1
|
Piórek A, Płużański A, Knetki-Wróblewska M, Winiarczyk K, Tabor S, Kowalski DM, Krzakowski M. Tracheal Tumors: Clinical Practice Guidelines for Palliative Treatment and Follow-Up. Oncol Rev 2024; 18:1451247. [PMID: 39360235 PMCID: PMC11445028 DOI: 10.3389/or.2024.1451247] [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: 06/18/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
A substantial portion of patients with advanced cancer cannot be cured, regardless of the therapeutic methods employed. Hence, rational palliative causal treatment becomes crucial. Representative studies specifically addressing the exclusive palliative treatment of patients diagnosed with tracheal cancers have not been identified. In most studies, patients treated palliatively constituted a subset of the overall evaluated group. A thorough literature review was conducted, focusing on three types of palliative treatment: palliative radiotherapy, palliative surgical procedures, and systemic treatment for advanced disease. This review uniquely fills a significant gap in the existing literature by providing the first comprehensive and updated clinical practice guidelines specifically focused on the palliative treatment of tracheal tumors. The proposed guidelines emphasize the unique clinical challenges and treatment strategies pertinent to palliative care in tracheal tumors, which are not adequately covered in existing guidelines for other thoracic malignancies.
Collapse
Affiliation(s)
- Aleksandra Piórek
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Adam Płużański
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Knetki-Wróblewska
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Kinga Winiarczyk
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Sylwia Tabor
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Dariusz M Kowalski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maciej Krzakowski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
2
|
Wang L, Tang J. SWI/SNF complexes and cancers. Gene 2023; 870:147420. [PMID: 37031881 DOI: 10.1016/j.gene.2023.147420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
Epigenetics refers to the study of genetic changes that can affect gene expression without altering the underlying DNA sequence, including DNA methylation, histone modification, chromatin remodelling, X chromosome inactivation and non-coding RNA regulation. Of these, DNA methylation, histone modification and chromatin remodelling constitute the three classical modes of epigenetic regulation. These three mechanisms alter gene transcription by adjusting chromatin accessibility, thereby affecting cell and tissue phenotypes in the absence of DNA sequence changes. In the presence of ATP hydrolases, chromatin remodelling alters the structure of chromatin and thus changes the transcription level of DNA-guided RNA. To date, four types of ATP-dependent chromatin remodelling complexes have been identified in humans, namely SWI/SNF, ISWI, INO80 and NURD/MI2/CHD. SWI/SNF mutations are prevalent in a wide variety of cancerous tissues and cancer-derived cell lines as discovered by next-generation sequencing technologies.. SWI/SNF can bind to nucleosomes and use the energy of ATP to disrupt DNA and histone interactions, sliding or ejecting histones, altering nucleosome structure, and changing transcriptional and regulatory mechanisms. Furthermore, mutations in the SWI/SNF complex have been observed in approximately 20% of all cancers. Together, these findings suggest that mutations targeting the SWI/SNF complex may have a positive impact on tumorigenesis and cancer progression.
Collapse
Affiliation(s)
- Liyuan Wang
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Oncology and Hematology, Jinan 250000, Shandong Province, China
| | - Jinglong Tang
- Adicon Medical Laboratory Center, Molecular Genetic Diagnosis Center, Pathological Diagnosis Center, Jinan 250014, Shandong Province, China.
| |
Collapse
|
3
|
Titmuss E, Milne K, Jones MR, Ng T, Topham JT, Brown SD, Schaeffer DF, Kalloger S, Wilson D, Corbett RD, Williamson LM, Mungall K, Mungall AJ, Holt RA, Nelson BH, Jones SJM, Laskin J, Lim HJ, Marra MA. Immune Activation following Irbesartan Treatment in a Colorectal Cancer Patient: A Case Study. Int J Mol Sci 2023; 24:5869. [PMID: 36982943 PMCID: PMC10051648 DOI: 10.3390/ijms24065869] [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: 02/18/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal cancers are one of the most prevalent tumour types worldwide and, despite the emergence of targeted and biologic therapies, have among the highest mortality rates. The Personalized OncoGenomics (POG) program at BC Cancer performs whole genome and transcriptome analysis (WGTA) to identify specific alterations in an individual's cancer that may be most effectively targeted. Informed using WGTA, a patient with advanced mismatch repair-deficient colorectal cancer was treated with the antihypertensive drug irbesartan and experienced a profound and durable response. We describe the subsequent relapse of this patient and potential mechanisms of response using WGTA and multiplex immunohistochemistry (m-IHC) profiling of biopsies before and after treatment from the same metastatic site of the L3 spine. We did not observe marked differences in the genomic landscape before and after treatment. Analyses revealed an increase in immune signalling and infiltrating immune cells, particularly CD8+ T cells, in the relapsed tumour. These results indicate that the observed anti-tumour response to irbesartan may have been due to an activated immune response. Determining whether there may be other cancer contexts in which irbesartan may be similarly valuable will require additional studies.
