1
|
Zhao Z, Xiong G, Wang C, Cao W. From pathogenesis to precision medicine: Transformative advances in research and treatment of ameloblastoma. Cancer Lett 2025; 612:217448. [PMID: 39800213 DOI: 10.1016/j.canlet.2025.217448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
Odontogenic neoplasms of the jaw are dominated by ameloblastoma (AM), a locally aggressive epithelial tumor with a significant propensity for recurrence. The World Health Organization's 2022 update to the AM classification system underscores recent progress in comprehending its underlying mechanisms and refining clinical approaches. Contemporary research has yielded significant insights into the genetic underpinnings of AM, paving the way for the development of precision-based treatment strategies. Advanced genetic profiling has revealed a significant frequency of BRAF (V-raf murine sarcoma viral oncogene homolog) V600E and SMO (Smoothened) gene alterations in AM. Importantly, therapeutic interventions specifically designed to target these genetic aberrations, including BRAF and MEK pathway blockers, have shown encouraging results in terms of both effectiveness and tolerability, as documented in individual case reports and small-scale clinical investigations. This comprehensive review summarizes the recent modifications to the World Health Organization's categorization of AMs, explores progress in elucidating their underlying molecular pathways, and evaluates emerging targeted treatment modalities. Our objective is to present a thorough synthesis of contemporary scientific discoveries and therapeutic interventions, potentially paving the way for more efficacious and individualized clinical management protocols for this complex neoplasm.
Collapse
Affiliation(s)
- Zhang Zhao
- Department of Oral and Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Gan Xiong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Cheng Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Wei Cao
- Department of Oral and Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|
2
|
Walker A, Fang CS, Schroff C, Serrano J, Vasudevaraja V, Yang Y, Belakhoua S, Faustin A, William CM, Zagzag D, Chiang S, Acosta AM, Movahed-Ezazi M, Park K, Moreira AL, Darvishian F, Galbraith K, Snuderl M. Deep learning-based classifier for carcinoma of unknown primary using methylation quantitative trait loci. J Neuropathol Exp Neurol 2025; 84:147-154. [PMID: 39607989 PMCID: PMC11747144 DOI: 10.1093/jnen/nlae123] [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] [Indexed: 11/30/2024] Open
Abstract
Cancer of unknown primary (CUP) constitutes between 2% and 5% of human malignancies and is among the most common causes of cancer death in the United States. Brain metastases are often the first clinical presentation of CUP; despite extensive pathological and imaging studies, 20%-45% of CUP are never assigned a primary site. DNA methylation array profiling is a reliable method for tumor classification but tumor-type-specific classifier development requires many reference samples. This is difficult to accomplish for CUP as many cases are never assigned a specific diagnosis. Recent studies identified subsets of methylation quantitative trait loci (mQTLs) unique to specific organs, which could help increase classifier accuracy while requiring fewer samples. We performed a retrospective genome-wide methylation analysis of 759 carcinoma samples from formalin-fixed paraffin-embedded tissue samples using Illumina EPIC array. Utilizing mQTL specific for breast, lung, ovarian/gynecologic, colon, kidney, or testis (BLOCKT) (185k total probes), we developed a deep learning-based methylation classifier that achieved 93.12% average accuracy and 93.04% average F1-score across a 10-fold validation for BLOCKT organs. Our findings indicate that our organ-based DNA methylation classifier can assist pathologists in identifying the site of origin, providing oncologists insight on a diagnosis to administer appropriate therapy, improving patient outcomes.
Collapse
Affiliation(s)
- Adam Walker
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Camila S Fang
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Chanel Schroff
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Jonathan Serrano
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Varshini Vasudevaraja
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Yiying Yang
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Sarra Belakhoua
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Arline Faustin
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Christopher M William
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - David Zagzag
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Sarah Chiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Misha Movahed-Ezazi
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Kyung Park
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Andre L Moreira
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Farbod Darvishian
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Kristyn Galbraith
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| |
Collapse
|
3
|
Ren C, D'Amato G, Hornicek FJ, Tao H, Duan Z. Advances in the molecular biology of the solitary fibrous tumor and potential impact on clinical applications. Cancer Metastasis Rev 2024; 43:1337-1352. [PMID: 39120790 PMCID: PMC11554739 DOI: 10.1007/s10555-024-10204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Solitary fibrous tumor (SFT) is a rare fibroblastic mesenchymal neoplasm. The current classification has merged SFT and hemangiopericytoma (HPC) into the same tumor entity, while the risk stratification models have been developed to compensate for clinical prediction. Typically, slow-growing and asymptomatic, SFT can occur in various anatomical sites, most commonly in the pleura. Histologically, SFT consists of spindle to oval cells with minimal patterned growth, surrounded by stromal collagen and unique vascular patterns. Molecularly, SFT is defined by the fusion of NGFI-A-binding protein 2 (NAB2) and signal transducer and activator of transcription 6 (STAT6) genes as NAB2-STAT6. This fusion transforms NAB2 into a transcriptional activator, activating early growth response 1 (EGR1) and contributing to SFT pathogenesis and development. There are several fusion variants of NAB2-STAT6 in tumor tissues, with the most frequent ones being NAB2ex4-STAT6ex2 and NAB2ex6-STAT6ex16/ex17. Diagnostic methods play a crucial role in SFT clinical practice and basic research, including RT-PCR, next-generation sequencing (NGS), FISH, immunohistochemistry (IHC), and Western blot analysis, each with distinct capabilities and limitations. Traditional treatment strategies of SFT encompass surgical resection, radiation therapy, and chemotherapy, while emerging management regimes include antiangiogenic agents, immunotherapy, RNA-targeting technologies, and potential targeted drugs. This review provides an update on SFT's clinical and molecular aspects, diagnostic methods, and potential therapies.
