1
|
Sofiani VH, Veisi P, Rukerd MRZ, Ghazi R, Nakhaie M. The complexity of human papilloma virus in cancers: a narrative review. Infect Agent Cancer 2023; 18:13. [PMID: 36843070 PMCID: PMC9969657 DOI: 10.1186/s13027-023-00488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/09/2023] [Indexed: 02/27/2023] Open
Abstract
Among human tumorigenic viruses, the role of Human papillomavirus (HPV) has been proven as one of the most important oncoviruses that are associated with a large number of cancers. Most cancers of the genital area such cervical and anal cancer as are caused by HPV, and in many other cancers, such as colorectal, gastric, liver, esophageal, urinary bladder, and head and neck cancers, it is considered as one of the important risk factors. Our search was conducted for published researches between 2000 and 2022 by using several international databases including Scopus, PubMed, and Web of Science as well as Google scholar. We also evaluated additional evidence from relevant published articles. It has been demonstrated that HPV can promote tumorigenesis via focusing on genes, proteins, and signaling pathways, by using E6 and E7 oncoproteins and inhibiting two crucial tumor suppressors, P53 and Rb. The following study was performed to investigate different malignant cancers under the influence of HPV infection and changes in molecular factors caused by HPV infection.
Collapse
Affiliation(s)
- Vahideh Hamidi Sofiani
- grid.411747.00000 0004 0418 0096Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Parsa Veisi
- grid.411747.00000 0004 0418 0096Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Rezaei Zadeh Rukerd
- grid.412105.30000 0001 2092 9755Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Ghazi
- Department of Biotechnology, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Mohsen Nakhaie
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Gunder LC, Moyer TH, Johnson HR, Auyeung AS, Leverson GE, Zhang W, Matkowskyj KA, Carchman EH. Anal Cancer Prevention Through the Topical Use of Single or Dual PI3K/mTOR Inhibitors. J Surg Res 2023; 282:137-146. [PMID: 36274448 DOI: 10.1016/j.jss.2022.09.025] [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: 01/06/2022] [Revised: 08/29/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Anal dysplasia and anal cancer are major health problems. This study seeks to determine if inhibition of mTOR and/or PI3K pathways is effective at anal cancer prevention in mice with/without established precancerous lesions of the anus (anal dysplasia). METHODS K14E6/E7 mice were entered into the study at 5 wk, 15 wk, or 25 wk of age. Mice were treated with a topical carcinogen, 7,12-Dimethylbenz[a]anthracene (DMBA), which ensures carcinoma development within 20 wk. Treatment groups included: no treatment, DMBA only, topical Pictilisib (PI3K inhibitor) with/without DMBA, topical Sapanisertib (mTOR inhibitor) with/without DMBA, and topical Samotolisib (dual PI3K/mTOR inhibitor) with/without DMBA. Mice underwent weekly observations for anal tumor development (tumor-free survival). After 20 wk of treatment, anal tissue was harvested and evaluated histologically for squamous cell carcinoma (SqCC). RESULTS All topical treatments in conjunction with DMBA increased tumor-free survival in mice that started treatment at 15 wk of age when compared to DMBA-only treatment, except for Pictilisib + DMBA in males. Topical Sapanisertib increased tumor-free survival in mice regardless of starting treatment age. When examining tissue for microscopic evidence of SqCC, only topical Samotolisib in males decreased SqCC in the 15 wk starting mice. CONCLUSIONS Sapanisertib, the mTOR inhibitor, had the greatest effect, in terms of increasing tumor-free survival, regardless of starting time point or sex. Unlike the other treatments, Samotolisib, the dual PI3K/mTOR inhibitor, decreased microscopic evidence of SqCC when starting treatment at 15 wk of age but only in male mice.
Collapse
Affiliation(s)
- Laura C Gunder
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin
| | - Tyra H Moyer
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin
| | - Hillary R Johnson
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin
| | - Andrew S Auyeung
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin
| | - Glen E Leverson
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin
| | - Wei Zhang
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), Madison, Wisconsin; University of Wisconsin-Madison, Carbone Cancer Center, Madison, Wisconsin
| | - Kristina A Matkowskyj
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), Madison, Wisconsin; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin; University of Wisconsin-Madison, Carbone Cancer Center, Madison, Wisconsin
| | - Evie H Carchman
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, 5148 Wisconsin Institute for Medical Research (WIMR), Madison, Wisconsin; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin; University of Wisconsin-Madison, Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
3
|
Standing D, Arnold L, Dandawate P, Ottemann B, Snyder V, Ponnurangam S, Sayed A, Subramaniam D, Srinivasan P, Choudhury S, New J, Kwatra D, Ramamoorthy P, Roy BC, Shadoin M, Al-Rajabi R, O’Neil M, Gunewardena S, Ashcraft J, Umar S, Weir SJ, Tawfik O, Padhye SB, Biersack B, Anant S, Thomas SM. Doublecortin-like kinase 1 is a therapeutic target in squamous cell carcinoma. Mol Carcinog 2023; 62:145-159. [PMID: 36218231 PMCID: PMC9852063 DOI: 10.1002/mc.23472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 01/25/2023]
Abstract
Doublecortin like kinase 1 (DCLK1) plays a crucial role in several cancers including colon and pancreatic adenocarcinomas. However, its role in squamous cell carcinoma (SCC) remains unknown. To this end, we examined DCLK1 expression in head and neck SCC (HNSCC) and anal SCC (ASCC). We found that DCLK1 is elevated in patient SCC tissue, which correlated with cancer progression and poorer overall survival. Furthermore, DCLK1 expression is significantly elevated in human papilloma virus negative HNSCC, which are typically aggressive with poor responses to therapy. To understand the role of DCLK1 in tumorigenesis, we used specific shRNA to suppress DCLK1 expression. This significantly reduced tumor growth, spheroid formation, and migration of HNSCC cancer cells. To further the translational relevance of our studies, we sought to identify a selective DCLK1 inhibitor. Current attempts to target DCLK1 using pharmacologic approaches have relied on nonspecific suppression of DCLK1 kinase activity. Here, we demonstrate that DiFiD (3,5-bis [2,4-difluorobenzylidene]-4-piperidone) binds to DCLK1 with high selectivity. Moreover, DiFiD mediated suppression of DCLK1 led to G2/M arrest and apoptosis and significantly suppressed tumor growth of HNSCC xenografts and ASCC patient derived xenografts, supporting that DCLK1 is critical for SCC growth.
Collapse
Affiliation(s)
- David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Levi Arnold
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Brendan Ottemann
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
| | - Vusala Snyder
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
| | - Sivapriya Ponnurangam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Afreen Sayed
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | | | | | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jacob New
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Deep Kwatra
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Prabhu Ramamoorthy
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Badal C. Roy
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Melissa Shadoin
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
| | - Raed Al-Rajabi
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Maura O’Neil
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John Ashcraft
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Shahid Umar
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Scott J. Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas
| | - Ossama Tawfik
- Department of Pathology, Saint Luke’s Health System, Kansas City, Missouri and MAWD Pathology Group, Kansas City, Kansas
| | | | | | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Sufi Mary Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
Wei E, Li J, Anand P, French LE, Wattad A, Clanner-Engelshofen B, Reinholz M. "From molecular to clinic": The pivotal role of CDC42 in pathophysiology of human papilloma virus related cancers and a correlated sensitivity of afatinib. Front Immunol 2023; 14:1118458. [PMID: 36936942 PMCID: PMC10014535 DOI: 10.3389/fimmu.2023.1118458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background Human papilloma virus (HPV)-related cancers are global health challenge. Insufficient comprehension of these cancers has impeded the development of novel therapeutic interventions. Bioinformatics empowered us to investigate these cancers from new entry points. Methods DNA methylation data of cervical squamous cell carcinoma (CESC) and anal squamous cell carcinoma (ASCC) were analyzed to identify the significantly altered pathways. Through analyses integrated with RNA sequencing data of genes in these pathways, genes with strongest correlation to the TNM staging of CESC was identified and their correlations with overall survival in patients were assessed. To find a potential promising drug, correlation analysis of gene expression levels and compound sensitivity was performed. In vitro experiments were conducted to validate these findings. We further performed molecular docking experiments to explain our findings. Results Significantly altered pathways included immune, HPV infection, oxidative stress, ferroptosis and necroptosis. 10 hub genes in these pathways (PSMD11, RB1, SAE1, TAF15, TFDP1, CORO1C, JOSD1, CDC42, KPNA2 and NUP62) were identified, in which only CDC42 high expression was statistically significantly correlated with overall survival (Hazard Ratio: 1.6, P = 0.045). Afatinib was then screened out to be tested. In vitro experiments exhibited that the expression level of CDC42 was upregulated in HaCaT/A431 cells transfected with HPV E6 and E7, and the inhibitory effect of afatinib on proliferation was enhanced after transfection. CDC42-GTPase-effector interface-EGFR-afatinib was found to be a stable complex with a highest ZDOCK score of 1264.017. Conclusion We identified CDC42 as a pivotal gene in the pathophysiology of HPV-related cancers. The upregulation of CDC42 could be a signal for afatinib treatment and the mechanism in which may be an increased affinity of EGFR to afatinib, inferred from a high stability in the quaternary complex of CDC42-GTPase-effector interface-EGFR-afatinib.
Collapse
Affiliation(s)
- Erdong Wei
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
| | - Jiahua Li
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
- *Correspondence: Jiahua Li,
| | - Philipp Anand
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
| | - Lars E. French
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, Miller School of Medicine, University of Miami, Miami, United States
| | - Adam Wattad
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
| | - Benjamin Clanner-Engelshofen
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
| | - Markus Reinholz
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilians University of Munich (LMU) Munich, Munich, Germany
| |
Collapse
|
5
|
Mathias-Machado MC, Peixoto RD, Moniz CMV, Jácome AA. Biomarkers in Anal Cancer: Current Status in Diagnosis, Disease Progression and Therapeutic Strategies. Biomedicines 2022; 10:2029. [PMID: 36009576 PMCID: PMC9405643 DOI: 10.3390/biomedicines10082029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Squamous cell carcinoma of the anal canal (SCCA) is a rare neoplasm, but with rising incidence rates in the past few decades; it is etiologically linked with the human papillomavirus (HPV) infection and is especially prevalent in immunocompromised patients, mainly those infected with HIV. Fluoropyrimidine-based chemoradiotherapy remains the cornerstone of the treatment of non-metastatic disease, but the locally advanced disease still presents high rates of disease recurrence and systemic therapy of SCCA is an unmet clinical need. Despite sharing common molecular aspects with other HPV-related malignancies, such as cervical and head and neck cancers, SCCA presents specific epigenomic, genomic, and transcriptomic abnormalities, which suggest that genome-guided personalized therapies should be specifically designed for this disease. Actionable mutations are rare in SCCA and immune checkpoint inhibition has not yet been proven useful in an unselected population of patients. Therefore, advances in systemic therapy of SCCA will only be possible with the identification of predictive biomarkers and the subsequent development of targeted therapies or immunotherapeutic approaches that consider the unique tumor microenvironment and the intra- and inter-tumoral heterogeneity. In the present review, we address the molecular characterization of SCCA and discuss potential diagnostic, predictive and prognostic biomarkers of this complex and challenging disease.
Collapse
Affiliation(s)
- Maria Cecília Mathias-Machado
- Department of Gastrointestinal Medical Oncology, Oncoclinicas, São Paulo 04538-132, Brazil
- Department of Oncology, ICESP—Instituto do Cancer do Estado de São Paulo, University of São Paulo, São Paulo 01246-000, Brazil
| | | | - Camila Motta Venchiarutti Moniz
- Department of Oncology, ICESP—Instituto do Cancer do Estado de São Paulo, University of São Paulo, São Paulo 01246-000, Brazil
| | - Alexandre A. Jácome
- Department of Gastrointestinal Medical Oncology, Oncoclinicas, Belo Horizonte 34000-000, Brazil
| |
Collapse
|
6
|
Collienne M, Loghmani H, Heineman TC, Arnold D. GOBLET: a phase I/II study of pelareorep and atezolizumab +/- chemo in advanced or metastatic gastrointestinal cancers. Future Oncol 2022; 18:2871-2878. [PMID: 35796248 DOI: 10.2217/fon-2022-0453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Most gastrointestinal (GI) cancers have microsatellite-stable (MSS) tumors, which have an immunologically 'cold' phenotype with fewer genetic mutations, reduced immune cell infiltration and downregulated immune checkpoint proteins. These attributes make MSS tumors resistant to conventional immunotherapy including checkpoint blockade therapy. Pelareorep is a naturally occurring, nongenetically modified reovirus. Upon intravenous administration, pelareorep selectively kills tumor cells and promotes several immunologic changes that prime tumors to respond to checkpoint blockade therapy. Given its demonstrated synergy with checkpoint blockade, as well as its encouraging efficacy in prior GI cancer studies, pelareorep plus atezolizumab will be evaluated in the GOBLET study in multiple GI cancer indications.