Collapse
Affiliation(s)
- E. Titmuss
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - K. Milne
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
| | - M. R. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - T. Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - J. T. Topham
- Pancreas Centre BC, Vancouver, BC V5Z 1G1, Canada
| | - S. D. Brown
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | | | - S. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - D. Wilson
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - R. D. Corbett
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - L. M. Williamson
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - K. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - A. J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - R. A. Holt
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - B. H. Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - S. J. M. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - J. Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - H. J. Lim
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - M. A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| |
Collapse
|
4
|
Ferrarotto R, Mishra V, Herz E, Yaacov A, Solomon O, Rauch R, Mondshine A, Motin M, Leibovich-Rivkin T, Davis M, Kaye J, Weber CR, Shen L, Pearson AT, Rosenberg AJ, Chen X, Singh A, Aster JC, Agrawal N, Izumchenko E. AL101, a gamma-secretase inhibitor, has potent antitumor activity against adenoid cystic carcinoma with activated NOTCH signaling. Cell Death Dis 2022; 13:678. [PMID: 35931701 PMCID: PMC9355983 DOI: 10.1038/s41419-022-05133-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023]
Abstract
Adenoid cystic carcinoma (ACC) is an aggressive salivary gland malignancy with limited treatment options for recurrent or metastatic disease. Due to chemotherapy resistance and lack of targeted therapeutic approaches, current treatment options for the localized disease are limited to surgery and radiation, which fails to prevent locoregional recurrences and distant metastases in over 50% of patients. Approximately 20% of patients with ACC carry NOTCH-activating mutations that are associated with a distinct phenotype, aggressive disease, and poor prognosis. Given the role of NOTCH signaling in regulating tumor cell behavior, NOTCH inhibitors represent an attractive potential therapeutic strategy for this subset of ACC. AL101 (osugacestat) is a potent γ-secretase inhibitor that prevents activation of all four NOTCH receptors. While this investigational new drug has demonstrated antineoplastic activity in several preclinical cancer models and in patients with advanced solid malignancies, we are the first to study the therapeutic benefit of AL101 in ACC. Here, we describe the antitumor activity of AL101 using ACC cell lines, organoids, and patient-derived xenograft models. Specifically, we find that AL101 has potent antitumor effects in in vitro and in vivo models of ACC with activating NOTCH1 mutations and constitutively upregulated NOTCH signaling pathway, providing a strong rationale for evaluation of AL101 in clinical trials for patients with NOTCH-driven relapsed/refractory ACC.
Collapse
Affiliation(s)
- Renata Ferrarotto
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vasudha Mishra
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Elad Herz
- Ayala Pharmaceuticals, Rehovot, Israel
| | | | | | | | | | | | | | | | - Joel Kaye
- Ayala Pharmaceuticals, Rehovot, Israel
| | | | - Le Shen
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Alexander T Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Xiangying Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Alka Singh
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nishant Agrawal
- Department of Surgery, Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, IL, USA
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
5
|
Wagner VP, Bingle CD, Bingle L. MYB-NFIB fusion transcript in Adenoid Cystic Carcinoma: current state of knowledge and future directions. Crit Rev Oncol Hematol 2022; 176:103745. [PMID: 35738530 DOI: 10.1016/j.critrevonc.2022.103745] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is the most common type of salivary gland cancer that can also arise in other primary sites. Regardless of the site, most ACC cases carry a recurrent chromosomal translocation - t(6;9)(q22-23;p23-24) - involving the MYB oncogene and the NFIB transcription factor. Generally, a long sequence of MYB is fused to the terminal exons of NFIB, yet the break can occur in different exons for both genes, resulting in multiple chimeric variants. The fusion status can be determined by a number of methods, each of them with particular advantages. In vitro and in vivo studies have been conducted to understand the biological consequences of MYB-NFIB translocation, and such findings could contribute to improving the current inefficient therapeutic options for disseminated ACC. This review provides a discussion on relevant evidence in the context of ACC MYB-NFIB translocations to determine the current state of knowledge and discuss future directions.