Collapse
Affiliation(s)
- Chongmin Ren
- Department of Bone Tumor, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
- The Orthopedic Hospital, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China
| | - Gina D'Amato
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
| | - Hao Tao
- The Orthopedic Hospital, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
4
|
Jawdekar R, Mishra V, Hatgoankar K, Tiwade YR, Bankar NJ. Precision medicine in cancer treatment: Revolutionizing care through proteomics, genomics, and personalized therapies. J Cancer Res Ther 2024; 20:1687-1693. [DOI: 10.4103/jcrt.jcrt_108_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/20/2024] [Indexed: 01/03/2025]
Abstract
ABSTRACT
Recent developments in biotechnology have allowed us to identify unique and complicated biological traits associated with cancer. Genomic profiling through next-generation sequencing (NGS) has revolutionized cancer therapy by evaluating hundreds of genes and biomarkers in a single assay. Proteomics offers blood-based biomarkers for cancer detection, categorization, and therapy monitoring. Immune oncology and chimeric antigen receptor (CAR-T cell) therapy use the immune system to combat cancer. Personalized cancer treatment is on the rise. Although precision medicine holds great promise, its widespread application faces obstacles such as lack of agreement on nomenclature, the difficulty of classifying patients into distinct groups, the difficulties of multimorbidity, magnitude, and the need for prompt intervention. This review studies advances in the era of precision medicine for cancer treatment; the application of genomic profiling techniques, NGS, proteomics, and targeted therapy; and the challenge in the application of precision medicine and the beneficial future it holds in cancer treatment.
Collapse
Affiliation(s)
- Riddhi Jawdekar
- Department of Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, Maharashtra, India
| | - Vaishnavi Mishra
- Department of Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Kajal Hatgoankar
- Department of Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, Maharashtra, India
| | - Yugeshwari R. Tiwade
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Nandkishor J. Bankar
- Department of Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| |
Collapse
|
5
|
Opoku KB, Santiago T, Kumar P, Roush SM, Fedoriw Y, Tomoka T, Leventaki V, Furtado LV, Bhakta N, Alexander TB, Wang JR. Transcriptome profiling of pediatric extracranial solid tumors and lymphomas enables rapid low-cost diagnostic classification. Sci Rep 2024; 14:19456. [PMID: 39169157 PMCID: PMC11339337 DOI: 10.1038/s41598-024-70541-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024] Open
Abstract
Approximately 80% of pediatric tumors occur in low- and middle-income countries (LMIC), where diagnostic tools essential for treatment decisions are often unavailable or incomplete. Development of cost-effective molecular diagnostics will help bridge the cancer diagnostic gap and ultimately improve pediatric cancer outcomes in LMIC settings. We investigated the feasibility of using nanopore whole transcriptome sequencing on formalin-fixed paraffin embedded (FFPE)-derived RNA and a composite machine learning model for pediatric solid tumor diagnosis. Transcriptome cDNA sequencing was performed on a heterogenous set of 221 FFPE and 32 fresh frozen pediatric solid tumor and lymphoma specimens on Oxford Nanopore Technologies' sequencing platforms. A composite machine learning model was then used to classify transcriptional profiles into clinically actionable tumor types and subtypes. In total, 95.6% and 89.7% of pediatric solid tumors and lymphoma specimens were correctly classified, respectively. 71.5% of pediatric solid tumors had prediction probabilities > 0.8 and were classified with 100% accuracy. Similarly, for lymphomas, 72.4% of samples that had prediction probabilities > 0.6 were classified with 97.6% accuracy. Additionally, FOXO1 fusion status was predicted accurately for 97.4% of rhabdomyosarcomas and MYCN amplification was predicted with 88% accuracy in neuroblastoma. Whole transcriptome sequencing from FFPE-derived pediatric solid tumor and lymphoma samples has the potential to provide clinical classification of both tissue lineage and core genomic classification. Further expansion, refinement, and validation of this approach is necessary to explore whether this technology could be part of the solution of addressing the diagnostic limitations in LMIC.
Collapse
Affiliation(s)
- Kofi B Opoku
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Teresa Santiago
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, USA
| | - Priya Kumar
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, USA
| | - Sophia M Roush
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Yuri Fedoriw
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Tamiwe Tomoka
- University of Malawi College of Medicine, Blantyre, Malawi
- University of North Carolina at Chapel Hill, Chapel Hill, USA
- UNC Project Malawi, Lilongwe, Malawi
| | - Vasiliki Leventaki
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Larissa V Furtado
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, USA
| | - Nickhill Bhakta
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, USA
| | - Thomas B Alexander
- Department of Pediatrics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Jeremy R Wang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, USA.
