel M, Claus R, Kuhlen M. A Retrospective 5-Year Single Center Study Highlighting the Risk of Cancer Predisposition in Adolescents and Young Adults. Cancers (Basel) 2021; 13:3033. [PMID: 34204522 PMCID: PMC8234548 DOI: 10.3390/cancers13123033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 11/23/2022] Open
Abstract
The knowledge of inherited cancer susceptibility opens a new field of cancer medicine. We conducted a retrospective single-center cohort study. Data of AYA cancer patients registered between January 2014 and December 2018 were analyzed. The median age at cancer diagnosis of 704 patients (343 males, 361 females) was 32 years (range, 15-39 years), median follow-up was 181 days (range, 1-1975 days). Solid tumors were diagnosed in 575 (81.7%) patients, hematologic malignancies in 129 (18.3%) patients. Multiple primary cancers were reported in 36 (5.1%) patients. Malignancies that may be indicators of inherited cancer susceptibility were diagnosed in 2.6% of patients with cancers of the endocrine system, in 73% of cancers of the gastrointestinal system, in 88% of tumors of the central nervous system, in 92% of cancers of the urinary tract, and in 59% of head and neck tumors. In addition, all patients with breast cancer, sarcoma, and peripheral nerve sheath tumor were in need of genetic counselling. In sum, at least 181 of 704 (25.7%) AYA cancer patients presented with malignancies suspicious of harboring pathogenic germline variants. Evaluation of AYA cancer patients for hereditary cancer predisposition needs to be integrated into daily practice.
Collapse
Affiliation(s)
- Frank Jordan
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany; (F.J.); (S.S.); (M.T.); (R.C.)
| | - Simon Huber
- Paediatrics and Adolescent Medicine, University Medical Center Augsburg, 86156 Augsburg, Germany; (S.H.); (M.C.F.)
| | - Sebastian Sommer
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany; (F.J.); (S.S.); (M.T.); (R.C.)
| | - Gerhard Schenkirsch
- Comprehensive Cancer Center Augsburg, University Medical Center Augsburg, 86156 Augsburg, Germany;
| | - Michael C. Frühwald
- Paediatrics and Adolescent Medicine, University Medical Center Augsburg, 86156 Augsburg, Germany; (S.H.); (M.C.F.)
| | - Martin Trepel
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany; (F.J.); (S.S.); (M.T.); (R.C.)
| | - Rainer Claus
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany; (F.J.); (S.S.); (M.T.); (R.C.)
| | - Michaela Kuhlen
- Paediatrics and Adolescent Medicine, University Medical Center Augsburg, 86156 Augsburg, Germany; (S.H.); (M.C.F.)
| |
Collapse
|
41
|
William WN, Zhao X, Bianchi JJ, Lin HY, Cheng P, Lee JJ, Carter H, Alexandrov LB, Abraham JP, Spetzler DB, Dubinett SM, Cleveland DW, Cavenee W, Davoli T, Lippman SM. Immune evasion in HPV - head and neck precancer-cancer transition is driven by an aneuploid switch involving chromosome 9p loss. Proc Natl Acad Sci U S A 2021; 118:e2022655118. [PMID: 33952700 PMCID: PMC8126856 DOI: 10.1073/pnas.2022655118] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An aneuploid-immune paradox encompasses somatic copy-number alterations (SCNAs), unleashing a cytotoxic response in experimental precancer systems, while conversely being associated with immune suppression and cytotoxic-cell depletion in human tumors, especially head and neck cancer (HNSC). We present evidence from patient samples and cell lines that alterations in chromosome dosage contribute to an immune hot-to-cold switch during human papillomavirus-negative (HPV-) head and neck tumorigenesis. Overall SCNA (aneuploidy) level was associated with increased CD3+ and CD8+ T cell microenvironments in precancer (mostly CD3+, linked to trisomy and aneuploidy), but with T cell-deficient tumors. Early lesions with 9p21.3 loss were associated with depletion of cytotoxic T cell infiltration in TP53 mutant tumors; and with aneuploidy were associated with increased NK-cell infiltration. The strongest driver of cytotoxic T cell and Immune Score depletion in oral cancer was 9p-arm level loss, promoting profound decreases of pivotal IFN-γ-related chemokines (e.g., CXCL9) and pathway genes. Chromosome 9p21.3 deletion contributed mainly to cell-intrinsic senescence suppression, but deletion of the entire arm was necessary to diminish levels of cytokine, JAK-STAT, and Hallmark NF-κB pathways. Finally, 9p arm-level loss and JAK2-PD-L1 codeletion (at 9p24) were predictive markers of poor survival in recurrent HPV- HNSC after anti-PD-1 therapy; likely amplified by independent aneuploidy-induced immune-cold microenvironments observed here. We hypothesize that 9p21.3 arm-loss expansion and epistatic interactions allow oral precancer cells to acquire properties to overcome a proimmunogenic aneuploid checkpoint, transform and invade. These findings enable distinct HNSC interception and precision-therapeutic approaches, concepts that may apply to other CN-driven neoplastic, immune or aneuploid diseases, and immunotherapies.
