1
|
Lu Z, Mo S, Xie D, Zhai X, Deng S, Zhou K, Wang K, Kang X, Zhang H, Tong J, Hou L, Hu H, Li X, Zhou D, Lee LTO, Liu L, Zhu Y, Yu J, Lan P, Wang J, He Z, He X, Hu Z. Polyclonal-to-monoclonal transition in colorectal precancerous evolution. Nature 2024; 636:233-240. [PMID: 39478225 DOI: 10.1038/s41586-024-08133-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/27/2024] [Indexed: 12/06/2024]
Abstract
Unravelling the origin and evolution of precancerous lesions is crucial for effectively preventing malignant transformation, yet our current knowledge remains limited1-3. Here we used a base editor-enabled DNA barcoding system4 to comprehensively map single-cell phylogenies in mouse models of intestinal tumorigenesis induced by inflammation or loss of the Apc gene. Through quantitative analysis of high-resolution phylogenies including 260,922 single cells from normal, inflamed and neoplastic intestinal tissues, we identified tens of independent cell lineages undergoing parallel clonal expansions within each lesion. We also found polyclonal origins of human sporadic colorectal polyps through bulk whole-exome sequencing and single-gland whole-genome sequencing. Genomic and clinical data support a model of polyclonal-to-monoclonal transition, with monoclonal lesions representing a more advanced stage. Single-cell RNA sequencing revealed extensive intercellular interactions in early polyclonal lesions, but there was significant loss of interactions during monoclonal transition. Therefore, our data suggest that colorectal precancer is often founded by many different lineages and highlight their cooperative interactions in the earliest stages of cancer formation. These findings provide insights into opportunities for earlier intervention in colorectal cancer.
Collapse
Affiliation(s)
- Zhaolian Lu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shanlan Mo
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Duo Xie
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xiangwei Zhai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Shanjun Deng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Kantian Zhou
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kun Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Mathematical Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Xueling Kang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hao Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Juanzhen Tong
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liangzhen Hou
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Huijuan Hu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xuefei Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Leo Tsz On Lee
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Li Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiguang Wang
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Division of Life Science, Department of Chemical and Biological Engineering, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Zhen He
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Xionglei He
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| | - Zheng Hu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
2
|
Carrothers E, Appleby M, Lai V, Kozbenko T, Alomar D, Smith BJ, Hamada N, Hinton P, Ainsbury EA, Hocking R, Yauk C, Wilkins RC, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to cataracts. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:31-56. [PMID: 38644659 DOI: 10.1002/em.22594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Abstract
Cataracts are one of the leading causes of blindness, with an estimated 95 million people affected worldwide. A hallmark of cataract development is lens opacification, typically associated not only with aging but also radiation exposure as encountered by interventional radiologists and astronauts during the long-term space mission. To better understand radiation-induced cataracts, the adverse outcome pathway (AOP) framework was used to structure and evaluate knowledge across biological levels of organization (e.g., macromolecular, cell, tissue, organ, organism and population). AOPs identify a sequence of key events (KEs) causally connected by key event relationships (KERs) beginning with a molecular initiating event to an adverse outcome (AO) of relevance to regulatory decision-making. To construct the cataract AO and retrieve evidence to support it, a scoping review methodology was used to filter, screen, and review studies based on the modified Bradford Hill criteria. Eight KEs were identified that were moderately supported by empirical evidence (e.g., dose-, time-, incidence-concordance) across the adjacent (directly linked) relationships using well-established endpoints. Over half of the evidence to justify the KER linkages was derived from the evidence stream of biological plausibility. Early KEs of oxidative stress and protein modifications had strong linkages to downstream KEs and could be the focus of countermeasure development. Several identified knowledge gaps and inconsistencies related to the quantitative understanding of KERs which could be the basis of future research, most notably directed to experiments in the range of low or moderate doses and dose-rates, relevant to radiation workers and other occupational exposures.
Collapse
Affiliation(s)
- Emma Carrothers
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Meghan Appleby
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vita Lai
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Dalya Alomar
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Patricia Hinton
- Defense Research & Development Canada, Canadian Forces Environmental Medicine Establishment, Toronto, Ontario, Canada
| | - Elizabeth A Ainsbury
- Radiation, Chemical and Environmental Hazards Division, UK Health Security Agency, Birmingham, UK
- Environmental Research Group within the School of Public Health, Faculty of Medicine at Imperial College of Science, Technology and Medicine, London, UK
| | - Robyn Hocking
- Learning and Knowledge and Library Services, Health Canada, Ottawa, Ontario, Canada
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth C Wilkins
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Parsons BL. Clonal expansion of cancer driver gene mutants investigated using advanced sequencing technologies. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108514. [PMID: 39369952 DOI: 10.1016/j.mrrev.2024.108514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Advanced sequencing technologies (ASTs) have revolutionized the quantitation of cancer driver mutations (CDMs) as rare events, which has utility in clinical oncology, cancer research, and cancer risk assessment. This review focuses on studies that have used ASTs to characterize clonal expansion (CE) of cells carrying CDMs and to explicate the selective pressures that shape CE. Importantly, high-sensitivity ASTs have made possible the characterization of mutant clones and CE in histologically normal tissue samples, providing the means to investigate nascent tumor development. Some ASTs can identify mutant clones in a spatially defined context; others enable integration of mutant data with analyses of gene expression, thereby elaborating immune, inflammatory, metabolic, and/or stromal microenvironmental impacts on CE. As a whole, these studies make it clear that a startlingly large fraction of cells in histologically normal tissues carry CDMs, CDMs may confer a context-specific selective advantage leading to CE, and only a small fraction of cells carrying CDMs eventually result in neoplasia. These observations were integrated with available literature regarding the mechanisms underlying clonal selection to interpret how measurements of CDMs and CE can be interpreted as biomarkers of cancer risk. Given the stochastic nature of carcinogenesis, the potential functional latency of driver mutations, the complexity of potential mutational and microenvironmental interactions, and involvement of other types of genetic and epigenetic changes, it is concluded that CDM-based measurements should be viewed as probabilistic rather than deterministic biomarkers. Increasing inter-sample variability in CDM levels (as a consequence of CE) may be interpretable as a shift away from normal tissue homeostasis and an indication of increased future cancer risk, a process that may reflect normal aging or carcinogen exposure. Consequently, analyses of variability in levels of CDMs have the potential to bolster existing approaches for carcinogenicity testing.
Collapse
Affiliation(s)
- Barbara L Parsons
- US Food and Drug Administration, National Center for Toxicological Research, Division of Genetic and Molecular Toxicology, 3900 NCTR Rd., Jefferson AR 72079, USA.
| |
Collapse
|
4
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Parsons BL, Beal MA, Dearfield KL, Douglas GR, Gi M, Gollapudi BB, Heflich RH, Horibata K, Kenyon M, Long AS, Lovell DP, Lynch AM, Myers MB, Pfuhler S, Vespa A, Zeller A, Johnson GE, White PA. Severity of effect considerations regarding the use of mutation as a toxicological endpoint for risk assessment: A report from the 8th International Workshop on Genotoxicity Testing (IWGT). ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024. [PMID: 38828778 DOI: 10.1002/em.22599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/13/2024] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
Exposure levels without appreciable human health risk may be determined by dividing a point of departure on a dose-response curve (e.g., benchmark dose) by a composite adjustment factor (AF). An "effect severity" AF (ESAF) is employed in some regulatory contexts. An ESAF of 10 may be incorporated in the derivation of a health-based guidance value (HBGV) when a "severe" toxicological endpoint, such as teratogenicity, irreversible reproductive effects, neurotoxicity, or cancer was observed in the reference study. Although mutation data have been used historically for hazard identification, this endpoint is suitable for quantitative dose-response modeling and risk assessment. As part of the 8th International Workshops on Genotoxicity Testing, a sub-group of the Quantitative Analysis Work Group (WG) explored how the concept of effect severity could be applied to mutation. To approach this question, the WG reviewed the prevailing regulatory guidance on how an ESAF is incorporated into risk assessments, evaluated current knowledge of associations between germline or somatic mutation and severe disease risk, and mined available data on the fraction of human germline mutations expected to cause severe disease. Based on this review and given that mutations are irreversible and some cause severe human disease, in regulatory settings where an ESAF is used, a majority of the WG recommends applying an ESAF value between 2 and 10 when deriving a HBGV from mutation data. This recommendation may need to be revisited in the future if direct measurement of disease-causing mutations by error-corrected next generation sequencing clarifies selection of ESAF values.
Collapse
Affiliation(s)
- Barbara L Parsons
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Marc A Beal
- Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Kerry L Dearfield
- U.S. Environmental Protection Agency and U.S. Department of Agriculture, Washington, DC, USA
| | - George R Douglas
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Min Gi
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | | | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | | | - Michelle Kenyon
- Portfolio and Regulatory Strategy, Drug Safety Research and Development, Pfizer, Groton, Connecticut, USA
| | - Alexandra S Long
- Existing Substances Risk Assessment Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - David P Lovell
- Population Health Research Institute, St George's Medical School, University of London, London, UK
| | | | - Meagan B Myers
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | | | - Alisa Vespa
- Pharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Andreas Zeller
- Pharmaceutical Sciences, pRED Innovation Center Basel, Hoffmann-La Roche Ltd, Basel, Switzerland
| | - George E Johnson
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
| | - Paul A White
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Vahle JL, Dybowski J, Graziano M, Hisada S, Lebron J, Nolte T, Steigerwalt R, Tsubota K, Sistare FD. ICH S1 prospective evaluation study and weight of evidence assessments: commentary from industry representatives. FRONTIERS IN TOXICOLOGY 2024; 6:1377990. [PMID: 38845817 PMCID: PMC11153695 DOI: 10.3389/ftox.2024.1377990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/03/2024] [Indexed: 06/09/2024] Open
Abstract
Industry representatives on the ICH S1B(R1) Expert Working Group (EWG) worked closely with colleagues from the Drug Regulatory Authorities to develop an addendum to the ICH S1B guideline on carcinogenicity studies that allows for a weight-of-evidence (WoE) carcinogenicity assessment in some cases, rather than conducting a 2-year rat carcinogenicity study. A subgroup of the EWG composed of regulators have published in this issue a detailed analysis of the Prospective Evaluation Study (PES) conducted under the auspices of the ICH S1B(R1) EWG. Based on the experience gained through the Prospective Evaluation Study (PES) process, industry members of the EWG have prepared the following commentary to aid sponsors in assessing the standard WoE factors, considering how novel investigative approaches may be used to support a WoE assessment, and preparing appropriate documentation of the WoE assessment for presentation to regulatory authorities. The commentary also reviews some of the implementation challenges sponsors must consider in developing a carcinogenicity assessment strategy. Finally, case examples drawn from previously marketed products are provided as a supplement to this commentary to provide additional examples of how WoE criteria may be applied. The information and opinions expressed in this commentary are aimed at increasing the quality of WoE assessments to ensure the successful implementation of this approach.