| |
Collapse
|
6
|
Tu T, Chen D, Jiang H, Ma J, Wang H, Chen C. The Prognosis of Advanced Non-Small Cell Lung Cancer Patients with Precision-Targeted Therapy Guided by NGS Testing or Routine Testing. Cancer Manag Res 2023; 15:1307-1318. [PMID: 38027245 PMCID: PMC10658950 DOI: 10.2147/cmar.s436808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose We aim to observe the potential survival benefits of driver gene-guided targeted therapy in advanced non-small cell lung cancer (NSCLC) patients compared to non-targeted therapy. Additionally, the study aims to assess whether Next-generation sequencing technology (NGS)-guided targeted therapy can provide survival advantages for advanced NSCLC patients compared to conventional Epidermal growth factor receptor (EGFR)/anaplastic lymphoma kinase (ALK) gene detection. Methods Clinical data, genetic testing results, and treatment information of 1663 advanced lung cancer patients diagnosed by pathology from January 2013 to June 2019 in Jiangsu University Affiliated Lianyungang Hospital were collected. Propensity score matching survival analysis was used to evaluate the differences in overall survival(OS) between groups. Results In the unadjusted survival curve, targeted therapy patients had significantly longer median OS than non-targeted therapy patients (28.3 months vs 15.4 months, Hazard ratio (HR) = 0.5426, 95% confidence interval (CI) 0.4768-0.6176, P < 0.0001); the conclusion was the same after propensity score matching analysis, with targeted therapy group patients having significantly prolonged OS median (27.5 months vs 14.8 months, HR = 0.5572, 95% CI 0.4796-0.6474, P < 0.0001). In the unadjusted survival curve, the NGS group had a significantly prolonged median OS compared to the conventional gene detection group (23.4 months vs 21.2 months, HR = 1.243, 95% CI = 1.017-1.519, P = 0.0495). However, after propensity score matching analysis, no statistically significant difference existed in the median OS between the two patient groups (23.1 months vs 21.5 months, HR = 1.288, 95% CI = 0.9557-1.735, P = 0.0926). Further analysis demonstrated no advantage in the five-, three-, and two-year survival rates of the NGS group compared to conventional gene detection group patients. However, the one-year survival rate of the NGS group was significantly increased (83.2% vs 68.1%, HR = 0.4890, 95% CI = 0.3170-0.7544, P = 0.0015). Conclusion Driver gene-guided targeted precision therapy significantly prolonged the median OS of advanced NSCLC patients compared to non-targeted therapy. NGS detection did not improve the median OS of advanced NSCLC patients compared to conventional EGFR/ALK gene detection but increased the one-year survival rate of patients.
Collapse
Affiliation(s)
- Tingting Tu
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| | - Dandan Chen
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| | - Houjun Jiang
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| | - Jianhua Ma
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| | - Hongwei Wang
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| | - Cheng Chen
- Department of Radiotherapy, Lianyungang Clinical Institute, Jiangsu University (The Second People’s Hospital of Lianyungang), Lianyungang, Jiangsu Province, People’s Republic of China
- Department of Radiotherapy, Lianyungang Cancer Hospital, Lianyungang, Jiangsu Province, People’s Republic of China
| |
Collapse
|
7
|
Wang Y, Mei J, Zhang Y, He X, Zheng X, Tan J, Jia Q, Li N, Li D, Wang Y, Meng Z. Cathepsin F genetic mutation is associated with familial papillary thyroid cancer. Am J Med Sci 2022; 364:414-424. [PMID: 35447134 DOI: 10.1016/j.amjms.2022.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 12/18/2021] [Accepted: 03/29/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Thyroid cancer is one of the most common cancers in the world. Genetic factors are important in the occurrence and development of thyroid cancer, and genetic diagnosis has become an important basis for the prognosis of benign and malignant nodules. We identify a family of six siblings with inherited thyroid cancer susceptibility. All six members of this generation have been definitely diagnosed with papillary thyroid carcinoma. This work aims at confirming the relevant causative genes for thyroid cancer in this pedigree. METHODS We extract DNA from the peripheral blood of six individuals and perform whole genome sequencing. Sanger sequencing and immunohistochemistry further testify the cathepsin F (CTSF) mutation and expression. RESULTS We identify 57 single nucleotide variations (SNVs) out of at least 4 affected family members via certain filter criteria. The CTSF gene found in five of the six family members is here considered the most promising candidate gene mutation for familial thyroid cancer. Besides, our research also proves several known genes including CTSB, TEKT4, ESR1, MSH6, DIRC3, GNAS, and BANCR that act as probable oncogenic drivers in this family. The Sanger sequencing identifies the existence and veracity of CTSF somatic mutations. The CTSF immunohistochemistry of thyroid cancer tissue specimens displays that higher CTSF expression in mutated patients than that in wild-type patient as well as pericarcinomatous tissue. CONCLUSIONS We conclude that the evaluation of CTSF gene mutations of patients in thyroid cancer families may be predictive and valuable for the familial heredity of thyroid cancer.
Collapse
Affiliation(s)
- Yaqiong Wang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Jingzhao Mei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P R China
| | - Yujie Zhang
- Department of Pathology, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, P R China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Qiang Jia
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, P R China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, P R China.
| | - Yan Wang
- Chinese Material Medical College, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, P R China; State Key Laboratory of Component-based Chinese Medicine, Jinghai District, Tianjin, P R China.
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, P R China.
| |
Collapse
|
8
|
Van der Mude A. A proposed Information-Based modality for the treatment of cancer. Biosystems 2021; 211:104587. [PMID: 34915101 DOI: 10.1016/j.biosystems.2021.104587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/20/2021] [Accepted: 12/01/2021] [Indexed: 11/02/2022]
Abstract
Treatment modalities for cancer involve physical manipulations such as surgery, immunology, radiation, chemotherapy or gene editing. This is a proposal for an information-based modality. This modality does not change the internal state of the cancer cell directly - instead, the cancer cell is manipulated by giving it information to instruct the cell to perform an action. This modality is based on a theory of Structure Encoding in DNA, where information about body part structure controls the epigenetic state of cells in the process of development from pluripotent cells to fully differentiated cells. It has been noted that cancer is often due to errors in morphogenetic differentiation accompanied by associated epigenetic processes. This implies a model of cancer called the Epigenetic Differentiation Model. A major feature of the Structure Encoding Theory is that the characteristics of the differentiated cell are affected by inter-cellular information passed in the tissue microenvironment, which specifies the exact location of a cell in a body part structure. This is done by exosomes that carry fragments of long non-coding RNA and transposons, which convey structure information. In the normal process of epigenetic differentiation, the information passed may lead to apoptosis due to the constraints of a particular body part structure. The proposed treatment involves determining what structure information is being passed in a particular tumor, then adding artificial exosomes that overwhelm the current information with commands for the cells to go into apoptosis.