Collapse
Affiliation(s)
- Vivian P Wagner
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield, UK.
| | - Colin D Bingle
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Lynne Bingle
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
6
|
Fusco N, Sajjadi E, Venetis K, Ivanova M, Andaloro S, Guerini-Rocco E, Montagna E, Caldarella P, Veronesi P, Colleoni M, Viale G. Low-risk triple-negative breast cancers: Clinico-pathological and molecular features. Crit Rev Oncol Hematol 2022; 172:103643. [PMID: 35217131 DOI: 10.1016/j.critrevonc.2022.103643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancers (TNBC) comprise biologically and clinically heterogeneous diseases characterized by the lack of hormone receptors (HR) and HER2 expression. This subset of tumors accounts for 15-20% of all breast cancers and pursues an ominous clinical course. However, there is a spectrum of low-risk TNBCs with no/minimal metastatic potential, including the salivary gland-type tumors, those with extensive apocrine differentiation and/or high tumor-infiltrating lymphocytes, and small-sized, early-stage (pT1a/bN0M0) TNBCs. De-escalating the treatment in low-risk TNBC, however, is not trivial because of the substantial lack of dedicated randomized clinical trials and cancer registries. The development of new diagnostic and/or prognostic biomarkers based on clinical and molecular aspects of low-risk TNBCs would lead to improved clinical treatment. Here, we sought to provide a portrait of the clinicopathological and molecular features of low-risk TNBC, with a focus on the diagnostic challenges along with the most important biological characteristics underpinning their favorable clinical course.
Collapse
Affiliation(s)
- Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy.
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Silvia Andaloro
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Emilia Montagna
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Pietro Caldarella
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Paolo Veronesi
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| |
Collapse
|
7
|
Small molecules targeting γ-secretase and their potential biological applications. Eur J Med Chem 2022; 232:114169. [DOI: 10.1016/j.ejmech.2022.114169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 12/14/2022]
|
8
|
Zheng S, Li H, Lin Y, Xie Y, Yin Z, Ge W, Yu T. Treatment response to eribulin and anlotinib in lung metastases from rare perianal adenoid cystic carcinoma: a case report. Anticancer Drugs 2022; 33:e548-e554. [PMID: 34321419 PMCID: PMC8670344 DOI: 10.1097/cad.0000000000001171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/07/2021] [Indexed: 11/25/2022]
Abstract
Adenoid cystic carcinoma (ACC) is a rare salivary glands tumor and often displays aggressive behavior with frequent relapse and metastasis. The terminal ACC lacks standard treatment guidelines and is always accompanied by poor prognosis. Here, we report a case of rare perianal ACC who received resection and palliative adjuvant radiation. Five years later, PET-computed tomography (CT) showed perianal recurrence and multiple pulmonary metastases. Combined chemotherapy with doxorubicin, carboplatin and cyclophosphamide was applied for two cycles but ineffective. Further next-generation sequencing analysis of perianal tissue demonstrated the v-myb avian myelobastosis viral oncogene homolog and nuclear factor I/B fusion gene and two novel BCL-6 corepressor (BCOR) mutations (p.F1106Tfs*5 and p.L1524Hfs*8). The therapy was switched to eribulin and anlotinib and has been performed for eight cycles. At recent follow-ups, MRI and CT examinations revealed the diminishing perianal and pulmonary lesions. This study presented the first case of perianal ACC with multiple pulmonary metastases and particular BCOR mutations, who presented a durable response to eribulin and anlotinib, providing a potential therapeutic option for advanced refractory ACC.