Collapse
Affiliation(s)
- John L. Vahle
- Lilly Research Laboratories, Indianapolis, IN, United States
| | - Joe Dybowski
- Alnylam Pharmaceuticals, Cambridge, MA, United States
| | | | - Shigeru Hisada
- Formerly ASKA Pharmaceutical Co., Ltd., Fujisawa-shi, Kanagawa, Japan
| | - Jose Lebron
- Merck & Co., Inc., Rahway, NJ, United States
| | - Thomas Nolte
- Development NCE, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | | | |
Collapse
|
7
|
Gourmet L, Walker-Samuel S, Mallick P. Examination of the role of mutualism in immune evasion. Front Oncol 2024; 14:1406744. [PMID: 38779085 PMCID: PMC11109368 DOI: 10.3389/fonc.2024.1406744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Though the earliest stages of oncogenesis, post initiation, are not well understood, it is generally appreciated that a successful transition from a collection of dysregulated cells to an aggressive tumour requires complex ecological interactions between cancer cells and their environment. One key component of tumorigenesis is immune evasion. To investigate the interplay amongst the ecological behaviour of mutualism and immune evasion, we used a computational simulation framework. Sensitivity analyses of the growth of a virtual tumour implemented as a 2D-hexagonal lattice model suggests tumour survival depends on the interplay between growth rates, mutualism and immune evasion. In 60% of simulations, cancer clones with low growth rates, but exhibiting mutualism were able to evade the immune system and continue progressing suggesting that tumours with equivalent growth rates and no mutualism are more likely to be eliminated than tumours with mutualism. Tumours with faster growth rates showed a lower dependence upon mutualism for progression. Geostatistical analysis showed decreased spatial heterogeneity over time for polyclonal tumours with a high division rate. Overall, these results suggest that in slow growing tumours, mutualism is critical for early tumorigenesis.
Collapse
Affiliation(s)
- Lucie Gourmet
- Centre for Computational Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Simon Walker-Samuel
- Centre for Computational Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Parag Mallick
- Canary Center for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
8
|
Qin L, Shi L, Wang Y, Yu H, Du Z, Chen M, Cai Y, Cao Y, Deng S, Wang J, Cheng D, Heng Y, Xu J, Cai K, Wu K. Fumarate Hydratase Enhances the Therapeutic Effect of PD-1 Antibody in Colorectal Cancer by Regulating PCSK9. Cancers (Basel) 2024; 16:713. [PMID: 38398104 PMCID: PMC10887080 DOI: 10.3390/cancers16040713] [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: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Despite the notable achievements of programmed death 1 (PD-1) antibodies in treating various cancers, the overall efficacy remains limited in the majority of colorectal cancer (CRC) cases. Metabolism reprogramming of tumors inhibits the tricarboxylic acid (TCA) cycle, leading to down-regulation of fumarate hydratase (FH), which is related to poor prognosis in CRC patients. By establishing a tumor-bearing mouse model of CRC with Fh1 expression deficiency, we confirmed that the therapeutic effect of PD-1 antibodies alone was suboptimal in mice with low Fh1 expression, which was improved by combination with a protein invertase subtilisin/kexin 9 (PCSK9) inhibitor. Mechanistically, FH binds to Ras-related nucleoprotein (RAN), which inhibits the nuclear import of the PCSK9 transcription factor SREBF1/2, thus reducing the expression of PCSK9. This leads to increased clonal expansion of CD8+ T cells while the number of Tregs remains unchanged, and the expression of PD-L1 does not change significantly, thus enhancing the immunotherapy response. On the contrary, the expression of PCSK9 increased in CRC cells with low FH expression, which antagonized the effects of immunotherapy. Overall, CRC patients with low FH expression may benefit from combinatorial therapy with PD-1 antibodies and PCSK9 inhibitors to enhance the curative effect.
Collapse
Affiliation(s)
- Le Qin
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China; (Y.H.); (J.X.)
| | - Liang Shi
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Yu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Haixin Yu
- Department of Digestive Surgical Oncology, Cancer Center, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (H.Y.); (Z.D.); (Y.C.)
| | - Zhouyuan Du
- Department of Digestive Surgical Oncology, Cancer Center, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (H.Y.); (Z.D.); (Y.C.)
| | - Mian Chen
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Yuxuan Cai
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Yinghao Cao
- Department of Digestive Surgical Oncology, Cancer Center, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (H.Y.); (Z.D.); (Y.C.)
| | - Shenghe Deng
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Jun Wang
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Denglong Cheng
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Yixin Heng
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China; (Y.H.); (J.X.)
| | - Jiaxin Xu
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China; (Y.H.); (J.X.)
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| | - Ke Wu
- Department of Gastrointestinal Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; (L.Q.); (L.S.); (M.C.); (Y.C.); (S.D.); (J.W.); (D.C.)
| |
Collapse
|
9
|
Wan M, Yang X, He L, Meng H. Elucidating the clonal relationship of esophageal second primary tumors in patients with laryngeal squamous cell carcinoma. Infect Agent Cancer 2023; 18:75. [PMID: 38017473 PMCID: PMC10685475 DOI: 10.1186/s13027-023-00558-z] [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: 09/01/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
Laryngeal cancer ranks as the second most prevalent upper airway malignancy, following Lung cancer. Although some progress has been made in managing laryngeal cancer, the 5-year survival rate is disappointing. The gradual increase in the incidence of second primary tumors (SPTs) plays a crucial role in determining survival outcomes during long-term follow-up, and the esophagus was the most common site with a worse prognosis. In clinical practice, the treatment of esophageal second primary tumors (ESPT) in patients with laryngeal squamous cell carcinoma (LSCC) has always been challenging. For patients with synchronous tumors, several treatment modalities, such as radiotherapy, chemotherapy and potentially curative surgery are necessary but are typically poorly tolerated. Secondary cancer therapy options for metachronous patients are always constrained by index cancer treatment indications. Therefore, understanding the clonal origin of the second primary tumor may be an important issue in the treatment of patients. LSCC cells demonstrate genetic instability because of two distinct aetiologies (human papillomavirus (HPV)-negative and HPV-positive) disease. Various etiologies exhibit distinct oncogenic mechanisms, which subsequently impact the tissue microenvironment. The condition of the tissue microenvironment plays a crucial role in determining the destiny and clonal makeup of mutant cells during the initial stages of tumorigenesis. This review focuses on the genetic advances of LSCC, the current research status of SPT, and the influence of key carcinogenesis of HPV-positive and HPV-negative LSCC on clonal evolution of ESPT cells. The objective is to gain a comprehensive understanding of the molecular basis underlying the clonal origins of SPT, thereby offering novel perspectives for future investigations in this field.
Collapse
Affiliation(s)
- Meixuan Wan
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xinxin Yang
- Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Lin He
- Department of Stomatology, Heilongjiang Province Hospital, Harbin, 150081, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
- Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
10
|
Menon V, Brash DE. Next-generation sequencing methodologies to detect low-frequency mutations: "Catch me if you can". MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 792:108471. [PMID: 37716438 PMCID: PMC10843083 DOI: 10.1016/j.mrrev.2023.108471] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/18/2023]
Abstract
Mutations, the irreversible changes in an organism's DNA sequence, are present in tissues at a variant allele frequency (VAF) ranging from ∼10-8 per bp for a founder mutation to ∼10-3 for a histologically normal tissue sample containing several independent clones - compared to 1%- 50% for a heterozygous tumor mutation or a polymorphism. The rarity of these events poses a challenge for accurate clinical diagnosis and prognosis, toxicology, and discovering new disease etiologies. Standard Next-Generation Sequencing (NGS) technologies report VAFs as low as 0.5% per nt, but reliably observing rarer precursor events requires additional sophistication to measure ultralow-frequency mutations. We detail the challenge; define terms used to characterize the results, which vary between laboratories and sometimes conflict between biologists and bioinformaticists; and describe recent innovations to improve standard NGS methodologies including: single-strand consensus sequence methods such as Safe-SeqS and SiMSen-Seq; tandem-strand consensus sequence methods such as o2n-Seq and SMM-Seq; and ultrasensitive parent-strand consensus sequence methods such as DuplexSeq, PacBio HiFi, SinoDuplex, OPUSeq, EcoSeq, BotSeqS, Hawk-Seq, NanoSeq, SaferSeq, and CODEC. Practical applications are also noted. Several methods quantify VAF down to 10-5 at a nt and mutation frequency (MF) in a target region down to 10-7 per nt. By expanding to > 1 Mb of sites never observed twice, thus forgoing VAF, other methods quantify MF < 10-9 per nt or < 15 errors per haploid genome. Clonal expansion cannot be directly distinguished from independent mutations by sequencing, so it is essential for a paper to report whether its MF counted only different mutations - the minimum independent-mutation frequency MFminI - or all mutations observed including recurrences - the larger maximum independent-mutation frequency MFmaxI which may reflect clonal expansion. Ultrasensitive methods reveal that, without their use, even mutations with VAF 0.5-1% are usually spurious.
Collapse
Affiliation(s)
- Vijay Menon
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520-8040, USA.
| | - Douglas E Brash
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520-8040, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT 06520-8059, USA; Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520-8028, USA.
| |
Collapse
|
11
|
Feller G, Khammissa RAG, Ballyram R, Beetge MM, Lemmer J, Feller L. Tumour Genetic Heterogeneity in Relation to Oral Squamous Cell Carcinoma and Anti-Cancer Treatment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2392. [PMID: 36767758 PMCID: PMC9915085 DOI: 10.3390/ijerph20032392] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
Oral squamous cell carcinoma (SCC) represents more than 90% of all oral cancers and is the most frequent SCC of the head and neck region. It may affect any oral mucosal subsite but most frequently the tongue, followed by the floor of the mouth. The use of tobacco and betel nut, either smoked or chewed, and abuse of alcohol are the main risk factors for oral SCC. Oral SCC is characterized by considerable genetic heterogeneity and diversity, which together have a significant impact on the biological behaviour, clinical course, and response to treatment and on the generally poor prognosis of this carcinoma. Characterization of spatial and temporal tumour-specific molecular profiles and of person-specific resource availability and environmental and biological selective pressures could assist in personalizing anti-cancer treatment for individual patients, with the aim of improving treatment outcomes. In this narrative review, we discuss some of the events in cancer evolution and the functional significance of driver-mutations in carcinoma-related genes in general and elaborate on mechanisms mediating resistance to anti-cancer treatment.
