1
|
The PTEN Tumor Suppressor Gene in Soft Tissue Sarcoma. Cancers (Basel) 2019; 11:cancers11081169. [PMID: 31416195 PMCID: PMC6721622 DOI: 10.3390/cancers11081169] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/26/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Soft tissue sarcoma (STS) is a rare malignancy of mesenchymal origin classified into more than 50 different subtypes with distinct clinical and pathologic features. Despite the poor prognosis in the majority of patients, only modest improvements in treatment strategies have been achieved, largely due to the rarity and heterogeneity of these tumors. Therefore, the discovery of new prognostic and predictive biomarkers, together with new therapeutic targets, is of enormous interest. Phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor that commonly loses its function via mutation, deletion, transcriptional silencing, or protein instability, and is frequently downregulated in distinct sarcoma subtypes. The loss of PTEN function has consequent alterations in important pathways implicated in cell proliferation, survival, migration, and genomic stability. PTEN can also interact with other tumor suppressors and oncogenic signaling pathways that have important implications for the pathogenesis in certain STSs. The aim of the present review is to summarize the biological significance of PTEN in STS and its potential role in the development of new therapeutic strategies.
Collapse
|
2
|
Rodon J, Soria JC, Berger R, Miller WH, Rubin E, Kugel A, Tsimberidou A, Saintigny P, Ackerstein A, Braña I, Loriot Y, Afshar M, Miller V, Wunder F, Bresson C, Martini JF, Raynaud J, Mendelsohn J, Batist G, Onn A, Tabernero J, Schilsky RL, Lazar V, Lee JJ, Kurzrock R. Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial. Nat Med 2019; 25:751-758. [PMID: 31011205 DOI: 10.1038/s41591-019-0424-4] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/14/2019] [Indexed: 12/21/2022]
Abstract
Precision medicine focuses on DNA abnormalities, but not all tumors have tractable genomic alterations. The WINTHER trial ( NCT01856296 ) navigated patients to therapy on the basis of fresh biopsy-derived DNA sequencing (arm A; 236 gene panel) or RNA expression (arm B; comparing tumor to normal). The clinical management committee (investigators from five countries) recommended therapies, prioritizing genomic matches; physicians determined the therapy given. Matching scores were calculated post-hoc for each patient, according to drugs received: for DNA, the number of alterations matched divided by the total alteration number; for RNA, expression-matched drug ranks. Overall, 303 patients consented; 107 (35%; 69 in arm A and 38 in arm B) were evaluable for therapy. The median number of previous therapies was three. The most common diagnoses were colon, head and neck, and lung cancers. Among the 107 patients, the rate of stable disease ≥6 months and partial or complete response was 26.2% (arm A: 23.2%; arm B: 31.6% (P = 0.37)). The patient proportion with WINTHER versus previous therapy progression-free survival ratio of >1.5 was 22.4%, which did not meet the pre-specified primary end point. Fewer previous therapies, better performance status and higher matching score correlated with longer progression-free survival (all P < 0.05, multivariate). Our study shows that genomic and transcriptomic profiling are both useful for improving therapy recommendations and patient outcome, and expands personalized cancer treatment.
Collapse
Affiliation(s)
- Jordi Rodon
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Wilson H Miller
- Segal Cancer Centre, Jewish General Hospital, QCROC-Quebec Cancer Consortium and Rossy Cancer Network, McGill University, Montreal, Québec, Canada
| | - Eitan Rubin
- Ben-Gurion University of the Negev, Beersheva, Israel
| | | | - Apostolia Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Irene Braña
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | | | - Fanny Wunder
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France
| | - Catherine Bresson
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France
| | | | | | - John Mendelsohn
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France.,Sheikh Khalifa Bin Zayad Al Nahyan Institute for Personalized Cancer Therapy (IPCT), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, QCROC-Quebec Cancer Consortium and Rossy Cancer Network, McGill University, Montreal, Québec, Canada
| | - Amir Onn
- Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Josep Tabernero
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Richard L Schilsky
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France.,American Society of Clinical Oncology (ASCO), Alexandria, VA, USA
| | - Vladimir Lazar
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Razelle Kurzrock
- Worldwide Innovative Network (WIN) Association-WIN Consortium, Villejuif, France. .,University of California San Diego, Moores Cancer Center, San Diego, CA, USA.