Collapse
Affiliation(s)
- Maike Collienne
- Asklepios Tumorzentrum Hamburg, AK Altona, Oncology, Hematology, Palliative Care, Rheumatology, Paul-Ehrlich-Straße 1, Hamburg, 22763, Germany
| | - Houra Loghmani
- Oncolytics Biotech Inc., Suite 804, 322 - 11th Avenue SW, Calgary, Alberta, T2R 0C5, Canada
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Oncology, Hematology, Palliative Care, Rheumatology, Paul-Ehrlich-Straße 1, Hamburg, 22763, Germany
| |
Collapse
|
7
|
Armstrong SA, Malley R, Wang H, Lenz HJ, Arguello D, El-Deiry WS, Xiu J, Gatalica Z, Hwang JJ, Philip PA, Shields AF, Marshall JL, Salem ME, Weinberg BA. Molecular characterization of squamous cell carcinoma of the anal canal. J Gastrointest Oncol 2021; 12:2423-2437. [PMID: 34790403 DOI: 10.21037/jgo-20-610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Squamous cell carcinoma of the anal canal (SCCA) is an uncommon malignancy with limited therapeutic options. Nivolumab and pembrolizumab show promising results in patients with SCCA. Human papillomavirus (HPV)-negative tumors are frequently TP53-mutated (TP53-MT) and often resistant to therapy. Methods We present a large molecularly-profiled cohort of SCCA, exploring the underlying biology of SCCA, differences between TP53-wild type (TP53-WT) and TP53-MT tumors, and differences between local and metastatic tumors. SCCA specimens (n=311) underwent multiplatform testing with immunohistochemistry (IHC), in situ hybridization (ISH) and next-generation sequencing (NGS). Tumor mutational burden (TMB) was calculated using only somatic nonsynonymous missense mutations. Chi-square testing was used for comparative analyses. Results The most frequently mutated genes included PIK3CA (28.1%), KMT2D (19.5%), FBXW7 (12%), TP53 (12%) and PTEN (10.8%). The expression of PD-1 was seen in 68.8% and PD-L1 in 40.5% of tumors. High TMB was present in 6.7% of specimens. HER2 IHC was positive in 0.9%, amplification by chromogenic in situ hybridization (CISH) was seen 1.3%, and mutations in ERBB2 were present in 1.8% of tumors. The latter mutation has not been previously described in SCCA. When compared with TP53-WT tumors, TP53-MT tumors had higher rates of CDKN2A, EWSR1, JAK1, FGFR1 and BRAF mutations. PD-1 and PD-L1 expression were similar, and high TMB did not correlate with PD-1 (P=0.50) or PD-L1 (P=0.52) expression. Conclusions Molecular profiling differences between TP53-MT and TP53-WT SCCA indicate different carcinogenic pathways which may influence response to therapy. Low frequency mutations in several druggable genes may provide therapeutic opportunities for patients with SCCA.
Collapse
Affiliation(s)
- Samantha A Armstrong
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rita Malley
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Hongkun Wang
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, CA, USA
| | | | | | | | | | - Jimmy J Hwang
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Philip A Philip
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - John L Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Mohamed E Salem
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
8
|
Moniz CMV, Riechelmann RP, Oliveira SCR, Bariani GM, Rivelli TG, Ortega C, Pereira AAL, Meireles SI, Franco R, Chen A, Bonadio RC, Nahas C, Sabbaga J, Coudry RA, Braghiroli MI, Hoff PM. A Prospective Cohort Study of Biomarkers in Squamous Cell Carcinoma of the Anal Canal (SCCAC) and their Influence on Treatment Outcomes. J Cancer 2021; 12:7018-7025. [PMID: 34729104 PMCID: PMC8558650 DOI: 10.7150/jca.57678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Although Chemoradiation (CRT) is the curative treatment for SCCAC, many patients present primary resistance. Since it is a rare tumor, response predictors remain unknown. Methods: We performed a prospective cohort study to evaluate biomarkers associated with CRT response, progression-free survival (PFS), and overall survival (OS). The primary endpoint was response at 6 months (m). Tumor DNA and HPV were analyzed by next-generation sequencing, while KI-67 and PD-L1 by immunohistochemistry in tumor tissue. Results: Seventy-eight patients were recruited between October/2011 and December/2015, and 75 were response evaluable. The median age was 57 years, 65% (n=49) were stage III and 12% (n=9) were HIV positive (HIV+). At 6m, 62.7% (n=47) presented CR. On multivariate analyses, stage II patients were 4.7 more likely to achieve response than stage III (OR, 4.70; 95%CI, 1.36-16.30; p=0.015). HIV+ was associated with a worse response (OR, 5.72; 95%CI, 2.5-13.0; p<0.001). 5-year PFS and OS rates were 63.3% and 76.4%, respectively, with a median follow up of 66m. On multivariate analyses, older age (HR 1.06, p=0.022, 95%IC 1.01-1.11) and absence of CR at 6m (HR 3.36, p=0.007, 95%IC 1.39-8.09) were associated with inferior OS. The 5-year OS rate was 62.5% in HIV+ group compared to 78% among HIV- pts, although this difference was not statistically significant (p=0.4). PIK3CA, MET and TP53 mutations, HPV, Ki-67 expression, and PD-L1 expression, were not associated with PFS and OS. Conclusions: Clinical stage III and HIV+ were associated with worse response to CRT at 6m. The absence of CR was the main factor associated with poor 5-year OS.
Collapse
Affiliation(s)
- Camila Motta Venchiarutti Moniz
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | | | | | - Giovanni Mendonça Bariani
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Thomas Giollo Rivelli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Cintia Ortega
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | | | - Rejane Franco
- Universidade Federal do Paraná - Hospital de Clínicas, Curitiba, PR, Brasil
| | - Andre Chen
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Renata Colombo Bonadio
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Caio Nahas
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Jorge Sabbaga
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Maria Ignez Braghiroli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | - Paulo Marcelo Hoff
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| |
Collapse
|
9
|
Walcheck MT, Matkowskyj KA, Turco A, Blaine-Sauer S, Nukaya M, Noel J, Ronnekleiv OK, Ronnekleiv-Kelly SM. Sex-dependent development of Kras-induced anal squamous cell carcinoma in mice. PLoS One 2021; 16:e0259245. [PMID: 34735515 PMCID: PMC8568287 DOI: 10.1371/journal.pone.0259245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/12/2021] [Indexed: 11/18/2022] Open
Abstract
Anal squamous cell carcinoma (SCC) will be diagnosed in an estimated 9,080 adults in the United States this year, and rates have been rising over the last several decades. Most people that develop anal SCC have associated human papillomavirus (HPV) infection (~85-95%), with approximately 5-15% of anal SCC cases occurring in HPV-negative patients from unknown etiology. This study identified and characterized the Kras-driven, female sex hormone-dependent development of anal squamous cell carcinoma (SCC) in the LSL-KrasG12D; Pdx1-Cre (KC) mouse model that is not dependent on papillomavirus infection. One hundred percent of female KC mice develop anal SCC, while no male KC mice develop tumors. Both male and female KC anal tissue express Pdx1 and Cre-recombinase mRNA, and the activated mutant KrasG12D gene. Although the driver gene mutation KrasG12D is present in anus of both sexes, only female KC mice develop Kras-mutant induced anal SCC. To understand the sex-dependent differences, KC male mice were castrated and KC female mice were ovariectomized. Castrated KC males displayed an unchanged phenotype with no anal tumor formation. In contrast, ovariectomized KC females demonstrated a marked reduction in anal SCC development, with only 15% developing anal SCC. Finally, exogenous administration of estrogen rescued the tumor development in ovariectomized KC female mice and induced tumor development in castrated KC males. These results confirm that the anal SCC is estrogen mediated. The delineation of the role of female sex hormones in mediating mutant Kras to drive anal SCC pathogenesis highlights a subtype of anal SCC that is independent of papillomavirus infection. These findings may have clinical applicability for the papillomavirus-negative subset of anal SCC patients that typically respond poorly to standard of care chemoradiation.
Collapse
Affiliation(s)
- Morgan T. Walcheck
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Kristina A. Matkowskyj
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States of America
| | - Anne Turco
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Simon Blaine-Sauer
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Manabu Nukaya
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Jessica Noel
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Oline K. Ronnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Sciences University, Portland, OR, United States of America
| | - Sean M. Ronnekleiv-Kelly
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| |
Collapse
|
10
|
Sia J, Mou W, Agas RA, Xie J, Burns M, Varghayee N, Chu J, Heriot A, Leong T, Ngan SY. Long-term Patterns of Failure and the Value of Blood Prognostic Markers in Anal Cancers Treated With Intensity-Modulated Radiation Therapy. Clin Colorectal Cancer 2021; 21:e102-e112. [PMID: 34799240 DOI: 10.1016/j.clcc.2021.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND To analyze the long-term outcomes and prognostic value of hematological parameters in anal cancer patients receiving intensity-modulated radiation therapy (IMRT). MATERIALS Hospital records of consecutive patients with anal squamous cell carcinoma who received curative-intent IMRT according to a standardized contouring protocol between 2010 and 2020 were reviewed. Locoregional failure-free survival (LRFS), distant metastasis-free survival (DMFS), progression-free survival (PFS), and overall survival (OS) were estimated using the Kaplan-Meier method. Coverage of locoregional recurrences by the initial IMRT volumes were assessed. The prognostic value of pretreatment blood counts for PFS and OS were determined using Cox regression analysis. RESULTS A total of 166 patients were analyzed with a median follow-up of 3.3 years. Forty-six percent and 54% of patients had Stage I-II and IIIA-B cancers, respectively. The 5-year LRFS, DMFS, PFS and OS were 81%, 89%, 65% and 76% respectively. Grade ≥ 3 toxicity occurred in 5% of patients. Of all patients who relapsed, 70% had only locoregional recurrence as first site of failure. Ninety percent of locoregional recurrences were in-field. Hemoglobin, neutrophil and platelet counts were associated with PFS on univariable analysis, but only cancer stage and p16 status remained prognostic on multivariable analysis. Patients with more advanced cancer stages also had higher baseline neutrophil counts. Performance status and neutrophil counts were prognostic for OS on multivariable analysis. CONCLUSION This study affirms the long-term efficacy and safety of IMRT. Treatment resistance, rather than radiation geographic miss, is a major issue underpinning locoregional recurrences. Pretreatment blood counts were not validated to be independently prognostic for disease recurrence.
Collapse
Affiliation(s)
- Joseph Sia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir PeterMacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| | - Wei Mou
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir PeterMacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Ryan Anthony Agas
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jing Xie
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Mark Burns
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Nahal Varghayee
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Julie Chu
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Alexander Heriot
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir PeterMacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Trevor Leong
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir PeterMacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Samuel Y Ngan
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir PeterMacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
11
|
Guerra GR, Kong JC, Millen RM, Read M, Liu DS, Roth S, Sampurno S, Sia J, Bernardi MP, Chittleborough TJ, Behrenbruch CC, Teh J, Xu H, Haynes NM, Yu J, Lupat R, Hawkes D, Di Costanzo N, Tothill RW, Mitchell C, Ngan SY, Heriot AG, Ramsay RG, Phillips WA. Molecular and genomic characterisation of a panel of human anal cancer cell lines. Cell Death Dis 2021; 12:959. [PMID: 34663790 PMCID: PMC8523722 DOI: 10.1038/s41419-021-04141-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022]
Abstract
Anal cancer is a rare disease that has doubled in incidence over the last four decades. Current treatment and survival of patients with this disease has not changed substantially over this period of time, due, in part, to a paucity of preclinical models to assess new therapeutic options. To address this hiatus, we set-out to establish, validate and characterise a panel of human anal squamous cell carcinoma (ASCC) cell lines by employing an explant technique using fresh human ASCC tumour tissue. The panel of five human ASCC cell lines were validated to confirm their origin, squamous features and tumourigenicity, followed by molecular and genomic (whole-exome sequencing) characterisation. This panel recapitulates the genetic and molecular characteristics previously described in ASCC including phosphoinositide-3-kinase (PI3K) mutations in three of the human papillomavirus (HPV) positive lines and TP53 mutations in the HPV negative line. The cell lines demonstrate the ability to form tumouroids and retain their tumourigenic potential upon xenotransplantation, with varied inducible expression of major histocompatibility complex class I (MHC class I) and Programmed cell death ligand 1 (PD-L1). We observed differential responses to standard chemotherapy, radiotherapy and a PI3K specific molecular targeted agent in vitro, which correlated with the clinical response of the patient tumours from which they were derived. We anticipate this novel panel of human ASCC cell lines will form a valuable resource for future studies into the biology and therapeutics of this rare disease.