Collapse
Affiliation(s)
- Gal Feller
- Department of Radiation Oncology, University of Witwatersrand, Johannesburg and Charlotte Maxeke Academic Hospital, Johannesburg 2193, South Africa
| | - Razia Abdool Gafaar Khammissa
- Department of Periodontics and Oral Medicine, School of Oral Health Sciences, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa
| | - Raoul Ballyram
- Department of Periodontology and Oral Medicine, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
| | - Mia-Michaela Beetge
- Department of Periodontics and Oral Medicine, School of Oral Health Sciences, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa
| | - Johan Lemmer
- Retired Professor, Silvela Street, Sandton, Johannesburg 2031, South Africa
| | - Liviu Feller
- Retired Professor, Bantry Bay, Cape Town 8005, South Africa
| |
Collapse
|
12
|
Adamczuk G, Humeniuk E, Adamczuk K, Grabarska A, Dudka J. 2,4-Dinitrophenol as an Uncoupler Augments the Anthracyclines Toxicity against Prostate Cancer Cells. Molecules 2022; 27:7227. [PMID: 36364051 PMCID: PMC9655928 DOI: 10.3390/molecules27217227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/25/2023] Open
Abstract
One of the strategies for the treatment of advanced cancer diseases is targeting the energy metabolism of the cancer cells. The compound 2,4-DNP (2,4-dinitrophenol) disrupts the cell energy metabolism through the ability to decouple oxidative phosphorylation. The aim of the study was to determine the ability of 2,4-DNP to sensitize prostate cancer cells with different metabolic phenotypes to the action of known anthracyclines (doxorubicin and epirubicin). The synergistic effect of the anthracyclines and 2,4-DNP was determined using an MTT assay, apoptosis detection and a cell cycle analysis. The present of oxidative stress in cancer cells was assessed by CellROX, the level of cellular thiols and DNA oxidative damage. The study revealed that the incubation of LNCaP prostate cancer cells (oxidative phenotype) with epirubicin and doxorubicin simultaneously with 2,4-DNP showed the presence of a synergistic effect for both the cytostatics. Moreover, it contributes to the increased induction of oxidative stress, which results in a reduced level of cellular thiols and an increased number of AP sites in the DNA. The synergistic activity may consist of an inhibition of ATP synthesis and the simultaneous production of toxic amounts of ROS, destroying the mitochondria. Additionally, the sensitivity of the LNCaP cell line to the anthracyclines is relatively higher compared to the other two (PC-3, DU-145).
Collapse
Affiliation(s)
- Grzegorz Adamczuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
| | - Ewelina Humeniuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
| | - Kamila Adamczuk
- Department of Biochemistry and Molecular Biology, Faculty of Medical Sciences, Medical University of Lublin, 20-090 Lublin, Poland
| | - Aneta Grabarska
- Department of Biochemistry and Molecular Biology, Faculty of Medical Sciences, Medical University of Lublin, 20-090 Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
13
|
Rahal Z, Sinjab A, Wistuba II, Kadara H. Game of clones: Battles in the field of carcinogenesis. Pharmacol Ther 2022; 237:108251. [PMID: 35850404 PMCID: PMC10249058 DOI: 10.1016/j.pharmthera.2022.108251] [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: 03/01/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022]
Abstract
Recent advances in bulk sequencing approaches as well as genomic decoding at the single-cell level have revealed surprisingly high somatic mutational burdens in normal tissues, as well as increased our understanding of the landscape of "field cancerization", that is, molecular and immune alterations in mutagen-exposed normal-appearing tissues that recapitulated those present in tumors. Charting the somatic mutational landscapes in normal tissues can have strong implications on our understanding of how tumors arise from mutagenized epithelium. Making sense of those mutations to understand the progression along the pathologic continuum of normal epithelia, preneoplasias, up to malignant tissues will help pave way for identification of ideal targets that can guide new strategies for preventing or eliminating cancers at their earliest stages of development. In this review, we will provide a brief history of field cancerization and its implications on understanding early stages of cancer pathogenesis and deviation from the pathologically "normal" state. The review will provide an overview of how mutations accumulating in normal tissues can lead to a patchwork of mutated cell clones that compete while maintaining an overall state of functional homeostasis. The review also explores the role of clonal competition in directing the fate of normal tissues and summarizes multiple mechanisms elicited in this phenomenon and which have been linked to cancer development. Finally, we highlight the importance of understanding mutations in normal tissues, as well as clonal competition dynamics (in both the epithelium and the microenvironment) and their significance in exploring new approaches to combatting cancer.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, USA
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, USA.
| |
Collapse
|
14
|
Corton JC, Mitchell CA, Auerbach S, Bushel P, Ellinger-Ziegelbauer H, Escobar PA, Froetschl R, Harrill AH, Johnson K, Klaunig JE, Pandiri AR, Podtelezhnikov AA, Rager JE, Tanis KQ, van der Laan JW, Vespa A, Yauk CL, Pettit SD, Sistare FD. A Collaborative Initiative to Establish Genomic Biomarkers for Assessing Tumorigenic Potential to Reduce Reliance on Conventional Rodent Carcinogenicity Studies. Toxicol Sci 2022; 188:4-16. [PMID: 35404422 PMCID: PMC9238304 DOI: 10.1093/toxsci/kfac041] [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] [Indexed: 11/24/2022] Open
Abstract
There is growing recognition across broad sectors of the scientific community that use of genomic biomarkers has the potential to reduce the need for conventional rodent carcinogenicity studies of industrial chemicals, agrochemicals, and pharmaceuticals through a weight-of-evidence approach. These biomarkers fall into 2 major categories: (1) sets of gene transcripts that can identify distinct tumorigenic mechanisms of action; and (2) cancer driver gene mutations indicative of rapidly expanding growth-advantaged clonal cell populations. This call-to-action article describes a collaborative approach launched to develop and qualify biomarker gene expression panels that measure widely accepted molecular pathways linked to tumorigenesis and their activation levels to predict tumorigenic doses of chemicals from short-term exposures. Growing evidence suggests that application of such biomarker panels in short-term exposure rodent studies can identify both tumorigenic hazard and tumorigenic activation levels for chemical-induced carcinogenicity. In the future, this approach will be expanded to include methodologies examining mutations in key cancer driver gene mutation hotspots as biomarkers of both genotoxic and nongenotoxic chemical tumor risk. Analytical, technical, and biological validation studies of these complementary genomic tools are being undertaken by multisector and multidisciplinary collaborative teams within the Health and Environmental Sciences Institute. Success from these efforts will facilitate the transition from current heavy reliance on conventional 2-year rodent carcinogenicity studies to more rapid animal- and resource-sparing approaches for mechanism-based carcinogenicity evaluation supporting internal and regulatory decision-making.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Constance A Mitchell
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Scott Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Pierre Bushel
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | | | - Patricia A Escobar
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Roland Froetschl
- BfArM-Bundesinstitut für Arzneimittel und Medizinprodukte, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Alison H Harrill
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - James E Klaunig
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public Health, Indiana University, Bloomington, Indiana, USA
| | - Arun R Pandiri
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - Julia E Rager
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Keith Q Tanis
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Jan Willem van der Laan
- Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht, The Netherlands
| | - Alisa Vespa
- Therapeutic Products Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Syril D Pettit
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Frank D Sistare
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
15
|
Preferential X Chromosome Inactivation as a Mechanism to Explain Female Preponderance in Myasthenia Gravis. Genes (Basel) 2022; 13:genes13040696. [PMID: 35456502 PMCID: PMC9031138 DOI: 10.3390/genes13040696] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Myasthenia gravis (MG) is a neuromuscular autoimmune disease characterized by prevalence in young women (3:1). Several mechanisms proposed as explanations for gender bias, including skewed X chromosome inactivation (XCI) and dosage or sex hormones, are often involved in the development of autoimmunity. The skewed XCI pattern can lead to an unbalanced expression of some X-linked genes, as observed in several autoimmune disorders characterized by female predominance. No data are yet available regarding XCI and MG. We hypothesize that the preferential XCI pattern may contribute to the female bias observed in the onset of MG, especially among younger women. XCI analysis was performed on blood samples of 284 women between the ages of 20 and 82. XCI was tested using the Human Androgen Receptor Assay (HUMARA). XCI patterns were classified as random (XCI < 75%) and preferential (XCI ≥ 75%). In 121 informative patients, the frequency of skewed XCI patterns was 47%, significantly higher than in healthy controls (17%; p ≤ 0.00001). Interestingly, the phenomenon was observed mainly in younger patients (<45 years; p ≤ 0.00001). Furthermore, considering the XCI pattern and the other clinical characteristics of patients, no significant differences were found. In conclusion, we observed preferential XCI in MG female patients, suggesting its potential role in the aetiology of MG, as observed in other autoimmune diseases in women.
Collapse
|
16
|
Simon Davis DA, Mun S, Smith JM, Hammill D, Garrett J, Gosling K, Price J, Elsaleh H, Syed FM, Atmosukarto II, Quah BJC. Machine learning predicts cancer subtypes and progression from blood immune signatures. PLoS One 2022; 17:e0264631. [PMID: 35226704 PMCID: PMC8884497 DOI: 10.1371/journal.pone.0264631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/14/2022] [Indexed: 12/23/2022] Open
Abstract
Clinical adoption of immune checkpoint inhibitors in cancer management has highlighted the interconnection between carcinogenesis and the immune system. Immune cells are integral to the tumour microenvironment and can influence the outcome of therapies. Better understanding of an individual's immune landscape may play an important role in treatment personalisation. Peripheral blood is a readily accessible source of information to study an individual's immune landscape compared to more complex and invasive tumour bioipsies, and may hold immense diagnostic and prognostic potential. Identifying the critical components of these immune signatures in peripheral blood presents an attractive alternative to tumour biopsy-based immune phenotyping strategies. We used two syngeneic solid tumour models, a 4T1 breast cancer model and a CT26 colorectal cancer model, in a longitudinal study of the peripheral blood immune landscape. Our strategy combined two highly accessible approaches, blood leukocyte immune phenotyping and plasma soluble immune factor characterisation, to identify distinguishing immune signatures of the CT26 and 4T1 tumour models using machine learning. Myeloid cells, specifically neutrophils and PD-L1-expressing myeloid cells, were found to correlate with tumour size in both the models. Elevated levels of G-CSF, IL-6 and CXCL13, and B cell counts were associated with 4T1 growth, whereas CCL17, CXCL10, total myeloid cells, CCL2, IL-10, CXCL1, and Ly6Cintermediate monocytes were associated with CT26 tumour development. Peripheral blood appears to be an accessible means to interrogate tumour-dependent changes to the host immune landscape, and to identify blood immune phenotypes for future treatment stratification.