| |
Collapse
|
3
|
Goodman AM, Kato S, Chattopadhyay R, Okamura R, Saunders IM, Montesion M, Frampton GM, Miller VA, Daniels GA, Kurzrock R. Phenotypic and Genomic Determinants of Immunotherapy Response Associated with Squamousness. Cancer Immunol Res 2019; 7:866-873. [PMID: 31003990 DOI: 10.1158/2326-6066.cir-18-0716] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/10/2019] [Accepted: 04/16/2019] [Indexed: 12/25/2022]
Abstract
Advanced and metastatic squamous cell carcinomas (SCC) are common and difficult-to-treat malignancies. We assessed 75 immunotherapy-treated patients with SCC from a clinically annotated database of 2,651 patients, as well as 9,407 patients from a deidentified database for molecular features that might influence checkpoint blockade response. SCCs had higher tumor mutational burdens (TMB) than non-SCCs (P < 0.0001). Cutaneous SCCs had the highest TMB (P < 0.0001), with 41.3% demonstrating a very high TMB (≥50 mutations/Mb). In immunotherapy-treated patients with SCC, higher TMB (≥12 mutations/Mb) correlated with a trend to higher clinical benefit rate [stable disease ≥ 6 months or partial/complete remission; 60% vs. 29%; (high vs. low TMB); P = 0.06] and significantly longer median time-to-treatment failure (TTF; 9.9 vs. 4.4 months; P = 0.0058). Cutaneous SCCs had the highest clinical benefit [11/15 patients (73%) vs. 20/60 (33%) non-cutaneous (P = 0.008)], TTF (P = 0.0015), and overall survival (P = 0.06) with immunotherapy treatment. In conclusion, among a diverse set of SCCs, higher TMB and cutaneous disease associated with better immunotherapy outcome.
Collapse
Affiliation(s)
- Aaron M Goodman
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, California. .,Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California
| | - Shumei Kato
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, California.,Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, California
| | - Ranajoy Chattopadhyay
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, California
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, California
| | - Ila M Saunders
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | | | | | | | - Gregory A Daniels
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, California
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, California.,Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, California
| |
Collapse
|
4
|
von der Grün J, Rödel F, Brandts C, Fokas E, Guckenberger M, Rödel C, Balermpas P. Targeted Therapies and Immune-Checkpoint Inhibition in Head and Neck Squamous Cell Carcinoma: Where Do We Stand Today and Where to Go? Cancers (Basel) 2019; 11:E472. [PMID: 30987257 PMCID: PMC6521064 DOI: 10.3390/cancers11040472] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 01/12/2023] Open
Abstract
With an increased understanding of the tumor biology of squamous cell carcinoma of the head and neck (SCCHN), targeted therapies have found their way into the clinical treatment routines against this entity. Nevertheless, to date platinum-based cytostatic agents remain the first line choice and targeting the epidermal growth factor-receptor (EGFR) with combined cetuximab and radiation therapy remains the only targeted therapy approved in the curative setting. Investigation of immune checkpoint inhibitors (ICI), such as antibodies targeting programmed cell death protein 1 (PD-1) and its ligand PD-L1, resulted in a change of paradigms in oncology and in the first approval of new drugs for treating SCCHN. Nivolumab and pembrolizumab, two anti-PD-1 antibodies, were the first agents shown to improve overall survival for patients with metastatic/recurrent tumors in recent years. Currently, several clinical trials investigate the role of ICI in different therapeutic settings. A robust set of biomarkers will be an inevitable tool for future individualized treatment approaches including radiation dose de-escalation and escalation strategies. This review aims to summarize achieved goals, the current status and future perspectives regarding targeted therapies and ICI in the management of SCCHN.