Collapse
Affiliation(s)
- William N William
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- Hospital BP, a Beneficência Portuguesa de São Paulo, 01323-001 São Paulo, Brazil
| | - Xin Zhao
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Langone Health, New York, NY 10016
| | - Joy J Bianchi
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Langone Health, New York, NY 10016
| | - Heather Y Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Pan Cheng
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Langone Health, New York, NY 10016
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Hannah Carter
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla, CA 92037
| | - Ludmil B Alexandrov
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92037
| | - Jim P Abraham
- Research and Development, Caris Life Sciences, Irving, TX 75039
| | | | - Steven M Dubinett
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024
| | - Don W Cleveland
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92037
| | - Webster Cavenee
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037;
- Department of Medicine, University of California San Diego, La Jolla, CA 92037
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92037
| | - Teresa Davoli
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Langone Health, New York, NY 10016;
| | - Scott M Lippman
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
42
|
Liang S, Mohanty V, Dou J, Miao Q, Huang Y, Müftüoğlu M, Ding L, Peng W, Chen K. Single-cell manifold-preserving feature selection for detecting rare cell populations. NATURE COMPUTATIONAL SCIENCE 2021; 1:374-384. [PMID: 36969355 PMCID: PMC10035340 DOI: 10.1038/s43588-021-00070-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/19/2021] [Indexed: 01/04/2023]
Abstract
A key challenge in studying organisms and diseases is to detect rare molecular programs and rare cell populations (RCPs) that drive development, differentiation, and transformation. Molecular features such as genes and proteins defining RCPs are often unknown and difficult to detect from unenriched single-cell data, using conventional dimensionality reduction and clustering-based approaches. Here, we propose an unsupervised approach, SCMER (Single-Cell Manifold presERving feature selection), which selects a compact set of molecular features with definitive meanings that preserve the manifold of the data. We applied SCMER in the context of hematopoiesis, lymphogenesis, tumorigenesis, and drug resistance and response. We found that SCMER can identify non-redundant features that sensitively delineate both common cell lineages and rare cellular states. SCMER can be used for discovering molecular features in a high dimensional dataset, designing targeted, cost-effective assays for clinical applications, and facilitating multi-modality integration.