Collapse
Affiliation(s)
| | - Sahngeun Mun
- Irradiation Immunity Interaction Lab, Canberra, ACT, Australia
| | | | - Dillon Hammill
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jessica Garrett
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Katharine Gosling
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jason Price
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Hany Elsaleh
- Radiation Oncology Department, The Alfred, Melbourne, VIC, Australia
| | - Farhan M. Syed
- Irradiation Immunity Interaction Lab, Canberra, ACT, Australia
- Radiation Oncology Department, Canberra Hospital, Canberra Health Services, Canberra, ACT, Australia
| | - Ines I. Atmosukarto
- Irradiation Immunity Interaction Lab, Canberra, ACT, Australia
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Benjamin J. C. Quah
- Irradiation Immunity Interaction Lab, Canberra, ACT, Australia
- Radiation Oncology Department, Canberra Hospital, Canberra Health Services, Canberra, ACT, Australia
| |
Collapse
|
17
|
Indave Ruiz BI, Armon S, Watanabe R, Uttley L, White VA, Lazar AJ, Cree IA. Clonality, Mutation and Kaposi Sarcoma: A Systematic Review. Cancers (Basel) 2022; 14:1201. [PMID: 35267506 PMCID: PMC8909603 DOI: 10.3390/cancers14051201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND It remains uncertain whether Kaposi sarcoma (KS) is a true neoplasm, in that it regresses after removal of the stimulus to growth (as HHV8) when immunosuppression is reduced. We aimed to summarize the available evidence on somatic mutations and clonality within KS to assess whether KS is a neoplasm or not. METHODS Medline and Web of Science were searched until September 2020 for articles on clonality or mutation in KS. Search strings were supervised by expert librarians, and two researchers independently performed study selection and data extraction. An adapted version of the QUADAS2 tool was used for methodological quality appraisal. RESULTS Of 3077 identified records, 20 publications reported on relevant outcomes and were eligible for qualitative synthesis. Five studies reported on clonality, 10 studies reported on various mutations, and 5 studies reported on chromosomal aberrations in KS. All studies were descriptive and were judged to have a high risk of bias. There was considerable heterogeneity of results with respect to clonality, mutation and cytogenetic abnormalities as well as in terms of types of lesions and patient characteristics. CONCLUSIONS While KS certainly produces tumours, the knowledge is currently insufficient to determine whether KS is a clonal neoplasm (sarcoma), or simply an aggressive reactive virus-driven lesion.
Collapse
Affiliation(s)
- Blanca Iciar Indave Ruiz
- International Agency for Research on Cancer (IARC), World Health Organization, 69372 Lyon, France; (S.A.); (R.W.); (V.A.W.); (I.A.C.)
| | - Subasri Armon
- International Agency for Research on Cancer (IARC), World Health Organization, 69372 Lyon, France; (S.A.); (R.W.); (V.A.W.); (I.A.C.)
| | - Reiko Watanabe
- International Agency for Research on Cancer (IARC), World Health Organization, 69372 Lyon, France; (S.A.); (R.W.); (V.A.W.); (I.A.C.)
| | - Lesley Uttley
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield S1 4DA, UK;
| | - Valerie A. White
- International Agency for Research on Cancer (IARC), World Health Organization, 69372 Lyon, France; (S.A.); (R.W.); (V.A.W.); (I.A.C.)
| | - Alexander J. Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Ian A. Cree
- International Agency for Research on Cancer (IARC), World Health Organization, 69372 Lyon, France; (S.A.); (R.W.); (V.A.W.); (I.A.C.)
| |
Collapse
|
18
|
Lim CA, Banyi N, Tucker T, Ionescu DN, Melosky B. A Case of ALK-Rearranged Combined Lung Adenocarcinoma and Neuroendocrine Carcinoma with Diffuse Bone Metastasis and Partial Response to Alectinib. Curr Oncol 2022; 29:848-852. [PMID: 35200571 PMCID: PMC8870951 DOI: 10.3390/curroncol29020072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 11/16/2022] Open
Abstract
We report a rare case of stage IV pulmonary combined large-cell neuroendocrine carcinoma (LCNEC) and adenocarcinoma (ACA), both demonstrating anaplastic lymphoma kinase (ALK) rearrangement by IHC and FISH. This 61-year-old lifelong nonsmoking Asian woman presented with a cough, and after diagnosis and surgical treatment, completed four cycles of adjuvant cisplatin and etoposide chemotherapy. She subsequently developed recurrence with bony metastases of exclusively ALK-positive LCNEC. Alectinib was started, and the patient experienced a partial response.
Collapse
Affiliation(s)
- Chloe A. Lim
- MD Undergraduate Program, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; (C.A.L.); (N.B.)
- Internal Medicine Residency Program, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Norbert Banyi
- MD Undergraduate Program, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; (C.A.L.); (N.B.)
- Department of Pathology, BC Cancer, Vancouver, BC V6T 1Z1, Canada;
| | - Tracy Tucker
- Cancer Genetics and Genomics Laboratory, Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, BC V6T 1Z1, Canada;
| | - Diana N. Ionescu
- Department of Pathology, BC Cancer, Vancouver, BC V6T 1Z1, Canada;
| | - Barbara Melosky
- Medical Oncology, BC Cancer, Vancouver, BC V6T 1Z1, Canada
- Correspondence: ; Tel.: +1-604-877-6000
| |
Collapse
|
19
|
Schoultz E, Johansson E, Moccia C, Jakubikova I, Ravi N, Liang S, Carlsson T, Montelius M, Patyra K, Kero J, Paulsson K, Fagman H, Bergo MO, Nilsson M. Tissue architecture delineates field cancerization in BRAFV600E-induced tumor development. Dis Model Mech 2022; 15:dmm048887. [PMID: 34379110 PMCID: PMC8380047 DOI: 10.1242/dmm.048887] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
Cancer cells hijack developmental growth mechanisms but whether tissue morphogenesis and architecture modify tumorigenesis is unknown. Here, we characterized a new mouse model of sporadic thyroid carcinogenesis based on inducible expression of BRAF carrying a Val600 Glu (V600E) point mutation (BRAFV600E) from the thyroglobulin promoter (TgCreERT2). Spontaneous activation of this Braf-mutant allele due to leaky activity of the Cre recombinase revealed that intrinsic properties of thyroid follicles determined BRAF-mutant cell fate. Papillary thyroid carcinomas developed multicentrically within a normal microenvironment. Each tumor originated from a single follicle that provided a confined space for growth of a distinct tumor phenotype. Lineage tracing revealed oligoclonal tumor development in infancy and early selection of BRAFV600E kinase inhibitor-resistant clones. Somatic mutations were few, non-recurrent and limited to advanced tumors. Female mice developed larger tumors than males, reproducing the gender difference of human thyroid cancer. These data indicate that BRAFV600E-induced tumorigenesis is spatiotemporally regulated depending on the maturity and heterogeneity of follicles. Moreover, thyroid tissue organization seems to determine whether a BRAF-mutant lineage becomes a cancerized lineage. The TgCreERT2;BrafCA/+ sporadic thyroid cancer mouse model provides a new tool to evaluate drug therapy at different stages of tumor evolution.
Collapse
Affiliation(s)
- Elin Schoultz
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Ellen Johansson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Carmen Moccia
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Iva Jakubikova
- Faculty of Medicine, Charles University, Hradec Kralove, Czech Republic
| | - Naveen Ravi
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund SE-22184, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Therese Carlsson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Mikael Montelius
- Department of Radiology, Institute of Clinical Sciences, University of Gothenburg, SE-41345 Göteborg, Sweden
| | - Konrad Patyra
- Department of Endocrinology, University of Turku, Åbo FI-20521, Finland
| | - Jukka Kero
- Department of Endocrinology, University of Turku, Åbo FI-20521, Finland
| | - Kajsa Paulsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund SE-22184, Sweden
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
- Department of Clinical Pathology, Sahlgrenska University Hospital, Göteborg SE-41345, Sweden
| | - Martin O. Bergo
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge SE-14183, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| |
Collapse
|
20
|
Zhong Z, Vong CT, Chen F, Tan H, Zhang C, Wang N, Cui L, Wang Y, Feng Y. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: Helping to fight against cancer via multiple targets. Med Res Rev 2022; 42:1246-1279. [PMID: 35028953 PMCID: PMC9306614 DOI: 10.1002/med.21876] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Immunotherapy sheds new light to cancer treatment and is satisfied by cancer patients. However, immunotoxicity, single‐source antibodies, and single‐targeting stratege are potential challenges to the success of cancer immunotherapy. A huge number of promising lead compounds for cancer treatment are of natural origin from herbal medicines. The application of natural products from herbal medicines that have immunomodulatory properties could alter the landscape of immunotherapy drastically. The present study summarizes current medication for cancer immunotherapy and discusses the potential chemicals from herbal medicines as immune checkpoint inhibitors that have a broad range of immunomodulatory effects. Therefore, this review provides valuable insights into the efficacy and mechanism of actions of cancer immunotherapies, including natural products and combined treatment with immune checkpoint inhibitors, which could confer an improved clinical outcome for cancer treatment.
Collapse
Affiliation(s)
- Zhangfeng Zhong
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Chi Teng Vong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Feiyu Chen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Horyue Tan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
21
|
Gomes CC. Recurrent driver mutations in benign tumors. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 789:108412. [PMID: 35690415 DOI: 10.1016/j.mrrev.2022.108412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 06/15/2023]
Abstract
The understanding of the molecular pathogenesis of benign tumors may bring essential information to clarify the process of tumorigenesis, and ultimately improve the understanding of events such as malignant transformation. The definition of benign neoplasia is not always straightforward and herein the issues surrounding this concept are discussed. Benign neoplasms share all cancer hallmarks with malignancies, except for metastatic potential. Recently, next-generation sequencing has provided unprecedented opportunities to unravel the genetic basis of benign neoplasms and, so far, we have learned that benign neoplasms are indeed characterized by the presence of genetic mutations, including genes rearrangements. Driver mutations in advanced cancer are those that confer growth advantage, and which have been positively selected during cancer evolution. Herein, some discussion will be brought about this concept in the context of cancer prevention, involving precursor lesions and benign neoplasms. When considering early detection and cancer prevention, a driver mutation should not only be advantageous (i.e., confer survival advantage), but predisposing (i.e., promoting a cancer phenotype). By including the benign counterparts of malignant neoplasms in tumor biology studies, it is possible to evaluate the risk posed by a given mutation and to differentiate advantageous from predisposing mutations, further refining the concept of driver mutations. Therefore, the study of benign neoplasms should be encouraged because it provides valuable information on tumorigenesis central for understanding the progression from initiation to malignant transformation.
Collapse
Affiliation(s)
- Carolina Cavalieri Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
22
|
Harris KL, McKim KL, Myers MB, Gong B, Xu J, Parsons BL. Assessment of clonal expansion using CarcSeq measurement of lung cancer driver mutations and correlation with mouse strain- and sex-related incidence of spontaneous lung neoplasia. Toxicol Sci 2021; 184:1-14. [PMID: 34373914 DOI: 10.1093/toxsci/kfab098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Quantification of variation in levels of spontaneously occurring cancer driver mutations (CDMs) was developed to assess clonal expansion and predict future risk of neoplasm development. Specifically, an error-corrected next generation sequencing method, CarcSeq, and a mouse CarcSeq panel (analogous to human and rat panels) were developed and used to quantify low-frequency mutations in a panel of amplicons enriched in hotspot CDMs. Mutations in a subset of panel amplicons, Braf, Egfr, Kras, Stk11 and Tp53, were related to incidence of lung neoplasms at two years. This was achieved by correlating median absolute deviation (MAD) from the overall median mutant fraction (MF) measured in the lung DNA of 16-week-old male and female, B6C3F1 and CD-1 mice (10 mice/sex/strain) with percentages of spontaneous alveolar/bronchioloalveolar adenomas and carcinomas reported in bioassay control groups. 1,586 mouse lung mutants with MFs >1 x 10-4 were recovered. The ratio of non-synonymous to synonymous mutations was used to assess the proportion of recovered mutations conferring a positive selective advantage. The greatest ratio was observed in what is considered the most lung tumor-sensitive model examined, male B6C3F1 mice. Of the recurrent, non-synonymous mouse mutations recovered, 55.5% have been reported in human tumors, with many located in or around the mouse equivalent of human cancer hotspot codons. MAD for the same subset of amplicons measured in normal human lung DNA samples showed a correlation of moderate strength and borderline significance) with age (a cancer risk factor), as well as age-related cumulative lung cancer risk, suggesting MAD may inform species extrapolation.