Collapse
Affiliation(s)
- Shengnan Zheng
- Department of Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Xuzhou, Jiangsu
- Department of Laboratory Medicine, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu
| | - Huiying Li
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yongjuan Lin
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yu Xie
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhenyu Yin
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Weihong Ge
- Department of Laboratory Medicine, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu
| | - Tingting Yu
- Department of Laboratory Medicine, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu
| |
Collapse
|
9
|
Dou S, Li R, He N, Zhang M, Jiang W, Ye L, Yang Y, Zhao G, Yang Y, Li J, Chen D, Zhu G. The Immune Landscape of Chinese Head and Neck Adenoid Cystic Carcinoma and Clinical Implication. Front Immunol 2021; 12:618367. [PMID: 34552580 PMCID: PMC8450584 DOI: 10.3389/fimmu.2021.618367] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
Novel systemic agents and effective treatment strategies for recurrence adenoid cystic carcinoma (ACC) of the head and neck are still worthy of further exploration. Here, we analyzed the mutations and expression profiles of 75 Chinese ACC patients, characterized the prognostic value of the immune signature for recurrence or distant metastasis, and explored the potential of immunotherapeutic biomarkers in ACC. In general, MYB fusion and somatic mutations accounted for a high proportion, which was 46.7% (35/75). ACCs displayed an overall low mutation burden and lack of programmed cell death ligand-1 (PD-L1) expression. The antigen-presenting machinery (APM) expression score and immune infiltration score (IIS) were the lowest among ACC patients, compared with other cancer types. For 61 primary cases, the locoregional recurrence-free survival (LRRFS) was statistically significantly correlated with the IIS [univariate analysis; hazard ratio (HR) = 0.32; 95% CI, 0.11–0.92; p = 0.035] and T-cell infiltration score (TIS) (univariate analysis; HR = 0.33; 95% CI, 0.12–0.94; p = 0.037]. Patients with lower IIS (log-rank p = 0.0079) or TIS (log-rank p = 0.0079) had shorter LRRFS. Additionally, solid pattern was also a prognostic factor related to locoregional recurrence, whereas postoperative radiotherapy (PORT) exerted its beneficial effects. We further evaluated the pretreatment immune profile of five ACC patients treated with PD-1 inhibitors. Patients who responded to camrelizumab or pembrolizumab observed elevated APM and TIS, compared with patients with progressive disease. Our study highlights the immune infiltration pattern and messenger RNA (mRNA) signatures of Chinese ACC patients, which has the potential value for prognosis and immunotherapy.
Collapse
Affiliation(s)
- Shengjin Dou
- Radiotherapy Division, Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Rongrong Li
- Radiotherapy Division, Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ning He
- GloriousMed Technology Co., Ltd, Shanghai, China
| | | | - Wen Jiang
- Radiotherapy Division, Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lulu Ye
- Radiotherapy Division, Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yining Yang
- GloriousMed Technology Co., Ltd, Shanghai, China
| | - Guodong Zhao
- GloriousMed Technology Co., Ltd, Shanghai, China
| | - Yadong Yang
- GloriousMed Technology Co., Ltd, Shanghai, China
| | - Jiang Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Chen
- GloriousMed Technology Co., Ltd, Shanghai, China
| | - Guopei Zhu
- Radiotherapy Division, Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
10
|
Wolkow N, Jakobiec FA, Afrogheh AH, Kidd M, Eagle RC, Pai SI, Faquin WC. PD-L1 and PD-L2 Expression Levels Are Low in Primary and Secondary Adenoid Cystic Carcinomas of the Orbit: Therapeutic Implications. Ophthalmic Plast Reconstr Surg 2021; 36:444-450. [PMID: 31990894 PMCID: PMC7423458 DOI: 10.1097/iop.0000000000001585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE To determine if there is a biologic rationale for using checkpoint inhibitor drugs targeting programmed cell death ligand 1 (PD-L1) and PD-L2 in the treatment of adenoid cystic carcinoma of the orbit. METHODS Twenty-three cases of adenoid cystic carcinoma involving the orbit (13 primary lacrimal gland, 5 secondarily extending into the orbit, and 5 unspecified) were examined histopathologically. Immunohistochemistry for PD-L1, PD-L2, and CD8 was performed. Charts were reviewed for clinical correlations. RESULTS Expression of PD-L1 and of PD-L2 was overall low in adenoid cystic carcinoma (mean expression 1.4 ± 0.9 of 5 for PD-L1, mean 0.83 ± 1.1 of 5 for PD-L2), and tumor-infiltrating CD8-positive T-lymphocytes were sparse (mean 1.1 ± 0.51 of 3). Only 13 of the 23 (57%) cases expressed PD-L1 as a combined positive score ≥1 of cells. No associations were found between expression levels of these markers and patient sex, tumor site of origin, Tumor, Node, Metastasis stage, or patient outcome. A significant association was observed between stromal PD-L1 expression and tumor histopathologic subtype (p = 0.05), and between tumor PD-L1 expression and prior exposure to radiation (p = 0.03). CONCLUSIONS Checkpoint inhibitor drugs may have limited impact in the treatment and clinical course of orbital adenoid cystic carcinoma based on the low frequency of CD8 infiltrate and low expression of PD-L1 and PD-L2. Pretreatment with radiation, however, may improve tumor response to checkpoint inhibitor drugs.