Collapse
|
9
|
Zhou X, Zeng B, Li Y, Wang H, Zhang X. LINC02532 Contributes to Radiosensitivity in Clear Cell Renal Cell Carcinoma through the miR-654-5p/YY1 Axis. Molecules 2021; 26:molecules26227040. [PMID: 34834139 PMCID: PMC8625588 DOI: 10.3390/molecules26227040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Studies have shown that long non-coding RNAs (lncRNAs) play essential roles in tumor progression and can affect the response to radiotherapy, including in clear cell renal cell carcinoma (ccRCC). LINC02532 has been found to be upregulated in ccRCC. However, not much is known about this lncRNA. Hence, this study aimed to investigate the role of LINC02532 in ccRCC, especially in terms of radioresistance. Methods: Quantitative real-time PCR was used to detect the expression of LINC02532, miR-654-5p, and YY1 in ccRCC cells. Protein levels of YY1, cleaved PARP, and cleaved-Caspase-3 were detected by Western blotting. Cell survival fractions, viability, and apoptosis were determined by clonogenic survival assays, CCK-8 assays, and flow cytometry, respectively. The interplay among LINC02532, miR-654-5p, and YY1 was detected by chromatin immunoprecipitation and dual-luciferase reporter assays. In addition, in vivo xenograft models were established to investigate the effect of LINC02532 on ccRCC radioresistance in 10 nude mice. Results: LINC02532 was highly expressed in ccRCC cells and was upregulated in the cells after irradiation. Moreover, LINC02532 knockdown enhanced cell radiosensitivity both in vitro and in vivo. Furthermore, YY1 activated LINC02532 in ccRCC cells, and LINC02532 acted as a competing endogenous RNA that sponged miR-654-5p to regulate YY1 expression. Rescue experiments indicated that miR-654-5p overexpression or YY1 inhibition recovered ccRCC cell functions that had been previously impaired by LINC02532 overexpression. Conclusions: Our results revealed a positive feedback loop of LINC02532/miR-654-5p/YY1 in regulating the radiosensitivity of ccRCC, suggesting that LINC02532 might be a potential target for ccRCC radiotherapy. This study could serve as a foundation for further research on the role of LINC02532 in ccRCC and other cancers.
Collapse
Affiliation(s)
- Xiaoguang Zhou
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.Z.); (B.Z.); (Y.L.); (H.W.)
| | - Bowen Zeng
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.Z.); (B.Z.); (Y.L.); (H.W.)
- Department of Urology, Affiliated Hospital of Sergeant School of Army Medical University, Shijiazhuang 050044, China
| | - Yansheng Li
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.Z.); (B.Z.); (Y.L.); (H.W.)
| | - Haozhou Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.Z.); (B.Z.); (Y.L.); (H.W.)
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.Z.); (B.Z.); (Y.L.); (H.W.)
- Correspondence: ; Tel.: +86-010-85231247
| |
Collapse
|
10
|
Xiao D, Deng Q, He D, Huang Y, Liang W, Wang F, Yang H. High Tumor Mutation Burden and DNA Repair Gene Mutations are Associated with Primary Resistance to Crizotinib in ALK-Rearranged Lung Cancer. Onco Targets Ther 2021; 14:4809-4817. [PMID: 34552337 PMCID: PMC8450189 DOI: 10.2147/ott.s325443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Background About 20% of patients with ALK-rearranged non-small cell lung cancer (NSCLC) develop acquired resistance to tyrosine kinase inhibitor (TKI) during the first 6 months. This study aimed to examine the molecular mechanisms of early TKI resistance and prognosis in ALK-rearranged NSCLC. Methods Ten patients with ALK-rearranged NSCLC were included: five who developed rapid resistance to crizotinib (progression-free survival (PFS) ≤3 months) and five who exhibited a good response to crizotinib (PFS ≥36 months). The tumor specimens were subjected to whole-exome sequencing (WES). The validation cohort included 19 patients with ALK-rearranged NSCLC who received crizotinib; targeted sequencing of 43 selected genes was performed. The effect of the TP53 G245S mutation on crizotinib sensitivity was tested in H3122 cells. Results Mutations in DNA repair-associated genes were identified in primary resistance to crizotinib. Patients with a poor response to crizotinib harbored a greater burden of somatic mutations than those with a good response [median somatic mutations, 136 (range, 72-180) vs 31 (range, 10-48)]. Compared with the patients carrying wild-type TP53 or TP53 exon 3 deletion, 29 patients with TP53 G245S mutation showed a shorter survival time (P < 0.05), with a median PFS of 3 (95% CI: 1.9-4.1) months and a median overall survival of 7 (95% CI: 3.4-10.5) months. TP53 mutation promoted the proliferation of EML4-ALK-rearranged H3122 cells by approximately 3 folds (P < 0.001). H3122 cells with TP53 mutant were more sensitive to crizotinib compared with control cells. Conclusion A higher mutation burden and mutations in DNA repair gene, including TP53, were potentially associated with primary resistance to crizotinib in ALK-rearranged NSCLC. An immune-checkpoint inhibition strategy could be examined, which might overcome primary resistance to crizotinib in ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- Dakai Xiao
- Research Center forTranslational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Qiuhua Deng
- Research Center forTranslational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Dongyun He
- Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Ying Huang
- Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Wenchi Liang
- Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Fengnan Wang
- Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Haihong Yang
- Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| |
Collapse
|
11
|
Jia X, Guan R, Cui X, Zhu J, Liu P, Zhang L, Wang D, Zhang Y, Dong K, Wu J, Ji W, Ji G, Bai J, Yu J, Yu Y, Sun W, Zhang F, Fu S. Molecular structure and evolution mechanism of two populations of double minutes in human colorectal cancer cells. J Cell Mol Med 2020; 24:14205-14216. [PMID: 33124133 PMCID: PMC7754069 DOI: 10.1111/jcmm.16035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/26/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022] Open
Abstract
Gene amplification chiefly manifests as homogeneously stained regions (HSRs) or double minutes (DMs) in cytogenetically and extrachromosomal DNA (ecDNA) in molecular genetics. Evidence suggests that gene amplification is becoming a hotspot for cancer research, which may be a new treatment strategy for cancer. DMs usually carry oncogenes or chemoresistant genes that are associated with cancer progression, occurrence and prognosis. Defining the molecular structure of DMs will facilitate understanding of the molecular mechanism of tumorigenesis. In this study, we re‐identified the origin and integral sequence of DMs in human colorectal adenocarcinoma cell line NCI‐H716 by genetic mapping and sequencing strategy, employing high‐resolution array‐based comparative genomic hybridization, high‐throughput sequencing, multiplex‐fluorescence in situ hybridization and chromosome walking techniques. We identified two distinct populations of DMs in NCI‐H716, confirming their heterogeneity in cancer cells, and managed to construct their molecular structure, which were not investigated before. Research evidence of amplicons distribution in two different populations of DMs suggested that a multi‐step evolutionary model could fit the module of DM genesis better in NCI‐H716 cell line. In conclusion, our data implicated that DMs play a very important role in cancer progression and further investigation is necessary to uncover the role of the DMs.