Collapse
Affiliation(s)
- Kelly L Harris
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Genetic and Molecular Toxicology, 3900 NCTR Rd, Jefferson, AR, 72079
| | - Karen L McKim
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Genetic and Molecular Toxicology, 3900 NCTR Rd, Jefferson, AR, 72079
| | - Meagan B Myers
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Genetic and Molecular Toxicology, 3900 NCTR Rd, Jefferson, AR, 72079
| | - Binsheng Gong
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Bioinformatics and Biostatistics, 3900 NCTR Rd, Jefferson, AR, 72079
| | - Joshua Xu
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Bioinformatics and Biostatistics, 3900 NCTR Rd, Jefferson, AR, 72079
| | - Barbara L Parsons
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Genetic and Molecular Toxicology, 3900 NCTR Rd, Jefferson, AR, 72079
| |
Collapse
|
23
|
Leystra AA, Gilsdorf BJ, Wisinger AM, Warda ER, Wiegand S, Zahm CD, Matkowskyj KA, Deming DA, Khan N, Rosemarie Q, Sievers CK, Schwartz AR, Albrecht DM, Clipson L, Mukhtar H, Newton MA, Halberg RB. Multi-ancestral origin of intestinal tumors: Impact on growth, progression, and drug efficacy. Cancer Rep (Hoboken) 2021; 5:e1459. [PMID: 34245130 PMCID: PMC8842699 DOI: 10.1002/cnr2.1459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/24/2021] [Accepted: 04/21/2021] [Indexed: 11/07/2022] Open
Abstract
Background Data are steadily accruing that demonstrate that intestinal tumors are frequently derived from multiple founding cells, resulting in tumors comprised of distinct ancestral clones that might cooperate or alternatively compete, thereby potentially impacting different phases of the disease process. Aim We sought to determine whether tumors with a multi‐ancestral architecture involving at least two distinct clones show increased tumor number, growth, progression, or resistance to drug intervention. Methods Mice carrying the Min allele of Apc were generated that were mosaic with only a subset of cells in the intestinal epithelium expressing an activated form of PI3K, a key regulatory kinase affecting several important cellular processes. These cells were identifiable as they fluoresced green, whereas all other cells fluoresced red. Results Cell lineage tracing revealed that many intestinal tumors from our mouse model were derived from at least two founding cells, those expressing the activated PI3K (green) and those which did not (red). Heterotypic tumors with a multi‐ancestral architecture as evidenced by a mixture of green and red cells exhibited increased tumor growth and invasiveness. Clonal architecture also had an impact on tumor response to low‐dose aspirin. Aspirin treatment resulted in a greater reduction of heterotypic tumors derived from multiple founding cells as compared to tumors derived from a single founding cell. Conclusion These data indicate that genetically distinct tumor‐founding cells can contribute to early intratumoral heterogeneity. The coevolution of the founding cells and their progeny enhances colon tumor progression and impacts the response to aspirin. These findings are important to a more complete understanding of tumorigenesis with consequences for several distinct models of tumor evolution. They also have practical implications to the clinic. Mouse models with heterogenous tumors are likely better for predicting drug efficacy as compared to models in which the tumors are highly homogeneous. Moreover, understanding how interactions among different populations in a single heterotypic tumor with a multi‐ancestral architecture impact response to a single agent and combination therapies are necessary to fully develop personalized medicine.
Collapse
Affiliation(s)
- Alyssa A Leystra
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Division of Gastroenterology and Hepatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Brock J Gilsdorf
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Amanda M Wisinger
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Elise R Warda
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Shanna Wiegand
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Christopher D Zahm
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kristina A Matkowskyj
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Dustin A Deming
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Naghma Khan
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Quincy Rosemarie
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Chelsie K Sievers
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Division of Gastroenterology and Hepatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Alexander R Schwartz
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Dawn M Albrecht
- Division of Gastroenterology and Hepatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Linda Clipson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Hasan Mukhtar
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Michael A Newton
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Richard B Halberg
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,Division of Gastroenterology and Hepatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
24
|
Thresholds for carcinogens. Chem Biol Interact 2021; 341:109464. [PMID: 33823170 DOI: 10.1016/j.cbi.2021.109464] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
Current regulatory cancer risk assessment principles and practices assume a linear dose-response relationship-the linear no-threshold (LNT) model-that theoretically estimates cancer risks occurring following low doses of carcinogens by linearly extrapolating downward from experimentally determined risks at high doses. The two-year rodent bioassays serve as experimental vehicles to determine the high-dose cancer risks in animals and then to predict, by extrapolation, the number of carcinogen-induced tumors (tumor incidence) that will arise during the lifespans of humans who are exposed to environmental carcinogens at doses typically orders of magnitude below those applied in the rodent assays. An integrated toxicological analysis is conducted herein to reconsider an alternative and once-promising approach, tumor latency, for estimating carcinogen-induced cancer risks at low doses. Tumor latency measures time-to-tumor following exposure to a carcinogen, instead of tumor incidence. Evidence for and against the concept of carcinogen-induced tumor latency is presented, discussed, and then examined with respect to its relationship to dose, dose rates, and the dose-related concepts of initiation, tumor promotion, tumor regression, tumor incidence, and hormesis. Considerable experimental evidence indicates: (1) tumor latency (time-to-tumor) is inversely related to the dose of carcinogens and (2) lower doses of carcinogens display quantifiably discrete latency thresholds below which the promotion and, consequently, the progression and growth of tumors are delayed or prevented during a normal lifespan. Besides reconciling well with the concept of tumor promotion, such latency thresholds also reconcile favorably with the existence of thresholds for tumor incidence, the stochastic processes of tumor initiation, and the compensatory repair mechanisms of hormesis. Most importantly, this analysis and the arguments presented herein provide sound theoretical, experimental, and mechanistic rationales for rethinking the foundational premises of low-dose linearity and updating the current practices of cancer risk assessment to include the concept of carcinogen thresholds.
Collapse
|
25
|
Chauhan S, Dhawan DK, Saini A, Preet S. Antimicrobial peptides against colorectal cancer-a focused review. Pharmacol Res 2021; 167:105529. [PMID: 33675962 DOI: 10.1016/j.phrs.2021.105529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/25/2022]
Abstract
Despite recent advances in the treatment of colorectal cancer (CRC), low patient survival rate due to emergence of drug resistant cancer cells, metastasis and multiple deleterious side effects of chemotherapy, is a cause of public concern globally. To negate these clinical conundrums, search for effective and harmless novel molecular entities for the treatment of CRC is an urgent necessity. Since antimicrobial peptides (AMPs) are part of innate immunity of living beings, it is quite imperative to look for essential attributes of these peptides which may contribute to their effectiveness against carcinogenesis. Once identified, those characteristics can be suitably modified using several synthetic and computational techniques to further enhance their selectivity and pharmacokinetic profiles. Hence, this review analyses scientific reports describing the antiproliferative action of AMPs derived from several sources, particularly focusing on various colon cancer in vitro/in vivo investigations. On perusal of the literature, it appears that AMPs based therapeutics would definitely find special place in CRC therapy in future either alone or as an adjunct to chemotherapy provided some necessary alterations are made in their natural structures to make them more compatible with modern clinical practice. In this context, further in-depth research is warranted in adequate in vivo models.
Collapse
Affiliation(s)
- Sonia Chauhan
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Devinder K Dhawan
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Avneet Saini
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Simran Preet
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| |
Collapse
|
26
|
Abstract
Introduction: Cystic sellar salivary gland-like lesions (CSSLs) are exceedingly rare, with fewer than a dozen case reports. They contain amorphous colloid identical to Rathke cleft cyst contents, but the cyst wall additionally shows cohesive aggregates of benign salivary glands. We report three new examples. Materials and methods: Two cases were seen at University of Colorado Denver and one at Memorial Sloan Kettering (MSK). Molecular testing was attempted on two of three. Results: Case 1 is a 20-year-old female who presented with panhypopituitarism and was found to have a suprasellar mass that proved to be a CSSL. She received no postoperative adjuvant therapy, but recurrence of headaches and blurred vision 2 years later prompted return to medical attention. A much smaller local cyst recurrence was now accompanied by a thickened, bulbous infundibular stalk. Second resection yielded a gliotic infundibular stalk and amorphous mucin, but no residual salivary-like glands. She is without further recurrence on 6-year follow-up. Case 2 is a 29-year-old female with headache; while seen initially at a tertiary care center, diagnosis was only made after consultation at MSK. Case 3 is 68-year-old female who had originally presented with apoplexy to an outside hospital 7 years prior to surgery and diagnosis. Molecular testing was uninformative on case 1 and negative for mutations or fusions on case 3. Conclusion: Few pathologists or neuropathologists have encountered CSSLs in their practices; case 1 produced recurrence and significant infundibular stalk damage, and case 3 originally manifested apoplexy, features not previously reported.
Collapse
|
27
|
Wang D, Wei X, Kalvakolanu DV, Guo B, Zhang L. Perspectives on Oncolytic Salmonella in Cancer Immunotherapy-A Promising Strategy. Front Immunol 2021; 12:615930. [PMID: 33717106 PMCID: PMC7949470 DOI: 10.3389/fimmu.2021.615930] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Since the first reported spontaneous regression of tumors in patients with streptococcus infection, cancer biological therapy was born and it evolved into today's immunotherapy over the last century. Although the original strategy was unable to impart maximal therapeutic benefit at the beginning, it laid the foundations for the development of immune checkpoint blockade and CAR-T which are currently used for cancer treatment in the clinics. However, clinical applications have shown that current cancer immunotherapy can cause a series of adverse reactions and are captious for patients with preexisting autoimmune disorders. Salmonellae was first reported to exert antitumor effect in 1935. Until now, numerous studies have proved its potency as an antitumor agent in the near future. In this review, we summarize the currently available data on the antitumor effects of Salmonella, and discussed a possibility of integrating Salmonella into cancer immunotherapy to overcome current obstacles.