Collapse
Affiliation(s)
- Glen R Guerra
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Joseph C Kong
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rosemary M Millen
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew Read
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - David S Liu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- UGI Surgery Unit, Austin Hospital, 145 Studley Road, Heidelberg, Victoria, 3084, Australia
| | - Sara Roth
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Shienny Sampurno
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Joseph Sia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Maria-Pia Bernardi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Timothy J Chittleborough
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Corina C Behrenbruch
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jiasian Teh
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Huiling Xu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Nicole M Haynes
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jiaan Yu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Richard Lupat
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - David Hawkes
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- VCS Foundation, Carlton, VIC, 3053, Australia
- Department of Pathology, University of Malaya, Kuala Lumpur, Malaysia
| | - Natasha Di Costanzo
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Richard W Tothill
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Cancer Research, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Samuel Y Ngan
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Alexander G Heriot
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Robert G Ramsay
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wayne A Phillips
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
12
|
Guren MG, Sebag-Montefiore D, Franco P, Johnsson A, Segelov E, Deutsch E, Rao S, Spindler KLG, Arnold D. Treatment of Squamous Cell Carcinoma of the Anus, Unresolved Areas and Future Perspectives for Research: Perspectives of Research Needs in Anal Cancer. Clin Colorectal Cancer 2021; 20:279-287. [PMID: 34645589 DOI: 10.1016/j.clcc.2021.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023]
Abstract
Anal cancer is a relatively rare, mostly HPV-related cancer. The curative treatment consists of concurrent chemoradiation delivered with modern radiotherapy techniques. The prognosis for most patients with early localized disease is very favourable; however patients with locally advanced disease and/or HPV negative tumours are at higher risk of locoregional and distant treatment failure. Tailored approaches are presently being investigated to determine the most suitable regimen in terms of radiotherapy dose prescription, target volume selection, normal tissue avoidance, and combination therapy. Metastatic anal cancer is treated with chemotherapy aiming at prolonged survival. The role of immune therapy in the clinical setting is being investigated. There is little knowledge on the biology of anal cancer, and an urgent need for more clinical and translational research dedicated to this disease. In this article, the evidence-base for the current treatment is briefly reviewed, and perspectives on future research needs are high-lighted.
Collapse
Affiliation(s)
| | | | - Pierfrancesco Franco
- Department of Translational Medicine, University of Eastern Piedmont and Department of Radiation Oncology, AOU ''Maggiore della Carità,'' Novara, Italy
| | - Anders Johnsson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Eva Segelov
- School of Clinical Sciences, Faculty of Medicine, Monash University, Clayton, Australia and Department of Oncology, Monash Health Clayton, Australia
| | | | - Sheela Rao
- GI Unit, Royal Marsden Hospital, London, UK
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| |
Collapse
|
13
|
Zhang X, Lu D, Szporn AH, Zakowski MF, Si Q. A comparative study of the genotype profiles of high-risk human papillomavirus infection in male and female HIV-positive patients and their correlation with anal cytology and biopsy. J Am Soc Cytopathol 2021; 11:21-30. [PMID: 34625384 DOI: 10.1016/j.jasc.2021.08.005] [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: 06/10/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Although anal cancer is more common in women, most of the studies on the role of high-risk human papillomavirus (hrHPV) infection in anal squamous lesions have focused on high-risk male patients. Therefore, we compared the genotype profile and clinicopathologic correlation of hrHPV infection in human immunodeficiency virus-positive (HIV+) men and women. MATERIALS AND METHODS We retrospectively analyzed 2254 HIV+ patients (1931 men and 323 women) who had undergone anal Papanicolaou tests at our institution; 1189 of them also had follow-up biopsy data available. HPV genotyping was performed using the Roche Cobas system and correlated with the cytologic and histologic diagnosis. RESULTS Compared with the HIV+ men, the HIV+ women had a significantly lower rate of hrHPV infection (67.5% versus 78.5%; P < 0.0001) but a significantly higher rate of high-grade squamous intraepithelial lesions (HSILs) on anal Papanicolaou tests (4.6% versus 2.5%; P < 0.05). Other high-risk HPV (ohrHPV), as a group, is much more common than HPV16 or HPV18 in both genders. HIV+ women had significantly lower HPV16 and ohrHPV infection rates than did HIV+ men. However, the HPV18 infection rates were similar between HIV+ women and HIV+ men. For both genders, the rates of HSILs or high-grade anal intraepithelial neoplasia (AIN2-3) were significantly increased when coinfection of ohrHPV with either HPV16 or HPV18 was present. CONCLUSIONS Although both HIV+ men and HIV+ women have an increased risk of hrHPV infection, HIV+ women have different hrHPV genotype profiles and higher rates of high-grade lesions. Coinfection with different genotypes of hrHPV can significantly increase the risk of HSILs or AIN2-3 in both genders and could requires vigilant clinical and laboratory follow-up.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin at Madison, Madison, Wisconsin
| | - Dan Lu
- Department of Pathology, Molecular, and Cell-Based Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arnold H Szporn
- Department of Pathology, Molecular, and Cell-Based Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Maureen F Zakowski
- Department of Pathology, Molecular, and Cell-Based Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Qiusheng Si
- Department of Pathology, Molecular, and Cell-Based Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
14
|
Spehner L, Boustani J, Cabel L, Doyen J, Vienot A, Borg C, Kim S. Present and Future Research on Anal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:3895. [PMID: 34359795 PMCID: PMC8345786 DOI: 10.3390/cancers13153895] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Squamous cell carcinoma of the anus is an orphan disease, and after more than three decades of no substantial advances in disease knowledge and treatment, it is finally gaining momentum with the arrival of a taxane-based chemotherapy and immunotherapy. Currently, about 20 combination clinical trials with an anti-PD1/L1 are ongoing in localized and advanced stages, in association with radiotherapy, chemotherapy, tumor vaccines, anti-CTLA4, anti-EGFR, or antiangiogenic molecules. Moreover, a new biomarker with high sensitivity and specificity such as HPV circulating tumor DNA (HPV ctDNA) by liquid biopsy, is improving not only the prognostic measurement but also the treatment strategy guidance for this disease. Finally, better understanding of potential targets is reshaping the present and future clinical research in this unique, HPV genotype-16-related disease in the great majority of patients.
Collapse
Affiliation(s)
- Laurie Spehner
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
| | - Jihane Boustani
- Department of Radiotherapy, University Hospital of Besançon, 25030 Besançon, France;
| | - Luc Cabel
- Department of Medical Oncology, Curie Institute, 75005 Paris, France;
| | - Jérôme Doyen
- Department of Medical Oncology, Centre Antoine-Lacassagne, 06189 Nice, France;
| | - Angélique Vienot
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
| | - Christophe Borg
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
| | - Stefano Kim
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
- Department of Oncology and Radiotherapy, Nord Franche Comté Hospital, 25209 Montbéliard, France
| |
Collapse
|
15
|
Wang GH, Ni K, Gu C, Huang J, Chen J, Wang XD, Ni Q. EphA8 inhibits cell apoptosis via AKT signaling and is associated with poor prognosis in breast cancer. Oncol Rep 2021; 46:183. [PMID: 34278497 DOI: 10.3892/or.2021.8134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/11/2021] [Indexed: 11/06/2022] Open
Abstract
Erythropoietin‑producing hepatocellular receptors (Ephs) comprise the largest subfamily of receptor tyrosine kinases and have been reported to be involved in a variety of biological cellular processes, including tumorigenesis and cancer progression. The present study aimed to determine the expression levels and clinicopathological significance of EphA8 in breast cancer (BC) using immunohistochemistry analysis of tissue microarrays. The results of the present study revealed that EphA8 expression levels were upregulated in BC tissue and were associated with tumor size and TNM stage. In addition, upregulated expression levels of EphA8 were identified to be a poor prognostic biomarker for patients with BC. The knockdown of EphA8 expression using short hairpin RNA resulted in increased levels of apoptosis as well as decreased proliferation, migration and invasion of BC cells both in vivo and in vitro. The knockdown of EphA8 also decreased the phosphorylation of AKT, which was accompanied by downregulation of Bcl‑2 expression levels and upregulation of p53, Caspase‑3 and Bax expression levels. Moreover, knockdown of EphA8 expression increased the chemosensitivity of BC cells to paclitaxel. In conclusion, the results of the present study indicated that EphA8 may be a useful prognostic marker in BC and that knockdown of EphA8 may represent a novel strategy in adjuvant chemotherapy for the treatment of BC.
Collapse
Affiliation(s)
- Gui-Hua Wang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Kan Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Changjiang Gu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianfei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jing Chen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xu-Dong Wang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qichao Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
16
|
|
17
|
Aldersley J, Lorenz DR, Mouw KW, D'Andrea AD, Gabuzda D. Genomic Landscape of Primary and Recurrent Anal Squamous Cell Carcinomas in Relation to HPV Integration, Copy-Number Variation, and DNA Damage Response Genes. Mol Cancer Res 2021; 19:1308-1321. [PMID: 33883185 DOI: 10.1158/1541-7786.mcr-20-0884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/02/2021] [Accepted: 04/16/2021] [Indexed: 12/26/2022]
Abstract
The incidence of anal squamous cell carcinoma (ASCC) has been increasing, particularly in populations with HIV. Human papillomavirus (HPV) is the causal factor in 85% to 90% of ASCCs, but few studies evaluated HPV genotypes and integrations in relation to genomic alterations in ASCC. Using whole-exome sequence data for primary (n = 56) and recurrent (n = 31) ASCC from 72 patients, we detected HPV DNA in 87.5% of ASCC, of which HPV-16, HPV-18, and HPV-6 were detected in 56%, 22%, and 33% of HIV-positive (n = 9) compared with 83%, 3.2%, and 1.6% of HIV-negative cases (n = 63), respectively. Recurrent copy-number variations (CNV) involving genes with documented roles in cancer included amplification of PI3KCA and deletion of APC in primary and recurrent tumors; amplifications of CCND1, MYC, and NOTCH1 and deletions of BRCA2 and RB1 in primary tumors; and deletions of ATR, FANCD2, and FHIT in recurrent tumors. DNA damage response genes were enriched among recurrently deleted genes in recurrent ASCCs (P = 0.001). HPV integrations were detected in 29 of 76 (38%) ASCCs and were more frequent in stage III-IV versus stage I-II tumors. HPV integrations were detected near MYC and CCND1 amplifications and recurrent targets included NFI and MUC genes. These results suggest HPV genotypes in ASCC differ by HIV status, HPV integration is associated with ASCC progression, and DNA damage response genes are commonly disrupted in recurrent ASCCs. IMPLICATIONS: These data provide the largest whole-exome sequencing study of the ASCC genomic landscape to date and identify HPV genotypes, integrations, and recurrent CNVs in primary or recurrent ASCCs.
Collapse
Affiliation(s)
- Jordan Aldersley
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - David R Lorenz
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Dana Gabuzda
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
18
|
Yan L, Chen H, Tang L, Jiang P, Yan F. Super-enhancer-associated long noncoding RNA AC005592.2 promotes tumor progression by regulating OLFM4 in colorectal cancer. BMC Cancer 2021; 21:187. [PMID: 33622275 PMCID: PMC7903608 DOI: 10.1186/s12885-021-07900-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/11/2021] [Indexed: 11/26/2022] Open
Abstract
Background Super-enhancer-associated long noncoding RNAs (SE-lncRNAs) have been reported to play essential roles in tumorigenesis, but the fundamental mechanism of SE-lncRNAs in colorectal cancer (CRC) remains largely unknown. Methods A microarray was performed to identify the differentially expressed SE-lncRNAs between CRC tissues and peritumoral tissues. A novel SE-lncRNA, AC005592.2, was selected from these differentially expressed SE-lncRNAs to explore its effects on CRC development. Fluorescence quantitative real-time PCR (qRT-PCR) was used to assay the expression of AC005592.2 in CRC tissues and cell lines. Functional assays were applied to identify the biological effects of AC005592.2 in CRC cells. Furthermore, RNA-seq was employed to predict potential targets of AC005592.2. Results AC005592.2 was significantly increased in CRC tissues and cells. High expression of AC005592.2 was significantly associated with TNM stage and tumor differentiation in CRC patients. Knockdown of AC005592.2 suppressed CRC cell proliferation, invasion and migration but promoted apoptosis, while AC005592.2 overexpression exerted the opposite effects on CRC cells. In addition, AC005592.2 positively regulated the expression of olfactomedin 4 (OLFM4), which was also upregulated in CRC tissues. Conclusion The findings suggested that AC005592.2 is a crucial promoter of CRC progression and may serve as an attractive therapeutic target for CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07900-x.