Collapse
Affiliation(s)
- Shaoheng Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Computer Science, Rice University, Houston, Texas, 77005, USA
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Jinzhuang Dou
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Biostatistics & Data Science, School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, 77030, USA
| | - Yuefan Huang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Biostatistics & Data Science, School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, 77030, USA
| | - Muharrem Müftüoğlu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63108
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, 77024
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| |
Collapse
|
43
|
Kavran AJ, Clauset A. Denoising large-scale biological data using network filters. BMC Bioinformatics 2021; 22:157. [PMID: 33765911 PMCID: PMC7992843 DOI: 10.1186/s12859-021-04075-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/15/2021] [Indexed: 11/29/2022] Open
Abstract
Background Large-scale biological data sets are often contaminated by noise, which can impede accurate inferences about underlying processes. Such measurement noise can arise from endogenous biological factors like cell cycle and life history variation, and from exogenous technical factors like sample preparation and instrument variation. Results We describe a general method for automatically reducing noise in large-scale biological data sets. This method uses an interaction network to identify groups of correlated or anti-correlated measurements that can be combined or “filtered” to better recover an underlying biological signal. Similar to the process of denoising an image, a single network filter may be applied to an entire system, or the system may be first decomposed into distinct modules and a different filter applied to each. Applied to synthetic data with known network structure and signal, network filters accurately reduce noise across a wide range of noise levels and structures. Applied to a machine learning task of predicting changes in human protein expression in healthy and cancerous tissues, network filtering prior to training increases accuracy up to 43% compared to using unfiltered data. Conclusions Network filters are a general way to denoise biological data and can account for both correlation and anti-correlation between different measurements. Furthermore, we find that partitioning a network prior to filtering can significantly reduce errors in networks with heterogenous data and correlation patterns, and this approach outperforms existing diffusion based methods. Our results on proteomics data indicate the broad potential utility of network filters to applications in systems biology. Supplementary Information The online version supplementary material available at 10.1186/s12859-021-04075-x.
Collapse
Affiliation(s)
- Andrew J Kavran
- Department of Biochemistry, University of Colorado, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Aaron Clauset
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA. .,Department of Computer Science, University of Colorado, Boulder, CO, USA. .,Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
44
|
Bommi PV, Bowen CM, Reyes-Uribe L, Wu W, Katayama H, Rocha P, Parra ER, Francisco-Cruz A, Ozcan Z, Tosti E, Willis JA, Wu H, Taggart MW, Burks JK, Lynch PM, Edelmann W, Scheet PA, Wistuba II, Sinha KM, Hanash SM, Vilar E. The Transcriptomic Landscape of Mismatch Repair-Deficient Intestinal Stem Cells. Cancer Res 2021; 81:2760-2773. [PMID: 34003775 DOI: 10.1158/0008-5472.can-20-2896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 02/16/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
Lynch syndrome is the most common cause of hereditary colorectal cancer and is secondary to germline alterations in one of four DNA mismatch repair (MMR) genes. Here we aimed to provide novel insights into the initiation of MMR-deficient (MMRd) colorectal carcinogenesis by characterizing the expression profile of MMRd intestinal stem cells (ISC). A tissue-specific MMRd mouse model (Villin-Cre;Msh2 LoxP/LoxP ) was crossed with a reporter mouse (Lgr5-EGFP-IRES-creERT2) to trace and isolate ISCs (Lgr5+) using flow cytometry. Three different ISC genotypes (Msh2-KO, Msh2-HET, and Msh2-WT) were isolated and processed for mRNA-seq and mass spectrometry, followed by bioinformatic analyses to identify expression signatures of complete MMRd and haplo-insufficiency. These findings were validated using qRT-PCR, IHC, and whole transcriptomic sequencing in mouse tissues, organoids, and a cohort of human samples, including normal colorectal mucosa, premalignant lesions, and early-stage colorectal cancers from patients with Lynch syndrome and patients with familial adenomatous polyposis (FAP) as controls. Msh2-KO ISCs clustered together with differentiated intestinal epithelial cells from all genotypes. Gene-set enrichment analysis indicated inhibition of replication, cell-cycle progression, and the Wnt pathway and activation of epithelial signaling and immune reaction. An expression signature derived from MMRd ISCs successfully distinguished MMRd neoplastic lesions of patients with Lynch syndrome from FAP controls. SPP1 was specifically upregulated in MMRd ISCs and colocalized with LGR5 in Lynch syndrome colorectal premalignant lesions and tumors. These results show that expression signatures of MMRd ISC recapitulate the initial steps of Lynch syndrome carcinogenesis and have the potential to unveil novel biomarkers of early cancer initiation. SIGNIFICANCE: The transcriptomic and proteomic profile of MMR-deficient intestinal stem cells displays a unique set of genes with potential roles as biomarkers of cancer initiation and early progression.