Collapse
Affiliation(s)
- Ding Wang
- Department of Pathophysiology and Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaodong Wei
- Department of Pathophysiology and Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dhan V. Kalvakolanu
- Department of Microbiology and Immunology and Greenebaum Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ling Zhang
- Department of Pathophysiology and Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
28
|
Gu B, Hakun MC. Challenges and Opportunities in NUT Carcinoma Research. Genes (Basel) 2021; 12:genes12020235. [PMID: 33562801 PMCID: PMC7915910 DOI: 10.3390/genes12020235] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
NUT carcinoma (NC) is a type of aggressive cancer driven by chromosome translocations. Fusion genes between a DNA-binding protein, such as bromodomain and extraterminal domain (BET) proteins, and the testis-specific protein NUTM1 generated by these translocations drive the formation of NC. NC can develop in very young children without significant accumulation of somatic mutations, presenting a relatively clean model to study the genetic etiology of oncogenesis. However, after 20 years of research, a few challenging questions still remain for understanding the mechanism and developing therapeutics for NC. In this short review, we first briefly summarize the current knowledge regarding the molecular mechanism and targeted therapy development of NC. We then raise three challenging questions: (1) What is the cell of origin of NC? (2) How does the germline analogous epigenetic reprogramming process driven by the BET-NUTM1 fusion proteins cause NC? and (3) How will BET-NUTM1 targeted therapies be developed? We propose that with the unprecedented technological advancements in genome editing, animal models, stem cell biology, organoids, and chemical biology, we have unique opportunities to address these challenges.
Collapse
Affiliation(s)
- Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University East Lansing, MI 48824, USA
- Department of Biomedical Engineering; Michigan State University East Lansing, MI 48824, USA;
- Correspondence:
| | - Maxwell C. Hakun
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University East Lansing, MI 48824, USA
- Department of Biomedical Engineering; Michigan State University East Lansing, MI 48824, USA;
| |
Collapse
|
29
|
Banda M, McKim KL, Myers MB, Inoue M, Parsons BL. Outgrowth of erlotinib-resistant subpopulations recapitulated in patient-derived lung tumor spheroids and organoids. PLoS One 2020; 15:e0238862. [PMID: 32898185 PMCID: PMC7478813 DOI: 10.1371/journal.pone.0238862] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
A model that recapitulates development of acquired therapeutic resistance is needed to improve oncology drug development and patient outcomes. To achieve this end, we established methods for the preparation and growth of spheroids from primary human lung adenocarcinomas, including methods to culture, passage, monitor growth, and evaluate changes in mutational profile over time. Primary lung tumor spheroids were cultured in Matrigel® with varying concentrations of erlotinib, a small molecule kinase inhibitor of epidermal growth factor receptor (EGFR) that is ineffective against KRAS mutant cells. Subtle changes in spheroid size and number were observed within the first two weeks of culture. Spheroids were cultured for up to 24 weeks, during which time interactions between different cell types, movement, and assembly into heterogeneous organoid structures were documented. Allele-specific competitive blocker PCR (ACB-PCR) was used to quantify low frequency BRAF V600E, KRAS G12D, KRAS G12V, and PIK3CA H1047R mutant subpopulations in tumor tissue residue (TR) samples and cultured spheroids. Mutant subpopulations, including multiple mutant subpopulations, were quite prevalent. Twelve examples of mutant enrichment were found in eight of the 14 tumors analyzed, based on the criteria that a statistically-significant increase in mutant fraction was observed relative to both the TR and the no-erlotinib control. Of the mutants quantified in erlotinib-treated cultures, PIK3CA H1047 mutant subpopulations increased most often (5/14 tumors), which is consistent with clinical observations. Thus, this ex vivo lung tumor spheroid model replicates the cellular and mutational tumor heterogeneity of human lung adenocarcinomas and can be used to assess the outgrowth of mutant subpopulations. Spheroid cultures with characterized mutant subpopulations could be used to investigate the efficacy of lung cancer combination therapies.
Collapse
Affiliation(s)
- Malathi Banda
- Division of Genetic and Molecular Toxicology, US Food & Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas, United States of America
| | - Karen L. McKim
- Division of Genetic and Molecular Toxicology, US Food & Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas, United States of America
| | - Meagan B. Myers
- Division of Genetic and Molecular Toxicology, US Food & Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas, United States of America
| | - Masahiro Inoue
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Barbara L. Parsons
- Division of Genetic and Molecular Toxicology, US Food & Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas, United States of America
| |
Collapse
|
30
|
Marinelli D, Mazzotta M, Pizzuti L, Krasniqi E, Gamucci T, Natoli C, Grassadonia A, Tinari N, Tomao S, Sperduti I, Sanguineti G, Botticelli A, Fabbri A, Botti C, Ciliberto G, Barba M, Vici P. Neoadjuvant Immune-Checkpoint Blockade in Triple-Negative Breast Cancer: Current Evidence and Literature-Based Meta-Analysis of Randomized Trials. Cancers (Basel) 2020; 12:cancers12092497. [PMID: 32899209 PMCID: PMC7565914 DOI: 10.3390/cancers12092497] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Breast cancer is a heterogeneous disease, which encompasses several subgroups of entities widely varying by clinical-pathological features. Triple negative breast cancer is characterized by a particularly aggressive biological behavior. The administration of chemotherapy has long represented the most efficacious weapon in combating triple negative breast cancer in both its initial and late phase of development. A pivot point has been recently reached throughout the approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer. Results from the registrative trial, IMpassion 130, have increasingly fueled the flourishing of studies of immune-checkpoint inhibitors in the early stage of triple negative breast cancer development. We critically interpret results from the most recent literature in light of relevant issues of methodological nature and also present a quantitative summary of data from the inherent trials. Future directions are also highlighted. Abstract Chemotherapy based on the sequential use of anthracyclines and taxanes has long represented the most efficacious approach in the management of early-stage, triple-negative breast cancer, whose aggressive behavior is widely renowned. This standard chemotherapy backbone was subsequently enriched by the use of carboplatin, based on its association with increased pathologic complete response and efficacy in the metastatic setting. Following the results from the IMpassion130 trial, the recent approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer increasingly fueled the flourishing of trials of immune-checkpoint inhibitors in the early setting. In this work, we review the most recent inherent literature in light of key methodological issues and provide a quantitative summary of the results from phase II–III randomized trials of immunotherapic agents combined with chemotherapy in the setting of interest. Hints regarding future directions are also discussed.
Collapse
Affiliation(s)
- Daniele Marinelli
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy;
| | - Marco Mazzotta
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Teresa Gamucci
- Medical Oncology, Sandro Pertini Hospital, 00157 Rome, Italy;
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Antonino Grassadonia
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, ‘Sapienza’ University of Rome, 00161 Rome, Italy;
| | - Isabella Sperduti
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | | | - Agnese Fabbri
- Medical Oncology Unit, Belcolle Hospital, 01100 Viterbo, Italy;
| | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| |
Collapse
|
31
|
Germline genomes have a dominant-heritable contribution to cancer immune evasion and immunotherapy response. QUANTITATIVE BIOLOGY 2020. [DOI: 10.1007/s40484-020-0212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
32
|
Klein-Scory S, Wahner I, Maslova M, Al-Sewaidi Y, Pohl M, Mika T, Ladigan S, Schroers R, Baraniskin A. Evolution of RAS Mutational Status in Liquid Biopsies During First-Line Chemotherapy for Metastatic Colorectal Cancer. Front Oncol 2020; 10:1115. [PMID: 32766143 PMCID: PMC7378792 DOI: 10.3389/fonc.2020.01115] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/04/2020] [Indexed: 12/31/2022] Open
Abstract
Treatment options for patients with metastatic colorectal cancer (mCRC) are limited. This particularly affects the largest group of patients with RAS mutations, who are considered ineligible for therapy with antiEGFR antibodies. In this liquid biopsy-based study, we performed the first in-depth analysis of the RAS mutational status in initially RAS-mutated patients during first-line therapy. RAS status of twelve patients with initially RAS-mutated mCRC was monitored longitudinally in 69 liquid biopsy samples. We focused on patients with stable disease (SD) or partial remission (PR) during first-line therapy (11 patients). Detection of fragmented RAS-mutated circulating cell-free tumor DNA (ctDNA) in plasma was performed by digital-droplet PCR (ddPCR) and BEAMing. Patients' total tumor masses were determined by measuring the tumor volumes using CT scan data. All patients with PR or SD at first follow-up showed a significant decrease of RAS mutational load. In ten patients (91%), the ctDNA-based RAS mutational status converted to wild-type in ddPCR and BEAMing. Remarkably, conversions were observed early after the first cycle of chemotherapy. Plasma concentration of ctDNA was controlled by determination of methylated WIF1-promotor ctDNA burden as a second tumor marker for mCRC. Persistent presence of methylated WIF1-promotor fragments confirmed the ongoing release of ctDNA during treatment. In patients with initially RAS-mutated mCRC, RAS mutations rapidly disappeared during first-line therapy in liquid biopsy, independent of type and intensity of chemotherapy and irrespective of anti-VEGF treatments. Following our results demonstrating conversion of RAS-mutational status, potential effectiveness of anti-EGFR antibodies in selected patients becomes an attractive hypothesis for future studies.
Collapse
Affiliation(s)
- Susanne Klein-Scory
- IMBL Medical Clinic, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Bochum, Germany
| | - Ingo Wahner
- IMBL Medical Clinic, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Bochum, Germany
| | - Marina Maslova
- Department of Radiology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Yosef Al-Sewaidi
- Department of Radiology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Michael Pohl
- Department of Medicine, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Thomas Mika
- Department of Medicine, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Swetlana Ladigan
- Department of Medicine, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| | - Alexander Baraniskin
- IMBL Medical Clinic, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Bochum, Germany
- Department of Medicine, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
33
|
Gilbert ES, Little MP, Preston DL, Stram DO. Issues in Interpreting Epidemiologic Studies of Populations Exposed to Low-Dose, High-Energy Photon Radiation. J Natl Cancer Inst Monogr 2020; 2020:176-187. [PMID: 32657345 PMCID: PMC7355296 DOI: 10.1093/jncimonographs/lgaa004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/02/2020] [Indexed: 01/19/2023] Open
Abstract
This article addresses issues relevant to interpreting findings from 26 epidemiologic studies of persons exposed to low-dose radiation. We review the extensive data from both epidemiologic studies of persons exposed at moderate or high doses and from radiobiology that together have firmly established radiation as carcinogenic. We then discuss the use of the linear relative risk model that has been used to describe data from both low- and moderate- or high-dose studies. We consider the effects of dose measurement errors; these can reduce statistical power and lead to underestimation of risks but are very unlikely to bring about a spurious dose response. We estimate statistical power for the low-dose studies under the assumption that true risks of radiation-related cancers are those expected from studies of Japanese atomic bomb survivors. Finally, we discuss the interpretation of confidence intervals and statistical tests and the applicability of the Bradford Hill principles for a causal relationship.