Collapse
Affiliation(s)
- Xueyuan Jia
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Rongwei Guan
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Xiaobo Cui
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jing Zhu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Ling Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Dong Wang
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Kexian Dong
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Wei Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Guohua Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jing Bai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jingcui Yu
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Yu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Sheeja K, Lakshmi S. Nod-like receptor protein 3 inflammasome in head-and-neck cancer. J Cancer Res Ther 2020; 16:405-409. [PMID: 32719244 DOI: 10.4103/jcrt.jcrt_849_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Activation of inflammasomes has a decisive role in host defense mechanism against pathogens and other intracellular risk factors, but recently, it has been revealed that they play a significant role in the pathogenesis of several diseases, including cancer. Nod-like receptor protein 3 (NLRP3) inflammasome, the best-studied inflammasome, has contrasting roles in cancer development and progressions. In head-and-neck cancers, the upregulated level of NLRP3 promotes tumor progression. The main objective of this review is to provide current knowledge on the involvement of NLRP3 inflammasome in head-and-neck cancers. Deeper understanding of the biology of this dynamic protein complex provides new scope for the development of more effective anticancer therapies.
Collapse
Affiliation(s)
- K Sheeja
- Division of Cancer Research, Regional Cancer Centre, Laboratory of Molecular Medicine, Medical College, Thiruvananthapuram, Kerala, India
| | - S Lakshmi
- Division of Cancer Research, Regional Cancer Centre, Laboratory of Molecular Medicine, Medical College, Thiruvananthapuram, Kerala, India
| |
Collapse
|
13
|
Raheem F, Kim P, Grove M, Kiel PJ. Precision Genomic Practice in Oncology: Pharmacist Role and Experience in an Ambulatory Care Clinic. PHARMACY 2020; 8:pharmacy8010032. [PMID: 32182657 PMCID: PMC7151676 DOI: 10.3390/pharmacy8010032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022] Open
Abstract
Recent advancements in molecular testing, the availability of cost-effective technology, and novel approaches to clinical trial design have facilitated the implementation of tumor genome sequencing into standard of care oncology practices. Current models of precision oncology practice include specialized clinics or consultation services based on a molecular tumor board (MTB) approach. MTBs are comprised of interprofessional teams of clinicians and scientists who evaluate tumors at the molecular level to guide patient-specific targeted therapy. The practice of precision oncology utilizing MTB-based models is an emerging approach, transforming precision genomics from a novel concept into clinical practice. This rapid shift in practice from cytotoxic therapy to targeted medicine poses challenges, yet brings exciting opportunities to clinical pharmacists practicing in hematology and oncology. Only a few precision genomics programs in the United States have a strong pharmacy presence with oncology pharmacists serving in leadership roles in research, interpreting genomic sequencing, making treatment recommendations, and facilitating off-label drug procurement. This article describes the experience of the precision medicine clinic at the Indiana University Health Simon Cancer Center, with emphasis on the role of the pharmacist in the precision oncology initiative.
Collapse
|
14
|
Zhao J, Dean DC, Hornicek FJ, Yu X, Duan Z. Emerging next-generation sequencing-based discoveries for targeted osteosarcoma therapy. Cancer Lett 2020; 474:158-167. [PMID: 31987920 DOI: 10.1016/j.canlet.2020.01.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/28/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy and is frequently lethal via metastasis to the lung. While surgical techniques and adjuvant chemotherapies have emerged to combat this deadly cancer, the 5-year survival rate has plateaued over the past four decades. Therapeutic progress has been notably poor because past technologies have not been able to reveal obscured OS biomarkers and targets. With the advent and implementation of large-scale next-generation sequencing (NGS) studies, various somatic mutations and copy number changes involved in OS progression and metastasis have surfaced. These findings have significantly expanded the amount of genome-informed pathways and candidate genes suitable for targeting in pre-clinical models. Furthermore, NGS analyses comparing primary and matched pulmonary metastatic tumor tissues have catalogued previously unknown prognostic biomarkers in OS. In this review, we delineate the most recent findings in NGS for OS therapy and how this technology has advanced personalized therapy.