Collapse
Affiliation(s)
- Jens von der Grün
- Department of Radiation Oncology, Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
| | - Franz Rödel
- Department of Radiation Oncology, Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
- German Cancer Consortium (DKTK), partner site: Frankfurt a. M., Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
| | - Christian Brandts
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
- German Cancer Consortium (DKTK), partner site: Frankfurt a. M., Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- Department of Medicine, Hematology/Oncology, University Cancer Center Frankfurt (UCT), Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
| | - Emmanouil Fokas
- Department of Radiation Oncology, Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
- German Cancer Consortium (DKTK), partner site: Frankfurt a. M., Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
| | - Matthias Guckenberger
- Department of Radiation Oncology, Rämistrasse 100, University Hospital Zurich, 8091 Zürich, Switzerland.
| | - Claus Rödel
- Department of Radiation Oncology, Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
- German Cancer Consortium (DKTK), partner site: Frankfurt a. M., Theodor-Stern-Kai 7, University of Frankfurt, 60590 Frankfurt, Germany.
| | - Panagiotis Balermpas
- Department of Radiation Oncology, Rämistrasse 100, University Hospital Zurich, 8091 Zürich, Switzerland.
| |
Collapse
|
5
|
Eze N, Lee JW, Yang DH, Zhu F, Neumeister V, Sandoval-Schaefer T, Mehra R, Ridge JA, Forastiere A, Chung CH, Burtness B. PTEN loss is associated with resistance to cetuximab in patients with head and neck squamous cell carcinoma. Oral Oncol 2019; 91:69-78. [PMID: 30926065 PMCID: PMC6855599 DOI: 10.1016/j.oraloncology.2019.02.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/06/2019] [Accepted: 02/23/2019] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Cetuximab, a monoclonal antibody to the epidermal growth factor receptor (EGFR), extends survival in combination with standard therapy in head and neck squamous cell carcinoma (HNSCC). However, as effects are modest, and patients experience side effects, a biomarker to predict resistance and personalize therapy is needed. Activation of signaling pathways downstream from receptor tyrosine kinases predicts resistance to such therapies in other cancers. The most common abnormalities downstream from EGFR in HNSCC are in the PI3K pathway, activated via loss of expression of the regulator PTEN, or via PI3K mutation. We studied whether PTEN and/or PI3K abnormalities predict resistance to cetuximab. METHODS Tumor PTEN and PIK3CA/PI3K p110α were analyzed in samples from subjects treated on two trials of cetuximab-based therapy for patients with metastatic or recurrent HNSCC: E5397, a randomized trial of cisplatin plus placebo versus cisplatin plus cetuximab; and NCI-8070, a randomized trial of cetuximab plus sorafenib versus cetuximab. In situ quantification of PTEN and PI3K p110 α was performed using the AQUA™ method of quantitative immunofluorescence. PI3KCA hot spot mutations were determined with BEAMing. RESULTS For E5397, in multivariable analysis, PTEN expressing/PIK3CA WT patients tended to improve PFS with cetuximab compared to placebo (N = 48; HR = 0.54, Wald p = 0.0502). High PTEN expression was significantly associated with superior PFS among patients treated on NCI-8070 (N = 37; HR = 0.35, p = 0.008). CONCLUSION Loss of PTEN expression may be associated with lack of benefit from cetuximab. This analysis is limited by small sample size, and PTEN as a potential predictive biomarker merits validation in larger sample sets.
Collapse
Affiliation(s)
- Nnamdi Eze
- Department of Medicine, Yale University School of Medicine and Yale Cancer Center, United States
| | - Ju-Whei Lee
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, United States
| | - Dong-Hua Yang
- Biosample Repository, Fox Chase Cancer Center, United States
| | - Fang Zhu
- Department of Biostatistics, Fox Chase Cancer Center, United States
| | | | - Teresa Sandoval-Schaefer
- Department of Medicine, Yale University School of Medicine and Yale Cancer Center, United States
| | - Ranee Mehra
- Department of Medical Oncology, Fox Chase Cancer Center, United States
| | - John A Ridge
- Department of Surgical Oncology, Fox Chase Cancer Center, United States
| | - Arlene Forastiere
- Departments of Medicine, Radiation Oncology and Otolaryngology, Sidney Kimmel Cancer Center, Johns Hopkins University, United States
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, United States
| | - Barbara Burtness
- Department of Medicine, Yale University School of Medicine and Yale Cancer Center, United States.