Collapse
Affiliation(s)
- Ethel S Gilbert
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mark P Little
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Daniel O Stram
- Department of Preventive Medicine, School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
34
|
Lin MT, Zheng G, Tseng LH, Zhang P, Ling H, Azad N, Gocke CD, Montgomery E, Eshleman JR. Multiclonal colorectal cancers with divergent histomorphological features and RAS mutations: one cancer or separate cancers? Hum Pathol 2020; 98:120-128. [PMID: 32171651 DOI: 10.1016/j.humpath.2020.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 02/05/2023]
Abstract
Detection of coexisting mutations within the same signal transduction pathway, which are expected to be mutually exclusive, raises a concern of laboratory errors. We have previously confirmed the presence of different RAS (KRAS and NRAS) mutations in the adenoma and/or adenocarcinoma subpopulations of colorectal cancers (CRCs). In this study, multiregional analyses by next-generation sequencing were conducted to elucidate the mechanisms underlying multiple RAS mutations seen in 5 CRC specimens. Multiregional analyses were initially conducted in a single tissue block originally submitted for mutational profiling. In 2 specimens, mutational status of the APC gene was not identical, indicating collisional adenoma and adenocarcinoma. In 3 specimens, the same APC mutation was present in different subpopulations with divergent RAS mutations, indicating a common clonal origin. Subsequent comprehensive multiregional analyses of additional adenoma and adenocarcinoma components revealed multiclonal CRCs with divergent histomorphological features and RAS mutations originating from a common APC-mutated founder lineage of adenoma, but from different RAS-mutated founder lineages of adenocarcinoma. These findings are consistent with the stepwise model of colorectal tumorigenesis along with parallel evolution, which affects RAS genes within the mitogen-activated protein kinase pathway and occurs during the progression from adenomas to adenocarcinomas. Evaluation of tumor subpopulations with divergent histomorphological features by pathologists may help identify multiclonal CRCs. Further studies are warranted to evaluate the incidence of multiclonality in CRCs and its impact on clinical outcomes. Perhaps, multiclonal CRCs originating from the same APC-mutated founder lineage of adenoma but from different RAS-mutated founder lineages of adenocarcinomas should be defined and managed as separate CRCs.
Collapse
Affiliation(s)
- Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Medical Genetics, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Peng Zhang
- Center for Inherited Disease Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hua Ling
- Center for Inherited Disease Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nilo Azad
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Christopher D Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Elizabeth Montgomery
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
35
|
Contreras HR, López-Moncada F, Castellón EA. Cancer stem cell and mesenchymal cell cooperative actions in metastasis progression and hormone resistance in prostate cancer: Potential role of androgen and gonadotropin‑releasing hormone receptors (Review). Int J Oncol 2020; 56:1075-1082. [PMID: 32319606 DOI: 10.3892/ijo.2020.5008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of male cancer‑associated mortality worldwide. Mortality is associated with metastasis and hormone resistance. Cellular, genetic and molecular mechanisms underlying metastatic progression and hormone resistance are poorly understood. Studies have investigated the local effects of gonadotropin‑releasing hormone (GnRH) analogs (used for androgen deprivation treatments) and the presence of the GnRH receptor (GnRH‑R) on PCa cells. Furthermore, cell subpopulations with stem‑like properties, or cancer stem cells, have been isolated and characterized using a cell culture system derived from explants of human prostate tumors. In addition, the development of preclinical orthotopic models of human PCa in a nonobese diabetic/severe combined immunodeficiency mouse model of compromised immunity has enabled the establishment of a reproducible system of metastatic progression in vivo. There is increasing evidence that metastasis is a complex process involving the cooperative actions of different cancer cell subpopulations, in which cancer stem‑like cells would be responsible for the final step of colonizing premetastatic niches. It has been hypothesized that PCa cells with stemness and mesenchymal signatures act cooperatively in metastatic progression and the inhibition of stemness genes, and that overexpression of androgen receptor (AR) and GnRH‑R decreases the rate the metastasis and sensitizes tumors to hormone therapy. The aim of the present review is to analyze the evidence regarding this cooperative process and the possible influence of stem‑like cell phenotypes, AR and GnRH‑R in metastatic progression and hormone resistance. These aspects may represent an important contribution in the understanding of the mechanisms underlying metastasis and hormone resistance in PCa, and potential routes to blocking these processes, enabling the development of novel therapies that would be particularly relevant for patients with metastatic and castration‑resistant PCa.
Collapse
Affiliation(s)
- Héctor R Contreras
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Fernanda López-Moncada
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Enrique A Castellón
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
36
|
Hegde PS, Chen DS. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020; 52:17-35. [PMID: 31940268 DOI: 10.1016/j.immuni.2019.12.011] [Citation(s) in RCA: 1175] [Impact Index Per Article: 293.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/01/2019] [Accepted: 12/14/2019] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a validated and critically important approach for treating patients with cancer. Given the vast research and clinical investigation efforts dedicated to advancing both endogenous and synthetic immunotherapy approaches, there is a need to focus on crucial questions and define roadblocks to the basic understanding and clinical progress. Here, we define ten key challenges facing cancer immunotherapy, which range from lack of confidence in translating pre-clinical findings to identifying optimal combinations of immune-based therapies for any given patient. Addressing these challenges will require the combined efforts of basic researchers and clinicians, and the focusing of resources to accelerate understanding of the complex interactions between cancer and the immune system and the development of improved treatment options for patients with cancer.
Collapse
|
37
|
Harris KL, Myers MB, McKim KL, Elespuru RK, Parsons BL. Rationale and Roadmap for Developing Panels of Hotspot Cancer Driver Gene Mutations as Biomarkers of Cancer Risk. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:152-175. [PMID: 31469467 PMCID: PMC6973253 DOI: 10.1002/em.22326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 05/24/2023]
Abstract
Cancer driver mutations (CDMs) are necessary and causal for carcinogenesis and have advantages as reporters of carcinogenic risk. However, little progress has been made toward developing measurements of CDMs as biomarkers for use in cancer risk assessment. Impediments for using a CDM-based metric to inform cancer risk include the complexity and stochastic nature of carcinogenesis, technical difficulty in quantifying low-frequency CDMs, and lack of established relationships between cancer driver mutant fractions and tumor incidence. Through literature review and database analyses, this review identifies the most promising targets to investigate as biomarkers of cancer risk. Mutational hotspots were discerned within the 20 most mutated genes across the 10 deadliest cancers. Forty genes were identified that encompass 108 mutational hotspot codons overrepresented in the COSMIC database; 424 different mutations within these hotspot codons account for approximately 63,000 tumors and their prevalence across tumor types is described. The review summarizes literature on the prevalence of CDMs in normal tissues and suggests such mutations are direct and indirect substrates for chemical carcinogenesis, which occurs in a spatially stochastic manner. Evidence that hotspot CDMs (hCDMs) frequently occur as tumor subpopulations is presented, indicating COSMIC data may underestimate mutation prevalence. Analyses of online databases show that genes containing hCDMs are enriched in functions related to intercellular communication. In its totality, the review provides a roadmap for the development of tissue-specific, CDM-based biomarkers of carcinogenic potential, comprised of batteries of hCDMs and can be measured by error-correct next-generation sequencing. Environ. Mol. Mutagen. 61:152-175, 2020. Published 2019. This article is a U.S. Government work and is in the public domain in the USA. Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Kelly L. Harris
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Meagan B. Myers
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Karen L. McKim
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Rosalie K. Elespuru
- Division of Biology, Chemistry and Materials ScienceCDRH/OSEL, US Food and Drug AdministrationSilver SpringMaryland
| | - Barbara L. Parsons
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| |
Collapse
|
38
|
Behnan J, Finocchiaro G, Hanna G. The landscape of the mesenchymal signature in brain tumours. Brain 2019; 142:847-866. [PMID: 30946477 PMCID: PMC6485274 DOI: 10.1093/brain/awz044] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
The complexity of glioblastoma multiforme, the most common and lethal variant of gliomas, is reflected by cellular and molecular heterogeneity at both the inter- and intra-tumoural levels. Molecular subtyping has arisen in the past two decades as a promising strategy to give better predictions of glioblastoma multiforme evolution, common disease pathways, and rational treatment options. The Cancer Genome Atlas network initially identified four molecular subtypes of glioblastoma multiforme: proneural, neural, mesenchymal and classical. However, further studies, also investigated glioma stem cells, have only identified two to three subtypes: proneural, mesenchymal and classical. The proneural-mesenchymal transition upon tumour recurrence has been suggested as a mechanism of tumour resistance to radiation and chemotherapy treatment. Glioblastoma multiforme patients with the mesenchymal subtype tend to survive shorter than other subtypes when analysis is restricted to samples with low transcriptional heterogeneity. Although the mesenchymal signature in malignant glioma may seem at odds with the common idea of the ectodermal origin of neural-glial lineages, the presence of the mesenchymal signature in glioma is supported by several studies suggesting that it can result from: (i) intrinsic expression of tumour cells affected with accumulated genetic mutations and cell of origin; (ii) tumour micro-environments with recruited macrophages or microglia, mesenchymal stem cells or pericytes, and other progenitors; (iii) resistance to tumour treatment, including radiotherapy, antiangiogenic therapy and possibly chemotherapy. Genetic abnormalities, mainly NF1 mutations, together with NF-κB transcriptional programs, are the main driver of acquiring mesenchymal-signature. This signature is far from being simply tissue artefacts, as it has been identified in single cell glioma, circulating tumour cells, and glioma stem cells that are released from the tumour micro-environment. All these together suggest that the mesenchymal signature in glioblastoma multiforme is induced and sustained via cell intrinsic mechanisms and tumour micro-environment factors. Although patients with the mesenchymal subtype tend to have poorer prognosis, they may have favourable response to immunotherapy and intensive radio- and chemotherapy.