Collapse
Affiliation(s)
- Natalie Wolkow
- David G. Cogan Ophthalmic Pathology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, U.S.A
- Ophthalmic Plastic and Reconstructive Surgery Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Frederick A. Jakobiec
- David G. Cogan Ophthalmic Pathology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Amir H. Afrogheh
- Department of Oral and Maxillofacial Pathology, National Health Laboratory Service, University of the Western Cape, Cape Town, South Africa
| | - Martin Kidd
- Centre for Statistical Consultation, Department of Statistics and Actuarial Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Ralph C. Eagle
- Department of Ophthalmic Pathology, Wills Eye Hospital, Philadelphia, Pennsylvania, U.S.A
| | - Sara I. Pai
- Department of Surgery, Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - William C. Faquin
- Division of Head and Neck Pathology, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, U.S.A
| |
Collapse
|
11
|
Palsgrove D, Allahabadi S, Khan SA. Genomic Analysis of Salivary Gland Cancer and Treatment of Salivary Gland Cancers. Surg Pathol Clin 2021; 14:151-163. [PMID: 33526219 DOI: 10.1016/j.path.2020.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Salivary gland cancer is a heterogenous group of tumors that presents challenges with both diagnosis and therapy. Recent advances in the classification of salivary gland cancers have led to distinct histologic and genomic criteria that successfully differentiate between cancers with similar clinical behavior and appearance. Genomic abnormalities have led to the emergence of targeted therapies being used in their therapy with drastic improvements in outcomes as well as reductions in treatment-related toxicity. Dramatic results seen with molecular targets, such as HER2, TRK, and others, indicate that this approach has the potential to yield even better treatments for the future.
Collapse
Affiliation(s)
- Doreen Palsgrove
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Sameer Allahabadi
- Texas Christian University, University of North Texas Health Science Center School of Medicine, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Saad A Khan
- Stanford Cancer Institute and Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Arroyo M, Larrosa R, Gómez-Maldonado J, Cobo MÁ, Claros MG, Bautista R. Expression-based, consistent biomarkers for prognosis and diagnosis in lung cancer. Clin Transl Oncol 2020; 22:1867-1874. [PMID: 32180209 DOI: 10.1007/s12094-020-02328-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 02/24/2020] [Indexed: 12/27/2022]
Abstract
OVERVIEW Lung cancer is one of the deadliest cancers in the world. Its histological classification depends on early diagnosis and successful treatment. Therefore, having specific biomarkers for a quick sorting widens the successful output of lung cancer treatment. MATERIAL AND METHODS High-throughput sequencing (RNA-seq) was performed of small cohorts of BioBanco samples from healthy and tumour cells from lung adenocarcinoma (LUAD) and squamous cell carcinoma of the lung (lSCC). RNA-seq samples from small cell lung cancer (SCLC) were downloaded from databases. A bioinformatic workflow has been programmed for the identification of differentially expressed genes (DEGs). RESULTS A total of 4777 DEGs were differentially expressed in SCLC, 3676 DEGs were in lSCC, while the lowest number of DEGs, 2819, appeared in LUAD. Among them, 945 DEGs were common to the three histological types. Once validated their expression profile and their survival predictive capacity in large, public cohorts, three DEGs can be exclusively considered as diagnostic biomarkers, three as prognosis biomarkers, and other three exhibit both diagnosis and prognosis capabilities. CONCLUSIONS This prospective study presents evidences for the diagnostic and prognostic capabilities of expression changes in CAPN8-2, TMC5 and MUC1 in LUAD, while they are non-significant in SCLC and lSCC. Their translation to clinical practice is proposed.
Collapse
Affiliation(s)
- M Arroyo
- U.G.C. Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - R Larrosa
- Department of Computer Architecture, Universidad de Málaga, Málaga, Spain
| | - J Gómez-Maldonado
- Sequencing and Genomics Unit at SCBI, Universidad de Málaga, Málaga, Spain
| | - M Á Cobo
- Unidad Intercentro, Hospital Regional Universitario de Málaga y VV. IBIMA, Málaga, Spain
| | - M G Claros
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, Málaga, Spain.