| |
Collapse
|
6
|
Dunn LA, Fury MG, Xiao H, Baxi SS, Sherman EJ, Korte S, Pfister C, Haque S, Katabi N, Ho AL, Pfister DG. A phase II study of temsirolimus added to low-dose weekly carboplatin and paclitaxel for patients with recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). Ann Oncol 2018; 28:2533-2538. [PMID: 28961834 DOI: 10.1093/annonc/mdx346] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Activating events along the PI3K/mTOR pathway are common in head and neck squamous cell carcinomas (HNSCC), and preclinical studies suggest additive or synergistic effects when combining mTORC1 inhibitors with carboplatin and paclitaxel chemotherapy. Patients and methods In this single-institution phase II study, the combination of temsirolimus 25 mg, carboplatin AUC 1.5, and paclitaxel 80 mg/m2 administered on days 1 and 8 of a 21-day cycle was evaluated in 36 patients with recurrent and/or metastatic (R/M) HNSCC. The primary end point was objective response rate after two cycles of treatment. Secondary end points include the safety and tolerability profile and overall survival. Correlative studies with exome mutational analysis were performed in pre-treatment biopsy samples from 21 patients. Results Fifteen (41.7%) patients had an objective response, which were all partial responses, and 19 (52.3%) patients had stable disease as best response. The two patients who were designated as 'non-responders' were removed from study prior to two cycles of treatment, but are included in the efficacy and safety analyses. The median duration on study was 5.3 months and the median progression-free survival and overall survival were 5.9 months (95% confidence interval, 4.8-7.1) and 12.8 months (95% confidence interval, 9.8-15.8), respectively. The most common grade 3 and 4 adverse events were hematologic toxicities. Three (3.8%) patients developed neutropenic fever on study. Three of four patients with PIK3CA mutations experienced tumor regressions, and responses were also seen in patients with other genetic alterations in the PI3K/mTOR pathway. Conclusion The combination of temsirolimus with low-dose weekly carboplatin and paclitaxel appears to have meaningful clinical efficacy in the treatment of R/M HNSCC. This regimen has a relatively high response rate compared to other treatments evaluated in R/M HNSCC, and potential associations with genetic alterations in the PI3K/mTOR pathway should be further explored.
Collapse
Affiliation(s)
- L A Dunn
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine;.
| | - M G Fury
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - H Xiao
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - S S Baxi
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - E J Sherman
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - S Korte
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - C Pfister
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | | | - N Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A L Ho
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| | - D G Pfister
- Section of Head and Neck Oncology, Division of Solid Tumor, Department of Medicine
| |
Collapse
|
7
|
Ferreira DM, Neves TJ, Lima LGCA, Alves FA, Begnami MD. Prognostic implications of the phosphatidylinositol 3-kinase/Akt signaling pathway in oral squamous cell carcinoma: overexpression of p-mTOR indicates an adverse prognosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s41241-017-0046-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
8
|
Niehr F, Eder T, Pilz T, Konschak R, Treue D, Klauschen F, Bockmayr M, Türkmen S, Jöhrens K, Budach V, Tinhofer I. Multilayered Omics-Based Analysis of a Head and Neck Cancer Model of Cisplatin Resistance Reveals Intratumoral Heterogeneity and Treatment-Induced Clonal Selection. Clin Cancer Res 2017; 24:158-168. [PMID: 29061642 DOI: 10.1158/1078-0432.ccr-17-2410] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/04/2017] [Accepted: 10/11/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Platinum-based drugs, in particular cisplatin (cis-diamminedichloridoplatinum(II), CDDP), are used for treatment of squamous cell carcinoma of the head and neck (SCCHN). Despite initial responses, CDDP treatment often results in chemoresistance, leading to therapeutic failure. The role of primary resistance at subclonal level and treatment-induced clonal selection in the development of CDDP resistance remains unknown.Experimental Design: By applying