Collapse
Affiliation(s)
- Prashant V Bommi
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Charles M Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenhui Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pedro Rocha
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Francisco-Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zuhal Ozcan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jason A Willis
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa W Taggart
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick M Lynch
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Paul A Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krishna M Sinha
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
45
|
Reyes-Uribe L, Wu W, Gelincik O, Bommi PV, Francisco-Cruz A, Solis LM, Lynch PM, Lim R, Stoffel EM, Kanth P, Samadder NJ, Mork ME, Taggart MW, Milne GL, Marnett LJ, Vornik L, Liu DD, Revuelta M, Chang K, You YN, Kopelovich L, Wistuba II, Lee JJ, Sei S, Shoemaker RH, Szabo E, Richmond E, Umar A, Perloff M, Brown PH, Lipkin SM, Vilar E. Naproxen chemoprevention promotes immune activation in Lynch syndrome colorectal mucosa. Gut 2021; 70:555-566. [PMID: 32641470 PMCID: PMC7790993 DOI: 10.1136/gutjnl-2020-320946] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Patients with Lynch syndrome (LS) are at markedly increased risk for colorectal cancer. It is being increasingly recognised that the immune system plays an essential role in LS tumour development, thus making an ideal target for cancer prevention. Our objective was to evaluate the safety, assess the activity and discover novel molecular pathways involved in the activity of naproxen as primary and secondary chemoprevention in patients with LS. DESIGN We conducted a Phase Ib, placebo-controlled, randomised clinical trial of two dose levels of naproxen sodium (440 and 220 mg) administered daily for 6 months to 80 participants with LS, and a co-clinical trial using a genetically engineered mouse model of LS and patient-derived organoids (PDOs). RESULTS Overall, the total number of adverse events was not different across treatment arms with excellent tolerance of the intervention. The level of prostaglandin E2 in the colorectal mucosa was significantly decreased after treatment with naproxen when compared with placebo. Naproxen activated different resident immune cell types without any increase in lymphoid cellularity, and changed the expression patterns of the intestinal crypt towards epithelial differentiation and stem cell regulation. Naproxen demonstrated robust chemopreventive activity in a mouse co-clinical trial and gene expression profiles induced by naproxen in humans showed perfect discrimination of mice specimens with LS and PDOs treated with naproxen and control. CONCLUSIONS Naproxen is a promising strategy for immune interception in LS. We have discovered naproxen-induced gene expression profiles for their potential use as predictive biomarkers of drug activity. TRIAL REGISTRATION NUMBER gov Identifier: NCT02052908.