Collapse
Affiliation(s)
- Jie Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China; Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA; Department of Orthopaedic Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, Shandong, 250031, China.
| | - Dylan C Dean
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| | - Francis J Hornicek
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| | - Xiuchun Yu
- Department of Orthopaedic Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, Shandong, 250031, China.
| | - Zhenfeng Duan
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
15
|
Udar N, Iyer A, Porter M, Haigis R, Smith S, Dhillon S, Meier K, Ward D, Lu J, Wenz P, Buchner L, Dunn T, Wise A, Mueller A, Gutekunst K. Development and Analytical Validation of a DNA Dual-Strand Approach for the US Food and Drug Administration-Approved Next-Generation Sequencing-Based Praxis Extended RAS Panel for Metastatic Colorectal Cancer Samples. J Mol Diagn 2019; 22:159-178. [PMID: 31837434 DOI: 10.1016/j.jmoldx.2019.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/03/2019] [Accepted: 09/27/2019] [Indexed: 12/28/2022] Open
Abstract
A next-generation sequencing method was developed that can distinguish single-stranded modifications from low-frequency somatic mutations present on both strands of DNA in formalin-fixed paraffin-embedded colorectal cancer samples. We applied this method for analytical validation of the Praxis Extended RAS Panel, a US Food and Drug Administration-approved companion diagnostic for panitumumab, on the Illumina MiSeqDx platform. With the use of the TruSeq amplicon workflow, both strands of DNA from the starting material were interrogated independently. Mutations were reported only if found on both strands; artifacts usually present on only one strand would not be reported. A total of 56 mutations were targeted within the KRAS and NRAS genes. A minimum read depth of 1800× per amplicon is required per sample but averaged >30,000× at maximum multiplexing levels. Analytical validation studies were performed to determine the simultaneous detection of mutations on both strands, reproducibility, assay detection level, precision of the assay across various factors, and the impact of interfering substances. In conclusion, this assay can clearly distinguish single-stranded artifacts from low-frequency mutations. Furthermore, the assay is accurate, precise, and reproducible, can achieve consistent detection of a mutation at 5% mutation frequency, exhibits minimal impact from tested interfering substances, and can simultaneously detect 56 mutations in a single run using 10 samples plus controls.
Collapse
Affiliation(s)
- Nitin Udar
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Anita Iyer
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California.
| | - Margaret Porter
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Robert Haigis
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Shannon Smith
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Shivani Dhillon
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Kristen Meier
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Diane Ward
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Jing Lu
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Paul Wenz
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Leonard Buchner
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Tamsen Dunn
- Department of Bioinformatics, Illumina, Inc., San Diego, California
| | - Aaron Wise
- Department of Bioinformatics, Illumina, Inc., San Diego, California
| | - Amy Mueller
- Department of Medical Affairs, Illumina, Inc., San Diego, California
| | - Karen Gutekunst
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| |
Collapse
|
16
|
Family history of cancer and risk of paediatric and young adult's testicular cancer: A Norwegian cohort study. Br J Cancer 2019; 120:1007-1014. [PMID: 30967648 PMCID: PMC6734662 DOI: 10.1038/s41416-019-0445-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/13/2019] [Accepted: 03/13/2019] [Indexed: 12/17/2022] Open
Abstract
Background The aim of this study was to examine the association of a family history of cancer with the risk of testicular cancer in young adults. Methods This is a prospective cohort study including 1,974,287 males born 1951–2015, of whom 2686 were diagnosed with TC before the age of 30. Results A history of TC in male relatives was significantly associated with a diagnosis of TC among children and young adults, including brothers (6.3-fold), sons (4.7-fold), fathers (4.4-fold), paternal uncles (2.0-fold) and maternal uncles (1.9-fold). Individuals with a father diagnosed with a carcinoma or sarcoma showed an elevated risk (1.1-fold and 1.8-fold, respectively). A family history of mesothelioma was positively associated with a risk of TC [(father (2.8-fold), mother (4.6-fold) and maternal uncles and aunt (4.4-fold)]. Elevated risks were also observed when siblings were diagnosed with malignant melanoma (1.4-fold). The risk of TC was also increased when fathers (11.1-fold), paternal (4.9-fold) and maternal uncles and aunts (4.6-fold) were diagnosed with malignant neuroepithelial-tumours. Conclusion We found an increased risk of TC among children and young adults with a family history of TC, carcinoma, mesothelioma, sarcoma, malignant melanoma and malignant neuroepithelial tumours. Hereditary cancer syndromes might underlie some of the associations reported in this study.
Collapse
|
17
|
Galata C, Wimmer E, Kasper B, Wenz F, Reißfelder C, Jakob J. Multidisciplinary Tumor Board Recommendations for Oligometastatic Malignancies: A Prospective Single-Center Analysis. Oncol Res Treat 2019; 42:87-94. [PMID: 30814474 DOI: 10.1159/000495474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/15/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND The treatment of oligometastatic disease is challenging and few data exist to guide treatment decisions. The objective of this study was to improve the data on the prevalence and treatment of patients with oligometastatic disease. METHODS We conducted a prospective single-center analysis that included all consecutive patients discussed in multidisciplinary tumor boards (MDT) between February and July 2017. Patients with oligometastatic disease were identified and treatment strategies were evaluated. RESULTS 1,673 patients were included in this study, 609 (36.4%) presented with metastatic disease, 151 (9%) had oligometastatic disease. Common metastatic sites were brain, liver, and lung. Lung cancer patients were the largest cohort (20.5%) among all patients with oligometastatic disease compared with other tumor entities. The majority of oligometastatic patients (68.9%) received local treatment with or without additional chemotherapy, 17.9% were recommended systemic therapy alone. MDT recommended watchful waiting for 4.6% of the patients. CONCLUSION Patients with oligometastatic disease represent a considerable proportion of all patients in MDT. In this study, 68.9% of patients with oligometastatic disease received regional treatment. This shows a possible treatment shift from palliative to potentially curative intent. These data may be used to design prospective clinical trials to optimize the treatment of oligometastatic disease.