targeted next-generation sequencing, fluorescence in situ hybridization, microarray-based transcriptome, and mass spectrometry-based phosphoproteome analysis to the CDDP-sensitive SCCHN cell line FaDu, a CDDP-resistant subline, and single-cell derived subclones, the molecular basis of CDDP resistance was elucidated. The causal relationship between molecular features and resistant phenotypes was determined by siRNA-based gene silencing. The clinical relevance of molecular findings was validated in patients with SCCHN with recurrence after CDDP-based chemoradiation and the TCGA SCCHN dataset.Results: Evidence of primary resistance at clonal level and clonal selection by long-term CDDP treatment was established in the FaDu model. Resistance was associated with aneuploidy of chromosome 17, increased TP53 copy-numbers and overexpression of the gain-of-function (GOF) mutant variant p53R248L siRNA-mediated knockdown established a causal relationship between mutant p53R248L and CDDP resistance. Resistant clones were also characterized by increased activity of the PI3K-AKT-mTOR pathway. The poor prognostic value of GOF TP53 variants and mTOR pathway upregulation was confirmed in the TCGA SCCHN cohort.Conclusions: Our study demonstrates a link of intratumoral heterogeneity and clonal evolution as important mechanisms of drug resistance in SCCHN and establishes mutant GOF TP53 variants and the PI3K/mTOR pathway as molecular targets for treatment optimization. Clin Cancer Res; 24(1); 158-68. ©2017 AACR.
Collapse
Affiliation(s)
- Franziska Niehr
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK) Partner Site Berlin, Berlin, Germany
| | - Theresa Eder
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK) Partner Site Berlin, Berlin, Germany
| | - Tanja Pilz
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany
| | - Robert Konschak
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK) Partner Site Berlin, Berlin, Germany
| | - Denise Treue
- Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Frederick Klauschen
- German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK) Partner Site Berlin, Berlin, Germany.,Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Michael Bockmayr
- Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Seval Türkmen
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Human Genetics, Berlin, Germany
| | - Korinna Jöhrens
- Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Volker Budach
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany
| | - Ingeborg Tinhofer
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiooncology and Radiotherapy, Berlin, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK) Partner Site Berlin, Berlin, Germany
| |
Collapse
|
9
|
Eze N, Lo YC, Burtness B. Biomarker driven treatment of head and neck squamous cell cancer. CANCERS OF THE HEAD & NECK 2017; 2:6. [PMID: 31093353 PMCID: PMC6460531 DOI: 10.1186/s41199-017-0025-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Abstract Treatment modalities of head and neck squamous cell cancer include surgery, radiation, chemotherapy, targeted agents and immune checkpoint inhibition. Treatment is often toxic and can affect long-term function and quality of life. In this context, identification of biomarker data that can help tailor therapy on an individualized basis and reduce treatment-related toxicity would be highly beneficial. A variety of predictive biomarkers have been discovered and are already utilized in clinical practice, while many more are being explored. We will review p16 overexpression as a surrogate biomarker in HPV-associated head and neck cancer and plasma EBV DNA as a biomarker in nasopharyngeal carcinoma, the two established biomarkers currently utilized in clinical practice. We will also examine novel predictive biomarkers that are in clinical development and may shape the future landscape of targeted head and neck cancer therapy. These emerging biomarkers include the tyrosine kinases and their signaling pathway, immune checkpoint biomarkers, tumor suppressor abnormalities, and molecular predictors of hypoxia-targeted therapy. We will also look at futuristic biomarkers including detection of circulating DNA from clinical specimens and rapid tumor profiling. We will highlight the ongoing effort that will see a shift from prognostic to predictive biomarker development in head and neck cancer with the goal of delivering individualized cancer therapy. Trial registration N/A.