Collapse
Affiliation(s)
- Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenhui Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Prashant V Bommi
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alejandro Francisco-Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Luisa M Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Patrick M Lynch
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ramona Lim
- Department of Gastroenterology, Dana Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Kanth
- Division of Gastroenterology, Department of Medicine, University of Utah/Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - N Jewel Samadder
- Department of Gastroenterology and Hepatology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Maureen E Mork
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Melissa W Taggart
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ginger L Milne
- Departments of Biochemistry, Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Lawrence J Marnett
- Departments of Biochemistry, Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Lana Vornik
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Diane D Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Kyle Chang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Y Nancy You
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shizuko Sei
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert H Shoemaker
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Ellen Richmond
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Marjorie Perloff
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
46
|
Heudel P, Chabaud S, Perol D, Flechon A, Fayette J, Combemale P, Tredan O, Desseigne F, de la Fouchardiere C, Boyle H, Perol M, Bachelot T, Cassier P, Avrillon V, Terret C, Michallet AS, Neidhardt-Berard EM, Nicolas-Virelizier E, Dufresne A, Belhabri A, Brahmi M, Lebras L, Nicolini F, Sarabi M, Rey P, Bonneville-Levard A, Rochefort P, Provensal AM, Eberst L, Assaad S, Swalduz A, Saintigny P, Toussaint P, Guillermin Y, Castets M, Coutzac C, Meeus P, Dupré A, Durand T, Crochet H, Fervers B, Gomez F, Rivoire M, Gregoire V, Claude L, Chassagne-Clement C, Pilleul F, Mognetti T, Russias B, Soubirou JL, Lasset C, Chvetzoff G, Mehlen P, Beaupère S, Zrounba P, Ray-Coquard I, Blay JY. Immune checkpoint inhibitor treatment of a first cancer is associated with a decreased incidence of second primary cancer. ESMO Open 2021; 6:100044. [PMID: 33516148 PMCID: PMC7844579 DOI: 10.1016/j.esmoop.2020.100044] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Second primary cancers (SPCs) are diagnosed in over 5% of patients after a first primary cancer (FPC). We explore here the impact of immune checkpoint inhibitors (ICIs) given for an FPC on the risk of SPC in different age groups, cancer types and treatments. Patients and methods The files of the 46 829 patients diagnosed with an FPC in the Centre Léon Bérard from 2013 to 2018 were analyzed. Structured data were extracted and electronic patient records were screened using a natural language processing tool, with validation using manual screening of 2818 files of patients. Univariate and multivariate analyses of the incidence of SPC according to patient characteristics and treatment were conducted. Results Among the 46 829 patients, 1830 (3.9%) had a diagnosis of SPC with a median interval of 11.1 months (range 0-78 months); 18 128 (38.7%) received cytotoxic chemotherapy (CC) and 1163 (2.5%) received ICIs for the treatment of the FPC in this period. SPCs were observed in 7/1163 (0.6%) patients who had received ICIs for their FPC versus 437/16 997 (2.6%) patients receiving CC and no ICIs for the FPC versus 1386/28 669 (4.8%) for patients receiving neither CC nor ICIs for the FPC. This reduction was observed at all ages and for all histotypes analyzed. Treatment with ICIs and/or CC for the FPC are associated with a reduced risk of SPC in multivariate analysis. Conclusion Immunotherapy with ICIs alone and in combination with CC was found to be associated with a reduced incidence of SPC for all ages and cancer types. From 2013 to 2018, 3.9% of the 46 829 patients diagnosed with a first cancer presented with an SPC. Treatment of the first cancer with ICIs was associated with a major reduction of SPC. CC given for an FPC was also associated with a lower magnitude of reduction of SPC. There were no SPC in cancer patients treated with ICIs in the localized phase of their first cancer.
Collapse
Affiliation(s)
- P Heudel
- Centre Léon Bérard, Lyon, France
| | | | - D Perol
- Centre Léon Bérard, Lyon, France
| | | | | | | | - O Tredan
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | | | | | - H Boyle
- Centre Léon Bérard, Lyon, France
| | - M Perol
- Centre Léon Bérard, Lyon, France
| | - T Bachelot
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | | | | | - C Terret
- Centre Léon Bérard, Lyon, France
| | | | | | | | - A Dufresne
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | | | - M Brahmi
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | - L Lebras
- Centre Léon Bérard, Lyon, France
| | - F Nicolini
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | - M Sarabi
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | - P Rey
- Centre Léon Bérard, Lyon, France
| | | | | | | | - L Eberst
- Centre Léon Bérard, Lyon, France
| | - S Assaad
- Centre Léon Bérard, Lyon, France
| | | | - P Saintigny
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | | | | | - M Castets
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | - C Coutzac
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France
| | - P Meeus
- Centre Léon Bérard, Lyon, France
| | - A Dupré
- Centre Léon Bérard, Lyon, France
| | - T Durand
- Centre Léon Bérard, Lyon, France
| | | | | | - F Gomez
- Centre Léon Bérard, Lyon, France
| | - M Rivoire
- Centre Léon Bérard, Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France
| | | | - L Claude
- Centre Léon Bérard, Lyon, France
| | | | - F Pilleul
- Centre Léon Bérard, Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France
| | | | | | | | - C Lasset
- Centre Léon Bérard, Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France
| | | | - P Mehlen
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France
| | - S Beaupère
- Centre Léon Bérard, Lyon, France; Unicancer, Paris, France
| | | | - I Ray-Coquard
- Centre Léon Bérard, Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France
| | - J-Y Blay
- Centre Léon Bérard, Lyon, France; Cancer Research Center of Lyon (CRCL), Lyon, France; Centre Léon Bérard & Université Claude Bernard, Lyon, France; Unicancer, Paris, France.