Collapse
Affiliation(s)
- Jinan Behnan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.,Duke Preclinical Translational Unit, Duke University Medical Center, Durham, North Carolina
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Neurological Institute C. Besta, Milan, Italy
| | - Gabi Hanna
- Duke Preclinical Translational Unit, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
39
|
Shademan M, Naseri Salanghuch A, Zare K, Zahedi M, Foroughi MA, Akhavan Rezayat K, Mosannen Mozaffari H, Ghaffarzadegan K, Goshayeshi L, Dehghani H. Expression profile analysis of two antisense lncRNAs to improve prognosis prediction of colorectal adenocarcinoma. Cancer Cell Int 2019; 19:278. [PMID: 31708689 PMCID: PMC6836367 DOI: 10.1186/s12935-019-1000-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 01/14/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) are involved in different pathogenesis pathways including cancer pathogenesis. The adenoma-carcinoma pathway in colorectal cancer may involve the aberrant and variable gene expression of regulatory RNAs. This study was conducted to analyse the expression and prognosis prediction ability of two natural antisense transcripts, protein kinase C theta antisense RNA 1 (PRKCQ-AS1), and special AT-rich sequence binding protein 1 antisense RNA 1 (SATB1-AS1) in colorectal low-grade adenoma, advanced adenoma, and adenocarcinomas. Methods In this study, from two RNA-seq analyses of CCAT1-ko cells and colorectal carcinoma biopsies having diminished and increased levels of CCAT1 transcription, respectively, we nominated two antisense lncRNAs of PRKCQ-AS1 and SATB1-AS1. Samples from colorectal low-grade adenomas, advanced adenomas, adenocarcinomas, and adjacent tissue were subjected to RT-qPCR to determine the expression of PRKCQ-AS1, SATB1-AS1 along with colon cancer-associated transcript 1 (CCAT1) and cMYC. In addition, we used different bioinformatics analyses and webservers (including GEPIA 2, TCGA, and CancerMine) to elucidate the prognosis prediction value, the expression correlation of sense–antisense pair of genes, and the expression profile of these antisense transcripts at the presence or absence of mutations in the driver genes, or the corresponding sense genes. Results PRKCQ-AS1 showed a wide range of expression levels in colorectal adenoma, advanced adenoma, and adenocarcinoma. Upregulation of PRKCQ-AS1 was related to a significant decrease in survival of colorectal cancer (CRC) patients. The expression levels of PRKCQ-AS1 and PRKCQ were strong and significantly concordant in normal and cancerous colorectal tissues. While SATB1-AS1 showed a wide range of expression in colorectal adenoma, advanced adenoma, and adenocarcinoma as well, its expression was not related to a decrease in survival of CRC patients. The expression levels of SATB1-AS1 and SATB1 (the sense gene) were not strong in normal colorectal tissues. In addition, where SATB1 gene was mutated, the expression of SATB1-AS1 was significantly downregulated. Conclusions We found the expression of PRKCQ-AS1 and SATB1-AS1 at a given stage of CRC very variable, and not all biopsy samples showed the increased expression of these antisense transcripts. PRKCQ-AS1 in contrast to SATB1-AS1 showed a significant prognostic value. Since a significantly concordant expression was observed for SATB1-AS1 and SATB1 in only cancerous, and for PRKCQ-AS1 and PRKCQ in both normal and cancerous colorectal tissues, it can be concluded that common mechanisms may regulate the expression of these sense and antisense genes.
Collapse
Affiliation(s)
- Milad Shademan
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974 Iran
| | - Azam Naseri Salanghuch
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974 Iran
| | - Khadijeh Zare
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974 Iran
| | - Morteza Zahedi
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974 Iran
| | - Mohammad Ali Foroughi
- 2Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Kambiz Akhavan Rezayat
- 3Department of Gastroenterology and Hepatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,5Gastroenterology and Hepatology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hooman Mosannen Mozaffari
- 3Department of Gastroenterology and Hepatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,5Gastroenterology and Hepatology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kamran Ghaffarzadegan
- Pathology Department, Education and Research Department, Razavi Hospital, Mashhad, Iran
| | - Ladan Goshayeshi
- 3Department of Gastroenterology and Hepatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,4Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Dehghani
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974 Iran.,2Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,7Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
40
|
Martín-Pardillos A, Valls Chiva Á, Bande Vargas G, Hurtado Blanco P, Piñeiro Cid R, Guijarro PJ, Hümmer S, Bejar Serrano E, Rodriguez-Casanova A, Diaz-Lagares Á, Castellvi J, Miravet-Verde S, Serrano L, Lluch-Senar M, Sebastian V, Bribian A, López-Mascaraque L, López-López R, Ramón Y Cajal S. The role of clonal communication and heterogeneity in breast cancer. BMC Cancer 2019; 19:666. [PMID: 31277602 PMCID: PMC6612119 DOI: 10.1186/s12885-019-5883-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022] Open
Abstract
Background Cancer is a rapidly evolving, multifactorial disease that accumulates numerous genetic and epigenetic alterations. This results in molecular and phenotypic heterogeneity within the tumor, the complexity of which is further amplified through specific interactions between cancer cells. We aimed to dissect the molecular mechanisms underlying the cooperation between different clones. Methods We produced clonal cell lines derived from the MDA-MB-231 breast cancer cell line, using the UbC-StarTrack system, which allowed tracking of multiple clones by color: GFP C3, mKO E10 and Sapphire D7. Characterization of these clones was performed by growth rate, cell metabolic activity, wound healing, invasion assays and genetic and epigenetic arrays. Tumorigenicity was tested by orthotopic and intravenous injections. Clonal cooperation was evaluated by medium complementation, co-culture and co-injection assays. Results Characterization of these clones in vitro revealed clear genetic and epigenetic differences that affected growth rate, cell metabolic activity, morphology and cytokine expression among cell lines. In vivo, all clonal cell lines were able to form tumors; however, injection of an equal mix of the different clones led to tumors with very few mKO E10 cells. Additionally, the mKO E10 clonal cell line showed a significant inability to form lung metastases. These results confirm that even in stable cell lines heterogeneity is present. In vitro, the complementation of growth medium with medium or exosomes from parental or clonal cell lines increased the growth rate of the other clones. Complementation assays, co-growth and co-injection of mKO E10 and GFP C3 clonal cell lines increased the efficiency of invasion and migration. Conclusions These findings support a model where interplay between clones confers aggressiveness, and which may allow identification of the factors involved in cellular communication that could play a role in clonal cooperation and thus represent new targets for preventing tumor progression. Electronic supplementary material The online version of this article (10.1186/s12885-019-5883-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana Martín-Pardillos
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain. .,CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain.
| | - Ángeles Valls Chiva
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Gemma Bande Vargas
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Roberto Piñeiro Cid
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain.,Cancer Modelling Lab, Roche-CHUS Joint Unit, Santiago de Compostela, Spain
| | - Pedro J Guijarro
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Stefan Hümmer
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain.,CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain
| | - Eva Bejar Serrano
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain.,CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain
| | - Aitor Rodriguez-Casanova
- Cancer Epigenomics, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Ángel Diaz-Lagares
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain.,Cancer Epigenomics, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Josep Castellvi
- Hospital Vall d'Hebron, Anatomía Patológica, Barcelona, Spain
| | - Samuel Miravet-Verde
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Institute of Science and Technology, Barcelona, Spain
| | - Luis Serrano
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - María Lluch-Senar
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Institute of Science and Technology, Barcelona, Spain
| | - Víctor Sebastian
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Zaragoza, Spain.,Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | - Ana Bribian
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Laura López-Mascaraque
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Rafael López-López
- Cancer Epigenomics, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain.,Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Santiago Ramón Y Cajal
- Translational Molecular Pathology Group, Vall d'Hebron Research Institute, Barcelona, Spain. .,CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Madrid, Spain. .,Hospital Vall d'Hebron, Anatomía Patológica, Barcelona, Spain.
| |
Collapse
|
41
|
Bochtler T, Krämer A. Does Cancer of Unknown Primary (CUP) Truly Exist as a Distinct Cancer Entity? Front Oncol 2019; 9:402. [PMID: 31165045 PMCID: PMC6534107 DOI: 10.3389/fonc.2019.00402] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer of unknown primary (CUP) designates an enigmatic cancer entity with histologic confirmation of malignancy from a metastasis but no identifiable primary tumor in spite of a thorough diagnostic work-up. In this review, we discuss the validity of CUP as a distinct cancer entity as well as diagnostic pitfalls. As arguments against a distinct entity, the diagnosis of CUP is erroneous in some cases. Diagnostic pitfalls include incomplete diagnostics, uncertainty in classifying a lesion as either primary or metastasis and mistaking a relapse of an antecedent malignancy as CUP due to histologic and immunohistologic disparities. Given the high frequency of prior malignancies in CUP patients, relapse of an antecedent cancer should always be carefully excluded. Gene expression profiling-based classifier assays aim at aligning the molecular profile of CUP patients with established primary cancer patterns for highest congruency in order to identify the putative primary and treat accordingly. However, the spectrum of predicted putative primaries by molecular techniques is somewhat at odds with the primaries identified in autopsy series. Also, a first randomized clinical trial did not show superiority of primary-tailored therapy over unspecific platinum-based chemotherapy. CUP cases share an aggressive clinical course, atypical metastasis pattern, rapid progression of metastases, a generally poor response to chemotherapy and dismal outcome as distinct clinical features. Metastatic spread appears to take place in the early stages of tumor evolution, with CUP metastases subsequently undergoing genetic evolution toward a chromosomally highly complex and instable karyotype independent from the primary tumor. In clinical practice, the diagnosis of CUP is valid when no primary tumor is detectable. Treatment should ideally offer broad spectrum coverage across numerous malignancies and be well-established in CUP as is the case for carboplatin/paclitaxel and cisplatin / gemcitabine in particular, but it should also cover the most likely putative primary. The diligent diagnosis of CUP is warranted for clinical trials, making the eligibility process particularly laborious. In conclusion, we deem CUP a distinct cancer entity and the diagnosis accurate in most patient cases.
Collapse
Affiliation(s)
- Tilmann Bochtler
- Clinical Cooperation Unit Molecular Hematology/Oncology, Department of Internal Medicine V, German Cancer Research Center, University Hospital Heidelberg, Heidelberg, Germany.,Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, Department of Internal Medicine V, German Cancer Research Center, University Hospital Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
Myers MB, McKim KL, Banda M, George NI, Parsons BL. Low-Frequency Mutational Heterogeneity of Invasive Ductal Carcinoma Subtypes: Information to Direct Precision Oncology. Int J Mol Sci 2019; 20:E1011. [PMID: 30813596 PMCID: PMC6429455 DOI: 10.3390/ijms20051011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022] Open
Abstract
Information regarding the role of low-frequency hotspot cancer-driver mutations (CDMs) in breast carcinogenesis and therapeutic response is limited. Using the sensitive and quantitative Allele-specific Competitor Blocker PCR (ACB-PCR) approach, mutant fractions (MFs) of six CDMs (PIK3CA H1047R and E545K, KRAS G12D and G12V, HRAS G12D, and BRAF V600E) were quantified in invasive ductal carcinomas (IDCs; including ~20 samples per subtype). Measurable levels (i.e., ≥ 1 × 10-5, the lowest ACB-PCR standard employed) of the PIK3CA H1047R, PIK3CA E545K, KRAS G12D, KRAS G12V, HRAS G12D, and BRAF V600E mutations were observed in 34/81 (42%), 29/81 (36%), 51/81 (63%), 9/81 (11%), 70/81 (86%), and 48/81 (59%) of IDCs, respectively. Correlation analysis using available clinicopathological information revealed that PIK3CA H1047R and BRAF V600E MFs correlate positively with maximum tumor dimension. Analysis of IDC subtypes revealed minor mutant subpopulations of critical genes in the MAP kinase pathway (KRAS, HRAS, and BRAF) were prevalent across IDC subtypes. Few triple-negative breast cancers (TNBCs) had appreciable levels of PIK3CA mutation, suggesting that individuals with TNBC may be less responsive to inhibitors of the PI3K/AKT/mTOR pathway. These results suggest that low-frequency hotspot CDMs contribute significantly to the intertumoral and intratumoral genetic heterogeneity of IDCs, which has the potential to impact precision oncology approaches.
Collapse
Affiliation(s)
- Meagan B Myers
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Karen L McKim
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Malathi Banda
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Nysia I George
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Barbara L Parsons
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
43
|
Modern conception of carcinogenesis creates opportunities to advance cancer risk assessment. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|