Collapse
Affiliation(s)
- Linping Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, China
| | - Huanhuan Chen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, China
| | - Li Tang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, China
| | - Pan Jiang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, China
| | - Feng Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, China.
| |
Collapse
|
19
|
Anal dysplasia and anal cancer. English version. Hautarzt 2020; 71:74-81. [PMID: 32303769 DOI: 10.1007/s00105-020-04562-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Anal intraepithelial neoplasia (AIN) and 89-100% of anal cancers are caused by persistent infections with high-risk (HR) human papillomaviruses (HPV). In HIV-positive patients, anal HPV infection and AIN are very common and these patients have a significantly increased risk for anal cancer. However, a continuous increase in the incidence of anal cancer has also been observed in the general population in recent decades. AIN can clinically present in diverse manners. In HIV-positive patients AIN can be hidden in condylomas. Approximately 3-14% of high-grade AIN progress to anal cancer within 5 years. Therefore, screening examinations should be offered to patients with an increased risk for anal cancer. The treatment options for AIN are similar to those for condylomas. HIV-positive patients with controlled immune status and HIV-negative patients with anal cancer respond comparably well to combined radiochemotherapy. A German-language S3 guideline for anal cancer will be available in 2020. In HIV-positive patients over 26 years of age, HPV vaccination showed no effect in a controlled phase‑3 study. To prevent AIN and anal cancer in the future, HPV vaccination rates need to be increased in HPV-naïve girls and boys.
Collapse
|
20
|
Williams EA, Montesion M, Sharaf R, Corines J, Patel PJ, Gillespie BJ, Pavlick DC, Sokol ES, Alexander BM, Williams KJ, Elvin JA, Ross JS, Ramkissoon SH, Hemmerich AC, Tse JY, Mochel MC. CYLD-mutant cylindroma-like basaloid carcinoma of the anus: a genetically and morphologically distinct class of HPV-related anal carcinoma. Mod Pathol 2020; 33:2614-2625. [PMID: 32461623 PMCID: PMC7685972 DOI: 10.1038/s41379-020-0584-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 11/12/2022]
Abstract
Rare reports of anal carcinoma (AC) describe histologic resemblance to cutaneous cylindroma, but mutations in the tumor suppressor CYLD, the gene responsible for familial and sporadic cylindromas, have not been systematically investigated in AC. Here, we investigate CYLD-mutant AC, focusing on molecular correlates of distinct histopathology. Comprehensive genomic profiling (hybrid-capture-based DNA sequencing) was performed on 574 ACs, of which 75 unique cases (13%) harbored a CYLD mutation. Clinical data, pathology reports, and histopathology were reviewed for each CYLD-mutant case. The spectrum of CYLD mutations included truncating (n = 50; 67%), homozygous deletion (n = 10; 13%), missense (n = 16; 21%), and splice-site (n = 3; 4%) events. Compared with CYLD-wildtype AC (n = 499), CYLD-mutant ACs were significantly enriched for females (88% vs. 67%, p = 0.0001), slightly younger (median age 59 vs. 61 years, p = 0.047), and included near-universal detection of high-risk HPV sequences (97% vs. 88%, p = 0.014), predominantly HPV16 (96%). The CYLD-mutant cohort also showed significantly lower tumor mutational burden (TMB; median 2.6 vs. 5.2 mut/Mb, p < 0.00001) and less frequent alterations in PIK3CA (13% vs. 31%, p = 0.0015). On histopathologic examination, 73% of CYLD-mutant AC (55/75 cases) showed a striking cylindroma-like histomorphology, composed of aggregates of basaloid cells surrounded by thickened basement membranes and containing characteristic hyaline globules, while only 8% of CYLD-wildtype tumors (n = 34/409) contained cylindroma-like hyaline globules (p < 0.0001). CYLD-mutant carcinomas with cylindroma-like histomorphology (n = 55) showed significantly lower TMB compared with CYLD-mutant cases showing basaloid histology without the distinctive hyaline globules (n = 14) (median 1.7 vs. 4.4 mut/Mb, p = 0.0058). Only five CYLD-mutant cases (7%) showed nonbasaloid conventional squamous cell carcinoma histology (median TMB = 5.2 mut/Mb), and a single CYLD-mutant case showed transitional cell carcinoma-like histology. Within our cohort of ACs, CYLD mutations characterize a surprisingly large subset (13%), with distinct clinical and genomic features and, predominantly, a striking cylindroma-like histopathology, representing a genotype-phenotype correlation which may assist in classification of AC.
Collapse
Affiliation(s)
- Erik A Williams
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA.
| | - Meagan Montesion
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - Radwa Sharaf
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - James Corines
- Department of Pathology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Parth J Patel
- Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | | | - Dean C Pavlick
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - Ethan S Sokol
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - Brian M Alexander
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - Kevin Jon Williams
- Department of Physiology and Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Julia A Elvin
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
| | - Jeffrey S Ross
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
- Department of Pathology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Shakti H Ramkissoon
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | | | - Julie Y Tse
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA, 02141, USA
- Department of Pathology & Laboratory Medicine, Tufts University School of Medicine, 145 Harrison Ave, Boston, MA, 02111, USA
| | - Mark C Mochel
- Departments of Pathology and Dermatology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| |
Collapse
|
21
|
O'Brien SJ, Ellis CT, McDowell J, Galandiuk S, Polk HC. Anal squamous cell carcinoma incidentally found at hemorrhoidectomy. Surgery 2020; 169:610-616. [PMID: 33004218 DOI: 10.1016/j.surg.2020.08.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/24/2020] [Accepted: 08/18/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anal squamous cell carcinoma incidence is increasing nationally and, more so, in Kentucky. Squamous cell carcinoma of the anus unexpectedly identified at hemorrhoidectomy pathologic evaluation is not uncommon. We hypothesized this is occurring more frequently and sought to evaluate its impact on outcomes. METHODS The Kentucky Cancer Registry, a premier population database, was queried for all squamous cell carcinoma of the anus cases between 2007 and 2016. Hemorrhoidal squamous cell carcinoma of the anus patients were compared with nonhemorrhoidal squamous cell carcinomas of the anus. Patient demographics, treatments, and outcomes were analyzed. RESULTS Of the 722 squamous cell carcinoma of the anus cases identified, 3.05% (n = 22) were within hemorrhoidectomy specimens. Demographics were similar between hemorrhoidal squamous cell carcinoma of the anus versus nonhemorrhoidal squamous cell carcinoma of the anus. Chemoradiation was the most common treatment strategy among all patients, and there were similar rates of disease, persistence, recurrence, and survival between hemorrhoidal and nonhemorrhoidal squamous cell carcinoma. Stage I disease was more common in the hemorrhoid group compared with the nonhemorrhoid group (63% vs 27%, P < .01). CONCLUSION Hemorrhoidal squamous cell carcinoma of the anus comprised 3.05% of our population-based cohort. Hemorrhoidal squamous cell carcinomas of the anus were more likely to receive chemoradiation compared with local excision, but there were similar oncologic outcomes. We postulate that some individuals may receive overtreatment with chemoradiation owing to imprecise labeling of hemorrhoid specimens. For this reason, we advocate for separate submission of each hemorrhoid specimen.
Collapse
Affiliation(s)
- Stephen J O'Brien
- Price Institute of Surgical Research, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - C Tyler Ellis
- Price Institute of Surgical Research, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Jaclyn McDowell
- Markey Cancer Control Program, Kentucky Cancer Registry, Lexington, KY
| | - Susan Galandiuk
- Price Institute of Surgical Research, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Hiram C Polk
- Price Institute of Surgical Research, Department of Surgery, University of Louisville School of Medicine, Louisville, KY.
| |
Collapse
|
22
|
Carter TJ, Jeyaneethi J, Kumar J, Karteris E, Glynne-Jones R, Hall M. Identification of Cancer-Associated Circulating Cells in Anal Cancer Patients. Cancers (Basel) 2020; 12:cancers12082229. [PMID: 32785154 PMCID: PMC7465059 DOI: 10.3390/cancers12082229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/26/2022] Open
Abstract
Whilst anal cancer accounts for less than 1% of all new cancer cases, incidence rates have increased by up to 70% in the last 30 years with the majority of cases driven by human papilloma virus (HPV) infection. Standard treatment for localised anal cancer is chemoradiotherapy (CRT). Localised progression is the predominant pattern of relapse but well under 50% of cases are salvaged by surgery, predominantly because confirming recurrence within post-radiation change is very challenging. Identifying cancer-associated circulating cells (CCs) in peripheral blood could offer a corroborative method of monitoring treatment efficacy and identifying relapse early. To study this, nucleated cells were isolated from the blood of patients with anal cancer prior to, during, and after CRT and processed through the Amnis® ImageStream®X Mk II Imaging Flow Cytometer, without prior enrichment, using Pan-cytokeratin (PCK), CD45 antibodies and making use of the DNA dye DRAQ5. Analysis was undertaken using IDEAS software to identify those cells that were PCK-positive and DRAQ5-positive as well as CD45-negative; these were designated as CCs. CCs were identified in 7 of 8 patients; range 60-876 cells per mL of blood. This first report of the successful identification of CCs in anal cancer patients raises the possibility that liquid biopsies will find a future role as a prognostic/diagnostic tool in this patient group.
Collapse
Affiliation(s)
- Thomas J. Carter
- Mount Vernon Cancer Centre, Middlesex HA6 2RN, UK; (T.J.C.); (R.G.-J.)
| | - Jeyarooban Jeyaneethi
- Department of Life Sciences, Brunel University, London UB83PH, UK; (J.J.); (J.K.); (E.K.)
| | - Juhi Kumar
- Department of Life Sciences, Brunel University, London UB83PH, UK; (J.J.); (J.K.); (E.K.)
| | - Emmanouil Karteris
- Department of Life Sciences, Brunel University, London UB83PH, UK; (J.J.); (J.K.); (E.K.)
| | - Rob Glynne-Jones
- Mount Vernon Cancer Centre, Middlesex HA6 2RN, UK; (T.J.C.); (R.G.-J.)
- Department of Life Sciences, Brunel University, London UB83PH, UK; (J.J.); (J.K.); (E.K.)
| | - Marcia Hall
- Mount Vernon Cancer Centre, Middlesex HA6 2RN, UK; (T.J.C.); (R.G.-J.)
- Department of Life Sciences, Brunel University, London UB83PH, UK; (J.J.); (J.K.); (E.K.)
- Correspondence:
| |
Collapse
|
23
|
Knight K, Choong JX, McKee RF, Anderson JH, Horgan PG, McMillan DC, McDonald A, Roxburgh CS. The Influence of Systemic Inflammation on Treatment Response and Survival in Anal Squamous Cell Cancer. Clin Oncol (R Coll Radiol) 2020; 33:e22-e30. [PMID: 32709540 DOI: 10.1016/j.clon.2020.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023]
Abstract
AIMS The incidence of anal squamous cell cancer (SCCA) is rising. Although chemoradiotherapy (CRT) provides a chance of cure, a proportion of patients have an incomplete response or develop recurrence. This study assessed the value of inflammation-based prognostic indicators, including the modified Glasgow Prognostic Score (mGPS) and neutrophil:lymphocyte ratio (NLR), in patients with SCCA treated by CRT with curative intent. MATERIAL AND METHODS Patients with histologically confirmed SCCA were identified from pathology records. Medical records were retrospectively reviewed and clinical, pathological and treatment characteristics were abstracted. The mGPS (0 = normal C-reactive protein [CRP] and albumin, 1 = CRP >10 mg/l and 2 = CRP >10 mg/l and albumin <35 mg/l) and NLR were calculated from routine blood tests obtained prior to CRT. RESULTS In total, 118 patients underwent CRT for SCCA between December 2007 and February 2018. Of these, 99 patients had appropriate pretreatment blood results available. Systemic inflammation as indicated by NLR >3 and mGPS >0 was present in 41% and 39% of patients, respectively. Most patients had T2 or larger tumours (n = 85, 86%) without nodal involvement (n = 64, 65%). An elevated mGPS was associated with more advanced T-stage (56% versus 35%, P = 0.036). NLR >5 was associated with nodal positivity (56% versus 31%, P = 0.047). On multivariate analysis, more advanced T-stage (odds ratio 7.49, 95% confidence interval 1.51-37.20, P = 0.014) and a raised mGPS (odds ratio 5.13, 95% confidence interval 1.25-21.14, P = 0.024) were independently related to incomplete CRT response. An elevated mGPS was prognostic of inferior survival (hazard ratio 3.09, 95% confidence interval 1.47-6.50, P = 0.003) and cancer-specific survival (hazard ratio 4.32, 95% confidence interval 1.54-12.15, P = 0.006), independent of TNM stage. CONCLUSION Systemic inflammation, as measured by the mGPS, is associated with an incomplete CRT response and is independently prognostic of inferior survival in patients with SCCA. The mGPS may offer a simple marker of inferior outcome that could be used to identify high-risk patients.