| | - R Bautista
- Andalusian Platform for Bioinformatics at SCBI, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
13
|
Jiang Y, Gao R, Cao C, Forbes L, Li J, Freeberg S, Fredenburg KM, Justice JM, Silver NL, Wu L, Varma S, West R, Licht JD, Zajac-Kaye M, Kentsis A, Kaye FJ. MYB-activated models for testing therapeutic agents in adenoid cystic carcinoma. Oral Oncol 2019; 98:147-155. [PMID: 31606723 DOI: 10.1016/j.oraloncology.2019.09.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/26/2019] [Accepted: 09/06/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE There are no effective systemic therapies for adenoid cystic cancer (ACC) and lack of tumor lines and mouse models have hindered drug development.We aim to develop MYB-activated models for testing new therapeutic agents. MATERIALS AND METHODS We studied new ACC patient-derived xenograft (PDX) models and generated a matched cell line from one patient. In addition, we generated a genetically-engineered MYB-NFIB mouse model (GEMM) that was crossed with Ink4a+/-/Arf+/- mice to study tumor spectrum and obtain tumor lines. Using human and murine ACC-like tumor lines, we analyzed MYB expression by RNA-Seq and immunoblot and tested efficacy of new MYB inhibitors. RESULTS We detected MYB-NFIB transcripts in both UFH1 and UFH2 PDX and observed tumor inhibition by MYB depletion using shRNA in vivo. We observed rapid loss of MYB expression when we cultured UFH1 in vitro, but were able to generate a UFH2 tumor cell line that retained MYB expression for 6 months. RNA-Seq expression detected an ACC-like mRNA signature in PDX samples and we confirmed an identical KMT2A/MLL variant in UFH2 PDX, matched cell line, and primary biopsy. Although the predominant phenotype of the MYB-NFIB GEMM was B-cell leukemia, we also generated a MYB-activated ACC-like mammary tumor cell line. We observed tumor inhibition using a novel MYB peptidomimetic in both human and murine tumor models. CONCLUSIONS We generated and studied new murine and human MYB-activated tumor samples and detected growth inhibition with MYB peptidomimetics. These data provide tools to define treatment strategies for patients with advanced MYB-activated ACC.
Collapse
Affiliation(s)
- Yue Jiang
- Department Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Ruli Gao
- Department Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunxia Cao
- Department Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Lauren Forbes
- Molecular Pharmacology Program, Sloan Kettering Institute and Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jianping Li
- Department Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Shelby Freeberg
- Department Medicine, University of Florida, Gainesville, FL 32608, USA
| | | | - Jeb M Justice
- Department Otolaryngology, University of Florida, Gainesville, FL 32608, USA
| | - Natalie L Silver
- Department Otolaryngology, University of Florida, Gainesville, FL 32608, USA
| | - Lizi Wu
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL 32608, USA
| | - Sushama Varma
- Department Pathology, Stanford University Medical Center, Palo Alto, CA 94304, USA
| | - Robert West
- Department Pathology, Stanford University Medical Center, Palo Alto, CA 94304, USA
| | - Jonathan D Licht
- Department Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Maria Zajac-Kaye
- Department Anatomy Cell Biology, University of Florida, Gainesville, FL 32608, USA
| | - Alex Kentsis
- Molecular Pharmacology Program, Sloan Kettering Institute and Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Frederic J Kaye
- Department Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
14
|
Abstract
BCOR is a gene that encodes for an epigenetic regulator involved in the specification of cell differentiation and body structure development and takes part in the noncanonical polycomb repressive complex 1. This review provides a comprehensive summary of BCOR’s involvement in oncology, illustrating that various BCOR aberrations, such as the internal tandem duplications of the PCGF Ub-like fold discriminator domain and different gene fusions (mainly BCOR–CCNB3, BCOR–MAML3 and ZC3H7B–BCOR), represent driver elements of various sarcomas such as clear cell sarcoma of the kidney, primitive mesenchymal myxoid tumor of infancy, small round blue cell sarcoma, endometrial stromal sarcoma and histologically heterogeneous CNS neoplasms group with similar genomic methylation patterns known as CNS-HGNET-BCOR. Furthermore, other BCOR alterations (often loss of function mutations) recur in a large variety of mesenchymal, epithelial, neural and hematological tumors, suggesting a central role in cancer evolution.