| |
Collapse
|
47
|
Elucidation of the Genomic-Epigenomic Interaction Landscape of Aggressive Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6641429. [PMID: 33511206 PMCID: PMC7825361 DOI: 10.1155/2021/6641429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022]
Abstract
Background Majority of prostate cancer (PCa) deaths are attributed to localized high-grade aggressive tumours which progress rapidly to metastatic disease. A critical unmet need in clinical management of PCa is discovery and characterization of the molecular drivers of aggressive tumours. The development and progression of aggressive PCa involve genetic and epigenetic alterations occurring in the germline, somatic (tumour), and epigenomes. To date, interactions between genes containing germline, somatic, and epigenetic mutations in aggressive PCa have not been characterized. The objective of this investigation was to elucidate the genomic-epigenomic interaction landscape in aggressive PCa to identify potential drivers aggressive PCa and the pathways they control. We hypothesized that aggressive PCa originates from a complex interplay between genomic (both germline and somatic mutations) and epigenomic alterations. We further hypothesized that these complex arrays of interacting genomic and epigenomic factors affect gene expression, molecular networks, and signaling pathways which in turn drive aggressive PCa. Methods We addressed these hypotheses by performing integrative data analysis combining information on germline mutations from genome-wide association studies with somatic and epigenetic mutations from The Cancer Genome Atlas using gene expression as the intermediate phenotype. Results The investigation revealed signatures of genes containing germline, somatic, and epigenetic mutations associated with aggressive PCa. Aberrant DNA methylation had effect on gene expression. In addition, the investigation revealed molecular networks and signalling pathways enriched for germline, somatic, and epigenetic mutations including the STAT3, PTEN, PCa, ATM, AR, and P53 signalling pathways implicated in aggressive PCa. Conclusions The study demonstrated that integrative analysis combining diverse omics data is a powerful approach for the discovery of potential clinically actionable biomarkers, therapeutic targets, and elucidation of oncogenic interactions between genomic and epigenomic alterations in aggressive PCa.
Collapse
|
48
|
Byng D, Retèl VP, Schaapveld M, Wesseling J, van Harten WH. Treating (low-risk) DCIS patients: What can we learn from real-world cancer registry evidence? Breast Cancer Res Treat 2021; 187:187-196. [PMID: 33389397 PMCID: PMC8062323 DOI: 10.1007/s10549-020-06042-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/28/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE Results from active surveillance trials for ductal carcinoma in situ (DCIS) will not be available for > 10 years. A model based on real-world data (RWD) can demonstrate the comparative impact of non-intervention for women with low-risk features. METHODS Multi-state models were developed using Surveillance, Epidemiology, and End Results Program (SEER) data for three treatment strategies (no local treatment, breast conserving surgery [BCS], BCS + radiotherapy [RT]), and for women with DCIS low-risk features. Eligible cases included women aged ≥ 40 years, diagnosed with primary DCIS between 1992 and 2016. Five mutually exclusive health states were modelled: DCIS, ipsilateral invasive breast cancer (iIBC) ≤ 5 years and > 5 years post-DCIS diagnosis, contralateral IBC, death preceded by and death not preceded by IBC. Propensity score-weighted Cox models assessed effects of treatment, age, diagnosis year, grade, ER status, and race. RESULTS Data on n = 85,982 women were used. Increased risk of iIBC ≤ 5 years post-DCIS was demonstrated for ages 40-49 (Hazard ratio (HR) 1.86, 95% Confidence Interval (CI) 1.34-2.57 compared to age 50-69), grade 3 lesions (HR 1.42, 95%CI 1.05-1.91) compared to grade 2, lesion size ≥ 2 cm (HR 1.66, 95%CI 1.23-2.25), and Black race (HR 2.52, 95%CI 1.83-3.48 compared to White). According to the multi-state model, propensity score-matched women with low-risk features who had not died or experienced any subsequent breast event by 10 years, had a predicted probability of iIBC as first event of 3.02% for no local treatment, 1.66% for BCS, and 0.42% for BCS+RT. CONCLUSION RWD from the SEER registry showed that women with primary DCIS and low-risk features demonstrate minimal differences by treatment strategy in experiencing subsequent breast events. There may be opportunity to de-escalate treatment for certain women with low-risk features: Hispanic and non-Hispanic white women aged 50-69 at diagnosis, with ER+, grade 1 + 2, < 2 cm DCIS lesions.