Collapse
Affiliation(s)
- K Knight
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK.
| | - J X Choong
- School of Medicine, University of Glasgow, Glasgow, UK
| | - R F McKee
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | - J H Anderson
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | - P G Horgan
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | - D C McMillan
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| | - A McDonald
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - C S Roxburgh
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
24
|
Luchini C, Bibeau F, Ligtenberg MJL, Singh N, Nottegar A, Bosse T, Miller R, Riaz N, Douillard JY, Andre F, Scarpa A. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol 2020; 30:1232-1243. [PMID: 31056702 DOI: 10.1093/annonc/mdz116] [Citation(s) in RCA: 591] [Impact Index Per Article: 147.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancers with a defective DNA mismatch repair (dMMR) system contain thousands of mutations most frequently located in monomorphic microsatellites and are thereby defined as having microsatellite instability (MSI). Therefore, MSI is a marker of dMMR. MSI/dMMR can be identified using immunohistochemistry to detect loss of MMR proteins and/or molecular tests to show microsatellite alterations. Together with tumour mutational burden (TMB) and PD-1/PD-L1 expression, it plays a role as a predictive biomarker for immunotherapy. METHODS To define best practices to implement the detection of dMMR tumours in clinical practice, the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project, based on a systematic review-approach, to generate consensus recommendations on the: (i) definitions related to the concept of MSI/dMMR; (ii) methods of MSI/dMMR testing and (iii) relationships between MSI, TMB and PD-1/PD-L1 expression. RESULTS The MSI-related definitions, for which a consensus frame-work was used to establish definitions, included: 'microsatellites', 'MSI', 'DNA mismatch repair' and 'features of MSI tumour'. This consensus also provides recommendations on MSI testing; immunohistochemistry for the mismatch repair proteins MLH1, MSH2, MSH6 and PMS2 represents the first action to assess MSI/dMMR (consensus with strong agreement); the second method of MSI/dMMR testing is represented by polymerase chain reaction (PCR)-based assessment of microsatellite alterations using five microsatellite markers including at least BAT-25 and BAT-26 (strong agreement). Next-generation sequencing, coupling MSI and TMB analysis, may represent a decisive tool for selecting patients for immunotherapy, for common or rare cancers not belonging to the spectrum of Lynch syndrome (very strong agreement). The relationships between MSI, TMB and PD-1/PD-L1 expression are complex, and differ according to tumour types. CONCLUSIONS This ESMO initiative is a response to the urgent questions raised by the growing success of immunotherapy and provides also important insights on the relationships between MSI, TMB and PD-1/PD-L1.
Collapse
Affiliation(s)
- C Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - F Bibeau
- Department of Pathology, Caen University Hospital, Caen, France
| | - M J L Ligtenberg
- Departments of Human Genetics Radboud university medical center, Nijmegen, The Netherlands; Departments of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - N Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - A Nottegar
- Department of Surgery, San Bortolo Hospital, Vicenza, Italy
| | - T Bosse
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - R Miller
- Department of Oncology, University College London, London, UK
| | - N Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J-Y Douillard
- European Society for Medical Oncology, Lugano, Switzerland
| | - F Andre
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France.
| | - A Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
25
|
Wieland U, Oellig F, Kreuter A. [Anal dysplasia and anal cancer]. Hautarzt 2020; 71:284-292. [PMID: 32065247 DOI: 10.1007/s00105-020-04548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Anal intraepithelial neoplasia (AIN) and 89-100% of anal cancers are caused by persistent infections with high-risk (HR) human papillomaviruses (HPV). In HIV-positive patients, anal HPV infection and AIN are very common and these patients have a greatly increased risk of developing anal cancer. However, a continuous increase in the incidence of anal cancer has also been observed in the general population in recent decades. AIN can clinically present in diverse manners. In HIV-positive patients AIN can be hidden in condylomas. Furthermore, 3-14% of high-grade AIN progress to anal cancer within 5 years. Therefore, screening examinations should be offered to patients with an increased risk for anal cancer. The treatment options for AIN are similar to those for condylomas. HIV-positive patients with controlled immune status and HIV-negative patients with anal cancer respond comparably well to combined radiochemotherapy. A German-language AWMF S3 guideline for anal cancer will be available in 2020. In HIV-positive patients over 26 years of age, HPV vaccination showed no effect in a controlled phase‑3 study. To prevent AIN and anal cancer in the future, HPV vaccination rates need to be increased in HPV-naïve girls and boys.
Collapse
Affiliation(s)
- Ulrike Wieland
- Institut für Virologie, Nationales Referenzzentrum für Papillom- und Polyomaviren, Universität zu Köln, Köln, Deutschland
| | - Frank Oellig
- Institut für Pathologie, Mühlheim an der Ruhr, Deutschland
| | - Alexander Kreuter
- Klinik für Dermatologie, Venerologie und Allergologie, HELIOS St. Elisabeth Klinik Oberhausen, Universität Witten/Herdecke, Josefstr. 3, 46045, Oberhausen, Deutschland.
| |
Collapse
|
26
|
Aigner F, Werner RN, Siegel R. Diagnostik und Therapie des Analkarzinoms. COLOPROCTOLOGY 2020. [DOI: 10.1007/s00053-020-00436-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Heriot AG. Whither Anal Cancer? Br J Cancer 2020; 122:733-734. [PMID: 31932753 PMCID: PMC7078260 DOI: 10.1038/s41416-019-0690-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 11/16/2022] Open
Abstract
Improving outcomes for uncommon tumours such as anal cancer can be challenging. Evolution of care tends to be incremental rather than transformative and identifying the specific factors facilitating improvement can be difficult. Outcome from specialist units may be better than that demonstrated in trials, posing challenges for development of future trials.
Collapse
Affiliation(s)
- Alexander G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
28
|
Walhart T, Isaacson-Wechsler E, Ang KH, Arkin M, Tugizov S, Palefsky JM. A Cell-Based Renilla Luminescence Reporter Plasmid Assay for High-Throughput Screening to Identify Novel FDA-Approved Drug Inhibitors of HPV-16 Infection. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2020; 25:79-86. [PMID: 31361520 PMCID: PMC6925341 DOI: 10.1177/2472555219860771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Like cervical cancer, anal cancer is caused by human papillomavirus (HPV). HPV is the most common sexually transmitted agent and is found in the anal canal of almost all HIV-positive men who have sex with men (MSM). Rates of HPV anal cancer are disproportionately higher in this population. Although the nanovalent HPV vaccine is efficacious in protecting against oncogenic HPV types, a substantial proportion of MSM remains unvaccinated and anal HPV infection continues to be an important public health burden. Therefore, it is important to identify strategies to prevent HPV infection. We report on two promising and interlinked strategies: (1) the development of a cell-based Renilla luminescence reporter assay using HPV-16 pseudovirions that encapsidate SV40-driven Renilla luminescence reporter expression plasmid and (2) use of this assay for high-throughput screening (HTS) of FDA- and internationally approved drugs to identify those that could be repurposed to prevent HPV infection. We conducted a screen of 1906 drugs. The assay was valid with a Z' of 0.67 ± 0.04, percent coefficient of variance of 10.0, and signal-to-background noise window of 424.0 ± 8.0. Five drugs were chosen for further analyses based on selection parameters of ≥77.0% infection of HPV-16 pseudovirion-driven Renilla expression with <20.0% cytotoxicity. Of these, the antifungal pentamidine and a gamma-amino butyric acid receptor agonist securinine exhibited ≥90.0% infection with <10.0% cytotoxicity. This luminescent cell-based reporter expression plasmid assay for HTS is a valid method to identify FDA- and internationally approved drugs with the potential to be repurposed into prevention modalities for HPV infection.
Collapse
Affiliation(s)
- Tara Walhart
- Department of Community Health Systems, School of Nursing, University of California, San Francisco, CA, USA
- Department of Infectious Disease, Palefsky Laboratory, School of Medicine, University of California, San Francisco, CA, USA
| | - Erin Isaacson-Wechsler
- Department of Infectious Disease, Palefsky Laboratory, School of Medicine, University of California, San Francisco, CA, USA
| | - Kean-Hooi Ang
- Department of Pharmaceutical Chemistry, Small Molecule Discovery Center, University of California, San Francisco, CA, USA
| | - Michelle Arkin
- Department of Pharmaceutical Chemistry, Small Molecule Discovery Center, University of California, San Francisco, CA, USA
| | - Sharof Tugizov
- Department of Infectious Disease, Palefsky Laboratory, School of Medicine, University of California, San Francisco, CA, USA
| | - Joel M. Palefsky
- Department of Infectious Disease, Palefsky Laboratory, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
29
|
Abstract
Anal squamous cell carcinoma (SCCA), among other malignancies, is associated with the human papillomavirus (HPV) and its incidence continues to rise. Anal SCCA will likely remain an existing healthcare concern given compliance issues with the HPV vaccination seen in the US. Localized disease is predominantly treated with standard of care (SOC) definitive chemoradiation that has remained unchanged for decades. Clinical and molecular prognostic factors have emerged to characterize patients unresponsive to SOC, revealing the need for an alternate approach. Metastatic disease is an extremely small subset and understudied population due to its rarity. Recent prospective trials and mutational analysis have opened treatment options for this subset in need. Our review details the pharmacotherapeutic treatment in localized and metastatic anal SCCA chronologically, while also describing future outlooks.