Collapse
|
18
|
Du H, Pan B, Chen T. Evaluation of chemical mutagenicity using next generation sequencing: A review. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2017; 35:140-158. [PMID: 28506110 DOI: 10.1080/10590501.2017.1328831] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Mutations are heritable changes in the nucleotide sequence of DNA that can lead to many adverse effects. Genotoxicity assays have been used to identify chemical mutagenicity. Recently, next generation sequencing (NGS) has been used for this purpose. In this review, we present the progress in NGS application for assessing mutagenicity of chemicals, including the methods used for detecting the induced mutations, bioinformatics tools for analyzing the sequencing data, and chemicals whose mutagenicity has been evaluated using NGS. Available information suggests that NGS technology has unparalleled advantages for evaluating mutagenicity of chemicals can be applied for the next generation of mutagenicity tests.
Collapse
Affiliation(s)
- Hua Du
- a Division of Genetic and Molecular Toxicology, National Center for Toxicological Research , U.S. Food and Drug Administration , Jefferson , Arkansas , USA
| | - Bohu Pan
- a Division of Genetic and Molecular Toxicology, National Center for Toxicological Research , U.S. Food and Drug Administration , Jefferson , Arkansas , USA
| | - Tao Chen
- a Division of Genetic and Molecular Toxicology, National Center for Toxicological Research , U.S. Food and Drug Administration , Jefferson , Arkansas , USA
| |
Collapse
|
19
|
Rahme K, Guo J, Holmes JD, O'Driscoll CM. Evaluation of the physicochemical properties and the biocompatibility of polyethylene glycol-conjugated gold nanoparticles: A formulation strategy for siRNA delivery. Colloids Surf B Biointerfaces 2015; 135:604-612. [PMID: 26322474 DOI: 10.1016/j.colsurfb.2015.08.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The potential of RNA interference (RNAi)-based therapeutics for cancer has received much attention; however, delivery of RNAi effectors, such as small interfering RNA (siRNA), remains an obstacle to clinical translation. Non-viral delivery vectors have been used extensively to enhance siRNA delivery. Recently, the potential of gold nanoparticles (AuNPs) for transporting drugs, proteins and genetic materials has been demonstrated. Previously, our laboratory synthesised positively charged, surfactant-free AuNPs in water by the reduction of gold (III) chloride (AuCl3) using hydroxylamine hydrochloride (NH2OH·HCl) in the presence of L-cysteine methyl ester hydrochloride (HSCH2CH(NH2)COOCH3·HCl) as a capping agent. These AuNPs, which achieve higher cell viability in comparison to cetyl trimethyl ammonium bromide (CTAB, a surfactant)-capped counterparts, have demonstrated potential for siRNA delivery. However, it is well known that systemic administration of cationic delivery systems without biological stablising moieties causes non-specific binding with negatively charged serum proteins, resulting in particle aggregation and opsonisation. Consequently, highly stable AuNPs capped with l-cysteine methyl ester hydrochloride conjugated to poly(ethylene glycol) (PEG) were synthesised in this study. PEGylation enhanced the biocompatibility of the AuNPs by reducing toxicity in a range of cell types, by inhibiting interaction with serum proteins thus avoiding aggregation, and, by providing protection against degradation by nucleases. Moreover, these PEGylated AuNPs formed nanoparticles (NPs) with siRNA (which was first compacted with protamine), and had a diameter within the nanoscale range (∼ 250 nm) and a near neutral surface charge (∼ 10 mV). In the future a bifunctional PEG chain on the AuNPs (i.e., SH-PEG-NH2, SH-PEG-COOH) will be used to facilitate conjugation of a targeting ligand to enhance cell specific uptake.
Collapse
Affiliation(s)
- Kamil Rahme
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland; Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Lebanon
| | - Jianfeng Guo
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Ireland
| | - Justin D Holmes
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
20
|
Bernstein C, Bernstein H. Epigenetic reduction of DNA repair in progression to gastrointestinal cancer. World J Gastrointest Oncol 2015; 7:30-46. [PMID: 25987950 PMCID: PMC4434036 DOI: 10.4251/wjgo.v7.i5.30] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 03/18/2015] [Accepted: 04/20/2015] [Indexed: 02/05/2023] Open
Abstract
Deficiencies in DNA repair due to inherited germ-line mutations in DNA repair genes cause increased risk of gastrointestinal (GI) cancer. In sporadic GI cancers, mutations in DNA repair genes are relatively rare. However, epigenetic alterations that reduce expression of DNA repair genes are frequent in sporadic GI cancers. These epigenetic reductions are also found in field defects that give rise to cancers. Reduced DNA repair likely allows excessive DNA damages to accumulate in somatic cells. Then either inaccurate translesion synthesis past the un-repaired DNA damages or error-prone DNA repair can cause mutations. Erroneous DNA repair can also cause epigenetic alterations (i.e., epimutations, transmitted through multiple replication cycles). Some of these mutations and epimutations may cause progression to cancer. Thus, deficient or absent DNA repair is likely an important underlying cause of cancer. Whole genome sequencing of GI cancers show that between thousands to hundreds of thousands of mutations occur in these cancers. Epimutations that reduce DNA repair gene expression and occur early in progression to GI cancers are a likely source of this high genomic instability. Cancer cells deficient in DNA repair are more vulnerable than normal cells to inactivation by DNA damaging agents. Thus, some of the most clinically effective chemotherapeutic agents in cancer treatment are DNA damaging agents, and their effectiveness often depends on deficient DNA repair in cancer cells. Recently, at least 18 DNA repair proteins, each active in one of six DNA repair pathways, were found to be subject to epigenetic reduction of expression in GI cancers. Different DNA repair pathways repair different types of DNA damage. Evaluation of which DNA repair pathway(s) are deficient in particular types of GI cancer and/or particular patients may prove useful in guiding choice of therapeutic agents in cancer therapy.