Collapse
Affiliation(s)
- Annalisa Astolfi
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Michele Fiore
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Fraia Melchionda
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Valentina Indio
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Salvatore N Bertuccio
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Andrea Pession
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy.,Department of Medical & Surgical Sciences, University of Bologna, S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| |
Collapse
|
15
|
Grewal JK, Tessier-Cloutier B, Jones M, Gakkhar S, Ma Y, Moore R, Mungall AJ, Zhao Y, Taylor MD, Gelmon K, Lim H, Renouf D, Laskin J, Marra M, Yip S, Jones SJM. Application of a Neural Network Whole Transcriptome-Based Pan-Cancer Method for Diagnosis of Primary and Metastatic Cancers. JAMA Netw Open 2019; 2:e192597. [PMID: 31026023 PMCID: PMC6487574 DOI: 10.1001/jamanetworkopen.2019.2597] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE A molecular diagnostic method that incorporates information about the transcriptional status of all genes across multiple tissue types can strengthen confidence in cancer diagnosis. OBJECTIVE To determine the practical use of a whole transcriptome-based pan-cancer method in diagnosing primary and metastatic cancers and resolving complex diagnoses. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional diagnostic study assessed Supervised Cancer Origin Prediction Using Expression (SCOPE), a machine learning method using whole-transcriptome RNA sequencing data. Training was performed on publicly available primary cancer data sets, including The Cancer Genome Atlas. Testing was performed retrospectively on untreated primary cancers and treated metastases from volunteer adult patients at BC Cancer in Vancouver, British Columbia, from January 1, 2013, to March 31, 2016, and testing spanned 10 822 samples and 66 output classes representing untreated primary cancers (n = 40) and adjacent normal tissues (n = 26). SCOPE's performance was demonstrated on 211 untreated primary mesothelioma cancers and 201 treatment-resistant metastatic cancers. Finally, SCOPE was used to identify the putative site of origin in 15 cases with initial presentation as cancers with unknown primary of origin. RESULTS A total of 10 688 adult patient samples representing 40 untreated primary tumor types and 26 adjacent-normal tissues were used for training. Demographic data were not available for all data sets. Among the training data set, 5157 of 10 244 (50.3%) were male and the mean (SD) age was 58.9 (14.5) years. Testing was performed on 211 patients with untreated primary mesothelioma (173 [82.0%] male; mean [SD] age, 64.5 [11.3] years); 201 patients with treatment-resistant cancers (141 [70.1%] female; mean [SD] age, 55.6 [12.9] years); and 15 patients with cancers of unknown primary of origin; among the treatment-resistant cancers, 168 were metastatic, and 33 were the primary presentation. An accuracy rate of 99% was obtained for primary epithelioid mesotheliomas tested (125 of 126). The remaining 85 mesotheliomas had a mixed etiology (sarcomatoid mesotheliomas) and were correctly identified as a mixture of their primary components, with potential implications in resolving subtypes and incidences of mixed histology. SCOPE achieved an overall mean (SD) accuracy rate of 86% (11%) and F1 score of 0.79 (0.12) on the 201 treatment-resistant cancers and matched 12 of 15 of the putative diagnoses for cancers with indeterminate diagnosis from conventional pathology. CONCLUSIONS AND RELEVANCE These results suggest that machine learning approaches incorporating multiple tumor profiles can more accurately identify the cancerous state and discriminate it from normal cells. SCOPE uses the whole transcriptomes from normal and tumor tissues, and results of this study suggest that it performs well for rare cancer types, primary cancers, treatment-resistant metastatic cancers, and cancers of unknown primary of origin. Genes most relevant in SCOPE's decision making were examined, and several are known biological markers of respective cancers. SCOPE may be applied as an orthogonal diagnostic method in cases where the site of origin of a cancer is unknown, or when standard pathology assessment is inconclusive.
Collapse
Affiliation(s)
- Jasleen K. Grewal
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Basile Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Jones
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Sitanshu Gakkhar
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Yussanne Ma
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Richard Moore
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Andrew J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Yongjun Zhao
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Michael D. Taylor
- Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen Gelmon
- Division of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Howard Lim
- Division of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Daniel Renouf
- Division of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Janessa Laskin
- Division of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Marco Marra
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven J. M. Jones
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Thierauf J, Ramamurthy N, Jo VY, Robinson H, Frazier RP, Gonzalez J, Pacula M, Dominguez Meneses E, Nose V, Nardi V, Dias-Santagata D, Le LP, Lin DT, Faquin WC, Wirth LJ, Hess J, Iafrate AJ, Lennerz JK. Clinically Integrated Molecular Diagnostics in Adenoid Cystic Carcinoma. Oncologist 2019; 24:1356-1367. [PMID: 30926674 PMCID: PMC6795155 DOI: 10.1634/theoncologist.2018-0515] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 02/27/2019] [Indexed: 01/29/2023] Open
Abstract