Collapse
Affiliation(s)
- Jane E Rogers
- Pharmacy Clinical Programs, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Rising Incidence and Improved Survival of Anal Squamous Cell Carcinoma in Norway, 1987-2016. Clin Colorectal Cancer 2019; 18:e96-e103. [DOI: 10.1016/j.clcc.2018.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/29/2023]
|
31
|
Cacheux W, Lièvre A, Richon S, Vacher S, El Alam E, Briaux A, El Botty R, Mariani P, Buecher B, Schnitzler A, Barbazan J, Roman-Roman S, Bièche I, Dangles-Marie V. Interaction between IGF2-PI3K axis and cancer-associated-fibroblasts promotes anal squamous carcinogenesis. Int J Cancer 2019; 145:1852-1859. [DOI: 10.1002/ijc.32178] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/03/2018] [Accepted: 01/28/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Wulfran Cacheux
- Department of Medical Oncology; Hôpital Privé Pays de Savoie; Annemasse France
- Department of Medical Oncology; Institut Curie, PSL Research University; Saint-Cloud France
- Faculty of Health Sciences; Versailles Saint-Quentin-en-Yvelines University; Versailles France
| | - Astrid Lièvre
- Department of Medical Oncology; Institut Curie, PSL Research University; Saint-Cloud France
| | - Sophie Richon
- Department of Translational Research; Institut Curie, PSL Research University; Paris France
- UMR 144; Institut Curie, PSL Research University, CNRS; Paris France
| | - Sophie Vacher
- Department of Genetics, Pharmacogenomics Unit; Institut Curie, PSL Research University; Paris France
| | - Elsy El Alam
- Department of Tumour Biology; Institut Curie, PSL Research University; Saint-Cloud France
| | - Adrien Briaux
- Department of Genetics, Pharmacogenomics Unit; Institut Curie, PSL Research University; Paris France
| | - Rania El Botty
- Department of Translational Research; Institut Curie, PSL Research University; Paris France
| | - Pascale Mariani
- Department of Surgical Oncology; Institut Curie, PSL Research University; Paris France
| | - Bruno Buecher
- Department of Medical Oncology; Institut Curie, PSL Research University; Saint-Cloud France
| | - Anne Schnitzler
- Department of Genetics, Pharmacogenomics Unit; Institut Curie, PSL Research University; Paris France
| | - Jorge Barbazan
- UMR 144; Institut Curie, PSL Research University, CNRS; Paris France
| | - Sergio Roman-Roman
- Department of Translational Research; Institut Curie, PSL Research University; Paris France
| | - Ivan Bièche
- Department of Genetics, Pharmacogenomics Unit; Institut Curie, PSL Research University; Paris France
| | - Virginie Dangles-Marie
- Department of Translational Research; Institut Curie, PSL Research University; Paris France
- Faculty of Pharmacy; Université Paris Descartes; Paris France
| |
Collapse
|
32
|
Eng C, Messick C, Glynne-Jones R. The Management and Prevention of Anal Squamous Cell Carcinoma. Am Soc Clin Oncol Educ Book 2019; 39:216-225. [PMID: 31099616 DOI: 10.1200/edbk_237433] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Our aim is to discuss the current established management of care and associated prevention strategies of anal squamous cell carcinoma (SCCA). In general, the development of SCCA is commonly linked to a prior history of HPV. Unfortunately, HPV vaccination continues to be underutilized in the United States versus other countries. Increased acknowledgment of the importance of HPV vaccination as an anticancer vaccine should be encouraged. The present standard of care is primary chemoradiotherapy (CRT), which results in a high level of disease control for small, early-stage SCCA. More advanced cancers still fare poorly with this treatment, and the disease relapses locoregionally in the majority of cases (30%-50% of patients), resulting in an abdominoperineal resection. Current treatment recommendations are associated with substantial morbidity; alternative radiation doses and/or novel combinations of agents with CRT are needed to improve quality of life and oncologic outcomes. Cytotoxic chemotherapy remains the standard of care for treatment-naïve patients with metastatic disease, with a possible new treatment paradigm of carboplatin/weekly paclitaxel. In addition, immune checkpoint inhibition appears to have a promising role in the setting of patients with refractory disease. Several clinical trials with immunotherapeutic and vaccine approaches for locally advanced and metastatic anal cancer are ongoing, as are HPV-agnostic umbrella trials. Whenever possible, clinical trial enrollment is always encouraged for further therapeutic development in the setting of a rare cancer, given the potentially substantial global impact for other HPV-associated malignancies.
Collapse
Affiliation(s)
- Cathy Eng
- 1 Gastrointestinal Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Craig Messick
- 2 Surgical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rob Glynne-Jones
- 3 East and North Hertfordshire NHS Trust, Lister Hospital, Stevenage, United Kingdom
| |
Collapse
|
33
|
Ngan D, Chu J, Chander S, Michael M, Heriot AG, Ngan SY, Rischin D, Leong T. A clinical trial with protracted infusion 5-fluorouracil and mitomycin C for localized squamous cell carcinoma of the anus. Asia Pac J Clin Oncol 2018; 15:75-81. [PMID: 30536770 DOI: 10.1111/ajco.13106] [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: 05/03/2018] [Accepted: 10/27/2018] [Indexed: 11/29/2022]
Abstract
PURPOSE Mitomycin C (MMC) plus standard 5-fluorouracil (FU) infusion in weeks 1 and 5 often contributes to radiotherapy interruptions and possibly less-than-ideal outcomes in anal cancer. This study was to evaluate alternative strategies for chemotherapy delivery that might be less toxic or more efficacious, and outcomes of patient-initiated treatment interruption for severe acute toxicity. MATERIALS AND METHODS This was a prospective, nonrandomized study for patients with T1-4N0-3M0 anal squamous carcinoma. Radiotherapy of 54 Gy in 30 fractions over 6 weeks was given with infusion FU 300 mg/m2 /day for 96 hours/week for 6 weeks plus bolus MMC at 10 mg/m2 on D1. RESULTS Fifty patients were recruited (72% female). Median age was 60.5 years (35-84). Forty-seven patients (94%) received 54 Gy. Median duration of chemoradiation was 39 days (37-105). Grade 3 and 4 acute toxicity were observed in 66%. Thirty-one percent with severe acute toxicity developed severe late toxicity. Of those who experienced severe late skin toxicity, 29% did not have severe acute toxicity. Disease-free survival at 5 years was 74% (95% confidence interval [CI], 60-84), and at 9 years 61% (95% CI, 46-74). Overall survival at 5 years was 84% (95% CI, 71-92), and at 9 years 67% (95% CI, 50-81). Colostomy-free survival at 5 years was 70% (95% CI, 56-81), and at 9 years 57% (95% CI, 40-72). CONCLUSION It is feasible to deliver chemoradiation with bolus MMC and protracted infusion FU for anal cancer. Efficacy and toxicity of this regimen seem similar to conventional chemoradiation with FU/MMC. Acute skin toxicity is not a reliable predictor of severe late skin toxicity.
Collapse
Affiliation(s)
- David Ngan
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Julie Chu
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sarat Chander
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alexander G Heriot
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Samuel Y Ngan
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Danny Rischin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Trevor Leong
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Cacheux W, Tsantoulis P, Briaux A, Vacher S, Mariani P, Richard-Molard M, Buecher B, Richon S, Jeannot E, Lazartigues J, Rouleau E, Mariani O, El Alam E, Cros J, Roman-Roman S, Mitry E, Girard E, Dangles-Marie V, Lièvre A, Bièche I. Array comparative genomic hybridization identifies high level of PI3K/Akt/mTOR pathway alterations in anal cancer recurrences. Cancer Med 2018; 7:3213-3225. [PMID: 29804324 PMCID: PMC6051172 DOI: 10.1002/cam4.1533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 01/31/2023] Open
Abstract
Genomic alterations of anal squamous cell carcinoma (ASCC) remain poorly understood due to the rarity of this tumor. Array comparative genomic hybridization and targeted gene sequencing were performed in 49 cases of ASCC. The most frequently altered regions (with a frequency greater than 25%) were 10 deleted regions (2q35, 2q36.3, 3p21.2, 4p16.3, 4p31.21, 7q36.1, 8p23.3, 10q23.2, 11q22.3, and 13q14.11) and 8 gained regions (1p36.33, 1q21.1, 3q26.32, 5p15.33, 8q24.3, 9q34.3, 16p13.3, and 19p13.3). The most frequent minimal regions of deletion (55%) encompassed the 11q22.3 region containing ATM, while the most frequent minimal regions of gain (57%) encompassed the 3q26.32 region containing PIK3CA. Recurrent homozygous deletions were observed for 5 loci (ie, TGFR2 in 4 cases), and recurrent focal amplifications were observed for 8 loci (ie, DDR2 and CCND1 in 3 cases, respectively). Several of the focal amplified genes are targets for specific therapies. Integrated analysis showed that the PI3K/Akt/mTOR signaling pathway was the pathway most extensively affected, particularly in recurrences compared to treatment‐naive tumors (64% vs 30%; P = .017). In patients with ASCC recurrences, poor overall survival (OS) was significantly correlated with a large number of altered regions (P = .024). These findings provide insight into the somatic genomic alterations in ASCC and highlight the key role of the druggable PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Wulfran Cacheux
- Département d'oncologie médicale, Institut Curie, Ensemble hospitalier, Hôpital René Huguenin, Saint-Cloud, France.,Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Petros Tsantoulis
- Centre d'oncologie, Hôpitaux universitaires de Genève, Genève, Switzerland
| | - Adrien Briaux
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Sophie Vacher
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Pascale Mariani
- Département de chirurgie oncologique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Marion Richard-Molard
- Département de radio-oncologie, Institut Curie, Ensemble hospitalier, Hôpital René Huguenin, Saint-Cloud, France
| | - Bruno Buecher
- Département d'oncologie médicale, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Sophie Richon
- Centre de recherche, Institut Curie, UMR144, Paris Cedex 05, France
| | - Emmanuelle Jeannot
- Département d'anatomopathologie, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Julien Lazartigues
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Etienne Rouleau
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Odette Mariani
- Département d'anatomopathologie, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Elsy El Alam
- Département d'anatomopathologie, Institut Curie, Ensemble hospitalier, Hôpital René Huguenin, Saint-Cloud, France
| | - Jérôme Cros
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| | - Sergio Roman-Roman
- Recherche translationnelle, Centre de recherche, Institut Curie, Paris Cedex 05, France
| | - Emmanuel Mitry
- Département d'oncologie médicale, Institut Curie, Ensemble hospitalier, Hôpital René Huguenin, Saint-Cloud, France
| | - Elodie Girard
- Département de bio-informatiques, Centre de recherche, Institut Curie, Paris Cedex 05, France
| | - Virginie Dangles-Marie
- Recherche translationnelle, Centre de recherche, Institut Curie, Paris Cedex 05, France.,IFR71, Faculté des sciences biologique et pharmacologiques, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Astrid Lièvre
- Département d'oncologie médicale, Institut Curie, Ensemble hospitalier, Hôpital René Huguenin, Saint-Cloud, France.,Département de gastroentérologie, Hôpital universitaire de Rennes, Université de Rennes 1, Rennes, France
| | - Ivan Bièche
- Unité de pharmacogénomique, Département de génétique, Institut Curie, Ensemble hospitalier, Paris Cedex 05, France
| |
Collapse
|
35
|
Salem ME, Puccini A, Grothey A, Raghavan D, Goldberg RM, Xiu J, Korn WM, Weinberg BA, Hwang JJ, Shields AF, Marshall JL, Philip PA, Lenz HJ. Landscape of Tumor Mutation Load, Mismatch Repair Deficiency, and PD-L1 Expression in a Large Patient Cohort of Gastrointestinal Cancers. Mol Cancer Res 2018; 16:805-812. [PMID: 29523759 PMCID: PMC6833953 DOI: 10.1158/1541-7786.mcr-17-0735] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/19/2018] [Accepted: 02/19/2018] [Indexed: 02/06/2023]
Abstract
The efficacy of immunotherapy varies widely among different gastrointestinal cancers. Response to immune checkpoint inhibitors is shown to correlate with tumor mutation load (TML), mismatch repair deficiency (dMMR) status, and programmed cell death-ligand 1 (PD-L1) expression. Herein, we quantify TML, dMMR, and PD-L1 expression and determine their interrelationship in gastrointestinal cancers. Here, a total of 4,125 tumors from 14 different gastrointestinal cancer sites were studied using validated assays. Next-generation sequencing was performed on genomic DNA isolated from formalin-fixed paraffin-embedded tumor specimens using the NextSeq platform. TML was calculated using only somatic nonsynonymous missense mutations sequenced with a 592-gene panel. Microsatellite instability (MSI) was assessed using direct analysis of altered known MSI loci in the target regions of the sequenced genes. PD-L1 expression was analyzed by IHC. Interestingly, right-sided colon and small-bowel adenocarcinomas had the highest prevalence of TML-high tumors (14.6% and 10.2%, respectively). Pancreatic neuroendocrine tumors and gastrointestinal stromal tumors had the lowest rates of TML-high (1.3% and 0%, respectively). TML-high was strongly associated with MSI-H (P < 0.0001). However, all TML-high anal cancers (8.3%) were microsatellite stable (MSS). Higher PD-L1 expression was more likely to be seen in MSI compared with MSS tumors (20.6% vs. 7.8%, P < 0.0001).Implications: TML-high rate varied widely among gastrointestinal cancers. Although MSI is conceivably the main driver for TML-high, other factors may be involved. Future clinical trials are needed to evaluate whether the integration of TML, MSI, and PD-L1 could better identify potential responders to immunotherapy. Mol Cancer Res; 16(5); 805-12. ©2018 AACR.