Collapse
Affiliation(s)
- Danalyn Byng
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Health Technology and Services Research Department, Technical Medical Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands.
| | - Valesca P Retèl
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Health Technology and Services Research Department, Technical Medical Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Michael Schaapveld
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Wim H van Harten
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Health Technology and Services Research Department, Technical Medical Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| |
Collapse
|
49
|
Baliu-Piqué M, Pandiella A, Ocana A. Breast Cancer Heterogeneity and Response to Novel Therapeutics. Cancers (Basel) 2020; 12:E3271. [PMID: 33167363 PMCID: PMC7694303 DOI: 10.3390/cancers12113271] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Targeted cancer therapies against oncogenic drivers are actively being developed and tested in clinical trials. Targeting an oncogenic driver may only prove effective if the mutation is present in most tumoral cells. Therefore, highly heterogeneous tumors may be refractory to these therapies. This makes tumor heterogeneity a major challenge in cancer therapy. Although heterogeneity has traditionally been attributed to genetic diversity within cancer cell populations, it is now widely recognized that human cancers are heterogeneous in almost all distinguishable phenotypic characteristics. Understanding the genetic variability and also the non-genetic influences of tumor heterogeneity will provide novel insights into how to reverse therapeutic resistance and improve cancer therapy.
Collapse
Affiliation(s)
- Mariona Baliu-Piqué
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos and CIBERONC, 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer and CIBERONC, CSIC-IBSAL, 37007 Salamanca, Spain;
| | - Alberto Ocana
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos and CIBERONC, 28040 Madrid, Spain;
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomedicas, Castilla-La Mancha University (CRIB-UCLM), 02008 Albacete, Spain
| |
Collapse
|
50
|
Improved detection of tumor suppressor events in single-cell RNA-Seq data. NPJ Genom Med 2020; 5:43. [PMID: 33083012 PMCID: PMC7541488 DOI: 10.1038/s41525-020-00151-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Tissue-specific transcription factors are frequently inactivated in cancer. To fully dissect the heterogeneity of such tumor suppressor events requires single-cell resolution, yet this is challenging because of the high dropout rate. Here we propose a simple yet effective computational strategy called SCIRA to infer regulatory activity of tissue-specific transcription factors at single-cell resolution and use this tool to identify tumor suppressor events in single-cell RNA-Seq cancer studies. We demonstrate that tissue-specific transcription factors are preferentially inactivated in the corresponding cancer cells, suggesting that these are driver events. For many known or suspected tumor suppressors, SCIRA predicts inactivation in single cancer cells where differential expression does not, indicating that SCIRA improves the sensitivity to detect changes in regulatory activity. We identify NKX2-1 and TBX4 inactivation as early tumor suppressor events in normal non-ciliated lung epithelial cells from smokers. In summary, SCIRA can help chart the heterogeneity of tumor suppressor events at single-cell resolution.
Collapse
|