Adenoid cystic carcinoma is a rare but aggressive type of salivary gland malignancy. This article addresses the need for more effective, biomarker‐informed therapies in rare cancers, focusing on clinical utility and financial sustainability of integrated next‐generation sequencing in routine practice. Background. Adenoid cystic carcinoma (ACC) is an aggressive salivary gland malignancy without effective systemic therapies. Delineation of molecular profiles in ACC has led to an increased number of biomarker‐stratified clinical trials; however, the clinical utility and U.S.‐centric financial sustainability of integrated next‐generation sequencing (NGS) in routine practice has, to our knowledge, not been assessed. Materials and Methods. In our practice, NGS genotyping was implemented at the discretion of the primary clinician. We combined NGS‐based mutation and fusion detection, with MYB break‐apart fluorescent in situ hybridization (FISH) and MYB immunohistochemistry. Utility was defined as the fraction of patients with tumors harboring alterations that are potentially amenable to targeted therapies. Financial sustainability was assessed using the fraction of global reimbursement. Results. Among 181 consecutive ACC cases (2011–2018), prospective genotyping was performed in 11% (n = 20/181; n = 8 nonresectable). Testing identified 5/20 (25%) NOTCH1 aberrations, 6/20 (30%) MYB‐NFIB fusions (all confirmed by FISH), and 2/20 (10%) MYBL1‐NFIB fusions. Overall, these three alterations (MYB/MYBL1/NOTCH1) made up 65% of patients, and this subset had a more aggressive course with significantly shorter progression‐free survival. In 75% (n = 6/8) of nonresectable patients, we detected potentially actionable alterations. Financial analysis of the global charges, including NGS codes, indicated 63% reimbursement, which is in line with national (U.S.‐based) and international levels of reimbursement. Conclusion. Prospective routine clinical genotyping in ACC can identify clinically relevant subsets of patients and is approaching financial sustainability. Demonstrating clinical utility and financial sustainability in an orphan disease (ACC) requires a multiyear and multidimensional program. Implications for Practice. Delineation of molecular profiles in adenoid cystic carcinoma (ACC) has been accomplished in the research setting; however, the ability to identify relevant patient subsets in clinical practice has not been assessed. This work presents an approach to perform integrated molecular genotyping of patients with ACC with nonresectable, recurrent, or systemic disease. It was determined that 75% of nonresectable patients harbor potentially actionable alterations and that 63% of charges are reimbursed. This report outlines that orphan diseases such as ACC require a multiyear, multidimensional program to demonstrate utility in clinical practice.
Collapse
Affiliation(s)
- Julia Thierauf
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Nisha Ramamurthy
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Vickie Y Jo
- Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hayley Robinson
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan P Frazier
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Gonzalez
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Maciej Pacula
- Department of Pathology, Computational Pathology, Boston, Massachusetts, USA
| | | | - Vania Nose
- Department of Pathology, Head and Neck Pathology, Boston, Massachusetts, USA
- Department of Pathology, Surgical Pathology, Boston, Massachusetts, USA
| | - Valentina Nardi
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Dora Dias-Santagata
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Long P Le
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Computational Pathology, Boston, Massachusetts, USA
| | - Derrick T Lin
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - William C Faquin
- Department of Pathology, Surgical Pathology, Boston, Massachusetts, USA
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Lori J Wirth
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A John Iafrate
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
A high-throughput protocol for isolating cell-free circulating tumor DNA from peripheral blood. Biotechniques 2019; 66:85-92. [PMID: 30744412 DOI: 10.2144/btn-2018-0148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The analysis of cell-free circulating tumor DNA (ctDNA) is potentially a less invasive, more dynamic assessment of cancer progression and treatment response than characterizing solid tumor biopsies. Standard isolation methods require separation of plasma by centrifugation, a time-consuming step that complicates automation. To address these limitations, we present an automatable magnetic bead-based ctDNA isolation method that eliminates centrifugation to purify ctDNA directly from peripheral blood (PB). To develop and test our method, ctDNA from cancer patients was purified from PB and plasma. We found that allelic fractions of somatic single-nucleotide variants from target gene capture libraries were comparable, indicating that the PB ctDNA purification method may be a suitable replacement for the plasma-based protocols currently in use.
Collapse
|
18
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019. [PMID: 30636931 DOI: 10.1186/s12935-019-0725-1]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. METHODS We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. RESULTS Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. CONCLUSIONS KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
19
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019. [PMID: 30636931 DOI: 10.1186/s12935-019-0725-1] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. Methods We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. Results Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. Conclusions KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
20
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019; 19:10. [PMID: 30636931 PMCID: PMC6325847 DOI: 10.1186/s12935-019-0725-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. Methods We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. Results Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. Conclusions KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC. Electronic supplementary material The online version of this article (10.1186/s12935-019-0725-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|