Collapse
|
21
|
Clinical massively parallel next-generation sequencing analysis of 409 cancer-related genes for mutations and copy number variations in solid tumours. Br J Cancer 2014; 111:2014-23. [PMID: 25314059 PMCID: PMC4229640 DOI: 10.1038/bjc.2014.518] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 02/06/2023] Open
Abstract
Background: In a clinical diagnostic laboratory, we evaluated the applicability of the Ion Proton sequencer for screening 409 cancer-related genes in solid tumours. Methods: DNA was extracted from formalin-fixed, paraffin-embedded (FFPE) tissue biopsy specimens of 55 solid tumours (20 with matched normal tissue) and four cell lines and screened for mutations in 409 genes using the Ion Proton system. The mutation profiles of these samples were known based on prior testing using the Ion Torrent Personal Genome Machine (46-gene hotspot panel), Sanger sequencing, or fluorescence in situ hybridisation (FISH). Concordance with retrospective findings and additional mutations were evaluated. Assay sensitivity and reproducibility were established. Gene copy number variations (CNVs) detected were confirmed by molecular inversion probe (MIP) array. Results: The average Ion Proton (409-gene panel) sequencing output per run was 8 gigabases with 128 million sequencing reads. Of the 15,992 amplicons in the 409-gene panel, 90% achieved a minimum average sequencing depth of 100X. In 59 samples, the Ion Proton detected 100 of 105 expected single-nucleotide variants (SNVs) and all expected deletions (n=8), insertions (n=5), and CNVs (n=7). Five SNVs were not detected due to failed amplification of targeted regions. In 20 tumours with paired normal tissue, Ion Proton detected 37 additional somatic mutations, several in genes of high prognostic or therapeutic significance, such as MET, ALK, TP53, APC, and PTEN. MIP array analysis confirmed all CNVs detected by Ion Proton. Conclusions: The Ion Proton (409-gene panel) system was found to be well suited for use in a clinical molecular diagnostic laboratory. It can simultaneously screen 409 genes for a variety of sequence variants in multiple samples using a low input of FFPE DNA with high reproducibility and sensitivity.
Collapse
|
22
|
Haas BJ, Papanicolaou A, Yassour M, Grabherr M, Blood PD, Bowden J, Couger MB, Eccles D, Li B, Lieber M, MacManes MD, Ott M, Orvis J, Pochet N, Strozzi F, Weeks N, Westerman R, William T, Dewey CN, Henschel R, LeDuc RD, Friedman N, Regev A. De novo transcript sequence reconstruction from RNA-seq using the Trinity platform for reference generation and analysis. Nat Protoc 2013; 8:1494-512. [PMID: 23845962 PMCID: PMC3875132 DOI: 10.1038/nprot.2013.084] [Citation(s) in RCA: 5557] [Impact Index Per Article: 463.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
De novo assembly of RNA-seq data enables researchers to study transcriptomes without the need for a genome sequence; this approach can be usefully applied, for instance, in research on 'non-model organisms' of ecological and evolutionary importance, cancer samples or the microbiome. In this protocol we describe the use of the Trinity platform for de novo transcriptome assembly from RNA-seq data in non-model organisms. We also present Trinity-supported companion utilities for downstream applications, including RSEM for transcript abundance estimation, R/Bioconductor packages for identifying differentially expressed transcripts across samples and approaches to identify protein-coding genes. In the procedure, we provide a workflow for genome-independent transcriptome analysis leveraging the Trinity platform. The software, documentation and demonstrations are freely available from http://trinityrnaseq.sourceforge.net. The run time of this protocol is highly dependent on the size and complexity of data to be analyzed. The example data set analyzed in the procedure detailed herein can be processed in less than 5 h.
Collapse
Affiliation(s)
- Brian J. Haas
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Alexie Papanicolaou
- CSIRO Ecosystem Sciences, Black Mountain Labs, Canberra, ACT 2601, Australia
| | - Moran Yassour
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
- The Selim and Rachel Benin School of Computer Science, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Manfred Grabherr
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Philip D. Blood
- Pittsburgh Supercomputing Center, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Joshua Bowden
- CSIRO Information Management & Technology, 306 Carmody Rd, St Lucia QLD 4067, Australia
| | - Matthew Brian Couger
- Department of Microbiology and Molecular Genetics, Oklahoma State University, USA
| | - David Eccles
- Genomics Research Centre, Griffith University, Gold Coast Campus, Qld 4222, Australia
| | - Bo Li
- Department of Computer Sciences, University of Wisconsin, Madison, WI, 53706, USA
| | - Matthias Lieber
- Technische Universität Dresden, Dresden, Saxony 01062, Germany
| | - Matthew D. MacManes
- University of California, Berkeley and California Institute for Quantitative Biosciences Berkeley, CA 94720, USA
| | - Michael Ott
- CSIRO Ecosystem Sciences, Black Mountain Labs, Canberra, ACT 2601, Australia
| | - Joshua Orvis
- Institute for Genome Sciences, Baltimore, MD, 21201, USA
| | - Nathalie Pochet
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
- Department of Plant Systems Biology, VIB, Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent B-9052, Belgium
| | | | - Nathan Weeks
- Corn Insects and Crop Genetics Research Unit, United States Department of Agriculture--Agricultural Research Service, Ames, IA 50011, USA
| | - Rick Westerman
- Genomics facility, Purdue University, West Lafayette, IN, 47907, USA
| | - Thomas William
- GWT-TUD GmbH, Blasewitzer Strasse 43, Dresden, Saxony 01307, Germany
| | - Colin N. Dewey
- Department of Computer Sciences, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53706, USA
| | - Robert Henschel
- Indiana University, 2709 East 10th Street, Bloomington, IN 47408,USA
| | - Richard D. LeDuc
- Indiana University, 2709 East 10th Street, Bloomington, IN 47408,USA
| | - Nir Friedman
- The Selim and Rachel Benin School of Computer Science, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Aviv Regev
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02140
| |
Collapse
|