Collapse
Affiliation(s)
- Mohamed E Salem
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina.
| | - Alberto Puccini
- University of Southern California, Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Axel Grothey
- Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina
| | | | | | | | - Benjamin A Weinberg
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Jimmy J Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - John L Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Heinz-Josef Lenz
- University of Southern California, Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
36
|
Shin S, Park HC, Kim MS, Han MR, Lee SH, Jung SH, Lee SH, Chung YJ. Whole-exome sequencing identified mutational profiles of squamous cell carcinomas of anus. Hum Pathol 2018; 80:1-10. [PMID: 29555573 DOI: 10.1016/j.humpath.2018.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/13/2022]
Abstract
Anal squamous cell carcinoma (ASCC), either with human papillomavirus (+) or (-), is a neoplastic disease with frequent recurrence and metastasis. To characterize ASCC genomes, we attempted to disclose novel alterations of ASCC genomes and other genetic features including mutation signatures. We performed whole-exome sequencing and copy number alteration (CNA) profiling for 8 ASCC samples from 6 patients (2 cases with primary and recurrent/metastatic tumors). We found known ASCC mutations (TP53, CDKN2A, and PIK3CA) and CNAs (gains on 3q and 19q and losses on 11q and 13q). In addition, we discovered novel mutations in HRAS and ARID1A and CNAs (gain on 8q and losses 5q, 9p, 10q, and 19p) that had not been reported in ASCCs. We identified 4 signature patterns of the mutations (signatures 1 and 2 with deamination of 5-methyl-cytosin, signature 3 with APOBEC, and signature 4 with mismatch repair) in the ASCCs. Although signatures 1 to 3 have been detected in other SCCs, signature 4 was first identified in ASCCs. In addition, we first found that ASCCs harbored chromothripsis, copy-neutral losses of heterozygosity, and focal amplification of KLF5 super-enhancer. Analyses of primary and recurrent/metastatic pair genomes revealed that driver events in development and progression of ASCC might not be uniform. Our data indicate that ASCCs may have similar mutation and CNA profiles to other SCCs, but that there are unique genomic features of ASCCs as well. Our data may provide useful information for ASCC pathogenesis and for developing clinical strategies for ASCC.
Collapse
Affiliation(s)
- Sun Shin
- Department of Microbiology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hyeon-Chun Park
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Min Sung Kim
- Department of Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi-Ryung Han
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seung Hyun Jung
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sug Hyung Lee
- Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Department of Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Yeun-Jun Chung
- Department of Microbiology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
37
|
Bong YS, Assefnia S, Tuohy T, Neklason DW, Burt RW, Ahn J, Bueno De Mesquita PJ, Byers SW. A role for the vitamin D pathway in non-intestinal lesions in genetic and carcinogen models of colorectal cancer and in familial adenomatous polyposis. Oncotarget 2018; 7:80508-80520. [PMID: 27768599 PMCID: PMC5348337 DOI: 10.18632/oncotarget.12768] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
Vitamin D is implicated in the etiology of cancers of the gastrointestinal tract, usually characterized by alteration in the APC/β-catenin/TCF tumor suppressor pathway. The vitamin D receptor (VDR) is also implicated in cardiovascular and skin diseases as well as in immunity. Activated VDR can indirectly alter β-catenin nuclear localization and directly suppress β-catenin/TCF mediated transcriptional activity. We treated VDR null mice with the carcinogen azoxymethane (AOM) and generated mice bearing a mutated APC (hypomorph) on a VDR null background (Apc1638N/+Vdr−/−). VDR null mice do not develop GI or extra-colonic tumors but loss of VDR decreased intestinal tumor latency and increased progression to adenocarcinoma in both models. AOM treatment of VDR null mice also caused squamous cell carcinoma of the anus. Although levels and distribution of total or activated β-catenin in the epithelial component of tumors were unaffected by loss of VDR, β-catenin dependent cyclin D1 expression was affected suggesting a direct VDR effect on β-catenin co-activator activity. Extra-colonic mucosa manifestations in Apc1638N/+Vdr−/− animals included increased nuclear β-catenin in submucosal stromal cells, spleno- and cardiomegaly and large epidermoid cysts characteristic of the FAP variant, Gardner's syndrome. Consistent with this, SNPs in the VDR, vitamin D binding protein and CYP24 as well as mutations in APC distal to codon 850 were strongly associated with Gardners syndrome in a cohort of 457 FAP patients, This work suggests that alterations in the vitamin D/VDR axis are important in Gardner's syndrome, as well as in the etiology of anal cancer.
Collapse
Affiliation(s)
- Yong-Sik Bong
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Shahin Assefnia
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Therese Tuohy
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Deborah W Neklason
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Randall W Burt
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Paul J Bueno De Mesquita
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Stephen W Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| |
Collapse
|
38
|
Zhao YJ, Sun WP, Peng JH, Deng YX, Fang YJ, Huang J, Zhang HZ, Wan DS, Lin JZ, Pan ZZ. Programmed death-ligand 1 expression correlates with diminished CD8+ T cell infiltration and predicts poor prognosis in anal squamous cell carcinoma patients. Cancer Manag Res 2017; 10:1-11. [PMID: 29296096 PMCID: PMC5739110 DOI: 10.2147/cmar.s153965] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective Increased expression of programmed death-ligand 1 (PD-L1) on tumor cells can be found in various malignancies; however, very limited information is known about its role in anal squamous cell carcinoma (ASCC). This study explored PD-L1 expression in ASCC patients and its association with patients' clinicopathological features, CD8+ T cell infiltration, and prognosis. Methods Formalin-fixed paraffin-embedded tumor samples from 26 patients with ASCC were retrieved. The levels of PD-L1 expression on the membrane of both tumor cells and tumor-infiltrating mononuclear cells (TIMCs) were evaluated by immunohistochemistry. CD8+ T cell densities, both within tumors and at the tumor-stromal interface, were also analyzed. Baseline clinicopathological characteristics, human papilloma virus (HPV) status, and outcome data correlated with PD-L1-positive staining. Results PD-L1 expression on tumor cells and TIMCs was observed in 46% and 50% of patients, respectively. Nineteen patients (73%) were HPV positive, with 7 showing PD-L1-positive staining on tumor cells and 9 showing PD-L1-positive staining on TIMCs. Increasing CD8+ density within tumors, but not immune stroma, was significantly associated with decreased PD-L1 expression by both tumor cells and TIMCs (P=0.0043 and P=0.0007). Patients with negative PD-L1 expression had significantly better progression-free survival (P=0.038 and P=0.0443) and a non-statistically significant trend toward longer overall survival (P=0.0882 and P=0.1222) compared with patients with positive PD-L1 expression. Conclusion PD-L1 is widely expressed on the membrane of tumor cells and TIMCs in ASCCs. Its negative impact on prognosis may be due to the diminished CD8+ T cell infiltration within tumors.
Collapse
Affiliation(s)
- Yu-Jie Zhao
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Wei-Peng Sun
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University
| | - Jian-Hong Peng
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Yu-Xiang Deng
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Yu-Jing Fang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Jun Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University
| | - Hui-Zhong Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - De-Sen Wan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Jun-Zhong Lin
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| |
Collapse
|
39
|
Exome sequencing reveals aberrant signalling pathways as hallmark of treatment-naive anal squamous cell carcinoma. Oncotarget 2017; 9:464-476. [PMID: 29416628 PMCID: PMC5787481 DOI: 10.18632/oncotarget.23066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Anal squamous cell carcinomas (ASCC) are rare tumours in humans. The etiological role of HPV infection is now well established but little is known about the molecular landscape and signalling pathways involved in the pathogenesis of this cancer. Here we report the results from a whole exome sequencing of a homogeneous group of 20 treatment-naive ASCC. A total of 2422 somatic single nucleotide variations (SNV) were found, with an overall moderate rate of somatic mutations per tumour (median: 105 relevant SNV per tumour) but a high mutational load in 3 tumours. The mutational signatures associated with age and APOBEC were observed in 100% and 60% of tumours respectively. The most frequently mutated genes were PIK3CA (25%) followed by FBXW7 (15%), FAT1 (15%), and TRIP12 (15%), the two last ones having never been described in ASCC. The main copy number alterations were gains of chromosome 3q (affecting PIK3CA) and losses of chromosome 11q (affecting ATM). The combined analysis of somatic mutations and copy number alterations show that recurrent alterations of the PI3K/AKT/mTOR pathway are frequent (60%) in these tumours, as well as potentially targetable alterations of other signalling pathways that have never been described in ASCC such as chromatin remodelling (45%) and ubiquitin mediated proteolysis (35%). These results highlight the possible implication of these aberrant signalling pathways in anal carcinogenesis and suggest promising new therapeutic approaches in ASCC. The high somatic mutation burden found in some tumours, suggesting an elevated neoantigen load could also predict sensitivity of ASCC to immunotherapy.
Collapse
|
40
|
Mirzaei H, Goudarzi H, Eslami G, Faghihloo E. Role of viruses in gastrointestinal cancer. J Cell Physiol 2017; 233:4000-4014. [PMID: 28926109 DOI: 10.1002/jcp.26194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/08/2017] [Indexed: 12/22/2022]
Abstract
Gastrointestinal cancers are a global public health problem, which represent a vast majority of all cancer-caused deaths in both men and women. On the other hand, viral pathogens have been long implicated as etiological factors in the onset of certain human cancers, including gastrointestinal tumors. In this regard, Human Papilloma Virus (HPV), Epstein-Barr Virus (EBV), and John Cunningham Virus (JCV) have been more strongly suggested to be involved in gastrointestinal carcinogenesis; so that, the association of HPV with oropharyngeal and anal cancers and also the association of EBV with gastric cancer have been etiologically confirmed by epidemiological and experimental investigations. Although, the association of other viruses is less evident, but may rely on co-factors for their oncogenic roles. Therefore, to improve the prevention and treatment of these classes of cancer, their association with viral agents as potential risk factors should be investigated with care. In this respect, the present review has focused on the existing literature on the subject of viral involvement in gastrointestinal tumorgenesis, by covering and discussing various gastrointestinal cancers, corresponding viral agents and their oncogenic aspects and then summarizing evidences either supporting or rejecting a causal role of these pathogens in gastrointestinal malignancies.
Collapse
Affiliation(s)
- Habibollah Mirzaei
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gita Eslami
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Mitkevich VA, Burnysheva KM, Petrushanko IY, Adzhubei AA, Schulga AA, Chumakov PM, Makarov AA. Binase treatment increases interferon sensitivity and apoptosis in SiHa cervical carcinoma cells by downregulating E6 and E7 human papilloma virus oncoproteins. Oncotarget 2017; 8:72666-72675. [PMID: 29069817 PMCID: PMC5641160 DOI: 10.18632/oncotarget.20199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/25/2017] [Indexed: 11/25/2022] Open
Abstract
In this study, we determined whether binase, a ribonuclease from Bacillus pumilus, increases interferon sensitivity and apoptosis in SiHa cervical cancer cells infected with high-risk human papilloma virus (HPV) strain 16. Binase treatment increased SiHa cell apoptosis in a time- and concentration-dependent manner, as determined by flow cytometry, WST tests and real time xCelligence cell index analysis. Binase-treated SiHa cells showed reduced expression of E6 and E7 viral oncoproteins and increased expression of their intracellular targets, p53 and pRb. Combined treatment with binase and IFNα2b enhanced the interferon sensitivity of HPV-positive SiHa cells. By contrast, combined treatment with binase and IFNα2b in HPV-negative C33A cervical cancer cells, which do no expess E6 and E7, elicited no changes in interferon sensitivity or p53 and pRb expression. These findings suggest binase enhances interferon sensitivity and apoptosis in HPV-positive SiHa cervical cancer cells by suppressing E6 and E7 viral protein expression.
Collapse
Affiliation(s)
- Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ksenia M. Burnysheva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexei A. Adzhubei
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey A. Schulga
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117871 Moscow, Russia
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
42
|
Darragh TM. HPV-associated disease of the anal canal: A pathology primer. SEMINARS IN COLON AND RECTAL SURGERY 2017. [DOI: 10.1053/j.scrs.2017.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Human Papillomavirus Genotyping of Incidental Malignant and Premalignant Lesions on Hemorrhoidectomy Specimens. Am J Surg Pathol 2017; 41:382-388. [DOI: 10.1097/pas.0000000000000809] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Cascinu S. Anal cancer: from an orphan disease to a curable malignancy? Lancet Oncol 2017; 18:415-416. [PMID: 28223061 DOI: 10.1016/s1470-2045(17)30091-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
|
45
|
Sclafani F, Morano F, Cunningham D, Baratelli C, Kalaitzaki E, Watkins D, Starling N, Chau I, Rao S. Platinum-Fluoropyrimidine and Paclitaxel-Based Chemotherapy in the Treatment of Advanced Anal Cancer Patients. Oncologist 2017; 22:402-408. [PMID: 28209745 DOI: 10.1634/theoncologist.2016-0241] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although treatment of localized anal cancer (AC) is well established, very little evidence is available to inform the management of advanced tumors, and the prognosis of these patients remains poor. We have analyzed treatment pathways and outcomes of a single-institution series of advanced AC patients in order to provide insight into the management of this rare condition. MATERIALS AND METHODS Inclusion criteria included epidermoid histology, inoperable locally recurrent or metastatic disease, and availability of full medical records. The primary objective was overall survival (OS). Prognostic factors were analyzed in univariate models. RESULTS Sixty-four patients (1997-2014) were included: 16 (25.0%) with inoperable locally advanced and 48 (75.0%) with metastatic tumors. Fifty-one (79.7%) received at least one line of chemotherapy; of these, 37% underwent multimodality treatment. A combination of a platinum agent plus a fluoropyrimidine was the most common first-line regimen (74.5%), with an objective response rate (ORR) of 34.4% (95% confidence interval [CI], 18.6%-53.2%). Paclitaxel-based chemotherapy was used in 15 patients as front-line or salvage treatment, and the overall ORR was 53.3% (95% CI, 26.6%-78.7%). Median progression-free survival (PFS) after first- and second-line chemotherapy was 5.8 (interquartile range [IQR], 2.8-7.6) and 3.2 (IQR, 2.5-7.1) months, respectively. Five-year OS in the overall population was 15% (95% CI, 7.0%-25.0%). Age ≤65 years and liver metastases were predictive of better PFS (hazard ratio [HR], 0.39; 95% CI, 0.16-0.97; p = .04) and worse OS (HR, 2.25; 95% CI, 1.25-4.03; p = .01), respectively. CONCLUSION A platinum agent plus a fluoropyrimidine and paclitaxel-based chemotherapy are active regimens for advanced AC. Clinical trials are needed to standardize treatment pathways, investigate the potential of novel therapeutics, and improve the poor prognosis of this rare condition. The Oncologist 2017;22:402-408Implications for Practice: Because of the lack of randomized trials, the optimal management of advanced anal cancer is uncertain. Despite its retrospective analysis and relatively small sample size, this is the second largest study ever conducted in this setting, and, as such, it has the potential to serve as a valuable source of information for everyday clinical practice. These findings suggest that chemotherapy with a platinum agent plus a fluoropyrimidine or paclitaxel-containing regimens are reasonable treatment options for patients with inoperable locally recurrent or metastatic anal carcinoma.
Collapse
Affiliation(s)
- Francesco Sclafani
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Federica Morano
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Chiara Baratelli
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Eleftheria Kalaitzaki
- Department of Clinical Research & Development, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
46
|
Neuzillet C, Rousseau B, Kocher H, Bourget P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers Part 1: GI carcinomas. Pharmacol Ther 2017; 174:145-172. [PMID: 28223233 DOI: 10.1016/j.pharmthera.2017.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Until the 1990s, cytotoxic chemotherapy has been the cornerstone of medical therapy for gastrointestinal (GI) cancers. Better understanding of the molecular biology of cancer cell has led to the therapeutic revolution of targeted therapies, i.e. monoclonal antibodies or small molecule inhibitors directed against proteins that are specifically overexpressed or mutated in cancer cells. These agents being more specific to cancer cells were expected to be less toxic than cytotoxic agents. Targeted agents have provided clinical benefit in many GI cancer types. For example, antiangiogenics and anti-EGFR therapies have significantly improved survival of patients affected by metastatic colorectal cancer and have deeply changed the therapeutic strategy in this disease. However, their effects have sometimes been disappointing, due to intrinsic or acquired resistance mechanisms (e.g., RAS mutation for anti-EGFR therapies), or to an activity restricted to some tumour settings (e.g., lack of activity in other cancer types, or on the microscopic residual disease in adjuvant setting). Many studies are negative in overall population but positive in some specific patient subgroups (e.g., trastuzumab in HER2-positive gastric cancer), illustrating the importance of patient selection and early identification of predictive biomarkers of response to these therapies. We propose a comprehensive two-part review providing a panoramic approach of the successes and failures of targeted agents in GI cancers to unravel the pharmacologic opportunities and future directions for these agents in GI oncology. In this first part, we will focus on adenocarcinomas and squamous cell carcinomas, for which targeted therapies are mostly used in combination with chemotherapy.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London, E1 1BB, United Kingdom.
| | - Benoît Rousseau
- Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Hemant Kocher
- Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London, E1 1BB, United Kingdom
| | - Philippe Bourget
- Department of Clinical Pharmacy, Necker-Enfants Malades University Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| |
Collapse
|
47
|
Balermpas P, Martin D, Wieland U, Rave-Fränk M, Strebhardt K, Rödel C, Fokas E, Rödel F. Human papilloma virus load and PD-1/PD-L1, CD8 + and FOXP3 in anal cancer patients treated with chemoradiotherapy: Rationale for immunotherapy. Oncoimmunology 2017; 6:e1288331. [PMID: 28405521 DOI: 10.1080/2162402x.2017.1288331] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022] Open
Abstract
We examined the prognostic role of immune markers programmed cell death protein-1 (PD-1) and its ligand (PD-L1), CD8+ tumor-infiltrating lymphocytes (TILs), FOXP3+ Tregs and phosphorylated Caspase-8 (T273) in patients with anal squamous cell cancer (ASCC) treated with standard chemoradiotherapy (CRT). The baseline immunohistochemical expression of immune markers was correlated with clinicopathologic characteristics, and cumulative incidence of local failure, disease-free survival (DFS) and overall survival (OS) in 150 patients, also in the context of human papilloma virus 16 (HPV16) DNA load and p16INK4a expression. After a median follow-up of 40 mo (1-205 mo), the 5-y cumulative incidence of local failure and DFS was 19.4% and 67.2%, respectively. Strong immune marker expression was significantly more common in tumors with high HPV16 viral load. In multivariant analysis, high CD8+ and PD-1+ TILs expression predicted for improved local control (p = 0.023 and p = 0.007, respectively) and DFS (p = 0.020 and p = 0.014, respectively). Also, high p16INK4a (p = 0.011) and PD-L1 (p = 0.033) expression predicted for better local control, whereas high FOXP3+ Tregs (p = 0.050) and phosphorylated Caspase-8 (p = 0.031) expression correlated with superior DFS. Female sex and high HPV16 viral load correlated with favorable outcome for all three clinical endpoints. The present data provide, for the first time, robust explanation for the favorable clinical outcome of HPV16-positive ASCC patients harboring strong immune cell infiltration. Our findings are relevant for treatment stratification with immune PD-1/PD-L1 checkpoint inhibitors to complement CRT and should be explored in a clinical trial.
Collapse
Affiliation(s)
- Panagiotis Balermpas
- Department of Radiotherapy and Oncology, Goethe-University, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt/Mainz, Heidelberg, Germany
| | - Daniel Martin
- Department of Radiotherapy and Oncology, Goethe-University , Frankfurt am Main, Germany
| | - Ulrike Wieland
- Institute of Virology, National Reference Centre for Papilloma- and Polyomaviruses, University of Cologne , Cologne, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiation Oncology, University Medical Center Göttingen , Göttingen, Germany
| | - Klaus Strebhardt
- German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt/Mainz, Heidelberg, Germany; Department of Gynecology, Goethe-University, Frankfurt am Main, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe-University, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt/Mainz, Heidelberg, Germany
| | - Emmanouil Fokas
- Department of Radiotherapy and Oncology, Goethe-University, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt/Mainz, Heidelberg, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe-University, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK) partner site: Frankfurt/Mainz, Heidelberg, Germany
| |
Collapse
|
48
|
Mouw KW, Cleary JM, Reardon B, Pike J, Braunstein LZ, Kim J, Amin-Mansour A, Miao D, Damish A, Chin J, Ott PA, Fuchs CS, Martin NE, Getz G, Carter S, Mamon HJ, Hornick JL, Van Allen EM, D'Andrea AD. Genomic Evolution after Chemoradiotherapy in Anal Squamous Cell Carcinoma. Clin Cancer Res 2016; 23:3214-3222. [PMID: 27852700 DOI: 10.1158/1078-0432.ccr-16-2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Squamous cell carcinoma of the anal canal (ASCC) accounts for 2% to 4% of gastrointestinal malignancies in the United States and is increasing in incidence; however, genomic features of ASCC are incompletely characterized. Primary treatment of ASCC involves concurrent chemotherapy and radiation (CRT), but the mutational landscape of resistance to CRT is unknown. Here, we aim to compare mutational features of ASCC in the pre- and post-CRT setting.Experimental Design: We perform whole-exome sequencing of primary (n = 31) and recurrent (n = 30) ASCCs and correlate findings with clinical data. We compare genomic features of matched pre- and post-CRT tumors to identify genomic features of CRT response. Finally, we investigate the mutational underpinnings of an extraordinary ASCC response to immunotherapy.Results: We find that both primary and recurrent ASCC tumors harbor mutations in genes, such as PIK3CA and FBXW7, that are also mutated in other HPV-associated cancers. Overall mutational burden was not significantly different in pre- versus post-CRT tumors, and several examples of shared clonal driver mutations were identified. In two cases, clonally related pre- and post-CRT tumors harbored distinct oncogenic driver mutations in the same cancer gene (KRAS or FBXW7). A patient with recurrent disease achieved an exceptional response to anti-programmed death (PD-1) therapy, and genomic dissection revealed high mutational burden and predicted neoantigen load.Conclusions: We perform comprehensive mutational analysis of ASCC and characterize mutational features associated with CRT. Although many primary and recurrent tumors share driver events, we identify several unique examples of clonal evolution in response to treatment. Clin Cancer Res; 23(12); 3214-22. ©2016 AACR.
Collapse
Affiliation(s)
- Kent W Mouw
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Brendan Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jonathan Pike
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lior Z Braunstein
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jaegil Kim
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Ali Amin-Mansour
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Diana Miao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Alexis Damish
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joanna Chin
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles S Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Neil E Martin
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Scott Carter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Harvey J Mamon
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Alan D D'Andrea
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.,Ludwig Center at Harvard, Boston, Massachusetts
| |
Collapse
|
49
|
Chung JH, Sanford E, Johnson A, Klempner SJ, Schrock AB, Palma NA, Erlich RL, Frampton GM, Chalmers ZR, Vergilio J, Rubinson DA, Sun JX, Chmielecki J, Yelensky R, Suh JH, Lipson D, George TJ, Elvin JA, Stephens PJ, Miller VA, Ross JS, Ali SM. Comprehensive genomic profiling of anal squamous cell carcinoma reveals distinct genomically defined classes. Ann Oncol 2016; 27:1336-41. [PMID: 27052656 DOI: 10.1093/annonc/mdw152] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/22/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Squamous cell cancers of the anal canal (ASCC) are increasing in frequency and lack effective therapies for advanced disease. Although an association with human papillomavirus (HPV) has been established, little is known about the molecular characterization of ASCC. A comprehensive genomic analysis of ASCC was undertaken to identify novel genomic alterations (GAs) that will inform therapeutic choices for patients with advanced disease. PATIENTS AND METHODS Hybrid-capture-based next-generation sequencing of exons from 236 cancer-related genes and intronic regions from 19 genes commonly rearranged in cancer was performed on 70 patients with ASCC. HPV status was assessed by aligning tumor sequencing reads to HPV viral genomes. GAs were identified using an established algorithm and correlated with HPV status. RESULTS Sixty-one samples (87%) were HPV-positive. A mean of 3.5 GAs per sample was identified. Recurrent alterations in phosphoinositol-3-kinase pathway (PI3K/AKT/mTOR) genes including amplifications and homozygous deletions were present in 63% of cases. Clinically relevant GAs in genes involved in DNA repair, chromatin remodeling, or receptor tyrosine kinase signaling were observed in 30% of cases. Loss-of-function mutations in TP53 and CDKN2A were significantly enhanced in HPV-negative cases (P < 0.0001). CONCLUSIONS This is the first comprehensive genomic analysis of ASCC, and the results suggest new therapeutic approaches. Differing genomic profiles between HPV-associated and HPV-negative ASCC warrants further investigation and may require novel therapeutic and preventive strategies.
Collapse
Affiliation(s)
| | | | | | - S J Klempner
- Division of Hematology-Oncology, University of California Irvine, Irvine
| | | | | | | | | | | | | | - D A Rubinson
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston
| | - J X Sun
- Foundation Medicine, Cambridge
| | | | | | - J H Suh
- Foundation Medicine, Cambridge
| | | | - T J George
- Division of Hematology-Oncology, University of Florida, Gainesville
| | | | | | | | - J S Ross
- Foundation Medicine, Cambridge Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, USA
| | - S M Ali
- Foundation Medicine, Cambridge
| |
Collapse
|