Collapse
Affiliation(s)
- Nnamdi Eze
- 1Section of Medical Oncology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, 333 Cedar Street, Room WWW-221, P.O. Box 208028, New Haven, CT 06520 USA
| | - Ying-Chun Lo
- 2Department of Pathology, Yale University School of Medicine, New Haven, CT USA
| | - Barbara Burtness
- 3Section of Medical Oncology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT USA
| |
Collapse
|
10
|
Predicting clinical benefit from everolimus in patients with advanced solid tumors, the CPCT-03 study. Oncotarget 2017; 8:55582-55592. [PMID: 28903445 PMCID: PMC5589684 DOI: 10.18632/oncotarget.16029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/07/2017] [Indexed: 01/18/2023] Open
Abstract
Background In this study, our aim was to identify molecular aberrations predictive for response to everolimus, an mTOR inhibitor, regardless of tumor type. Methods To generate hypotheses about potential markers for sensitivity to mTOR inhibition, drug sensitivity and genomic profiles of 835 cell lines were analyzed. Subsequently, a multicenter study was conducted. Patients with advanced solid tumors lacking standard of care treatment options were included and underwent a pre-treatment tumor biopsy to enable DNA sequencing of 1,977 genes, derive copy number profiles and determine activation status of pS6 and pERK. Treatment benefit was determined according to TTP ratio and RECIST. We tested for associations between treatment benefit and single molecular aberrations, clusters of aberrations and pathway perturbation. Results Cell line screens indicated several genes, such as PTEN (P = 0.016; Wald test), to be associated with sensitivity to mTOR inhibition. Subsequently 73 patients were included, of which 59 started treatment with everolimus. Response and molecular data were available from 43 patients. PTEN aberrations, i.e. copy number loss or mutation, were associated with treatment benefit (P = 0.046; Fisher's exact test). Conclusion Loss-of-function aberrations in PTEN potentially represent a tumor type agnostic biomarker for benefit from everolimus and warrants further confirmation in subsequent studies.
Collapse
|
11
|
Schwaederle M, Daniels GA, Piccioni DE, Kesari S, Fanta PT, Schwab RB, Shimabukuro KA, Parker BA, Kurzrock R. Next generation sequencing demonstrates association between tumor suppressor gene aberrations and poor outcome in patients with cancer. Cell Cycle 2016; 14:1730-7. [PMID: 25928476 PMCID: PMC4614790 DOI: 10.1080/15384101.2015.1033596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Next generation sequencing is transforming patient care by allowing physicians to customize and match treatment to their patients’ tumor alterations. Our goal was to study the association between key molecular alterations and outcome parameters. We evaluated the characteristics and outcomes (overall survival (OS), time to metastasis/recurrence, and best progression-free survival (PFS)) of 392 patients for whom next generation sequencing (182 or 236 genes) had been performed. The Kaplan-Meier method and Cox regression models were used for our analysis, and results were subjected to internal validation using a resampling method (bootstrap analysis). In a multivariable analysis (Cox regression model), the parameters that were statistically associated with a poorer overall survival were the presence of metastases at diagnosis (P = 0.014), gastrointestinal histology (P < 0.0001), PTEN (P < 0.0001), and CDKN2A alterations (P = 0.0001). The variables associated with a shorter time to metastases/recurrence were gastrointestinal histology (P = 0.004), APC (P = 0.008), PTEN (P = 0.026) and TP53 (P = 0.044) alterations. TP53 (P = 0.003) and PTEN (P = 0.034) alterations were independent predictors of a shorter best PFS. A personalized treatment approach (matching the molecular aberration with a cognate targeted drug) also correlated with a longer best PFS (P = 0.046). Our study demonstrated that, across diverse cancers, anomalies in specific tumor suppressor genes (PTEN, CDKN2A, APC, and/or TP53) were independently associated with a worse outcome, as reflected by time to metastases/recurrence, best PFS on treatment, and/or overall survival. These observations suggest that molecular diagnostic tests may provide important prognostic information in patients with cancer.
Collapse
Affiliation(s)
- Maria Schwaederle
- a Center for Personalized Cancer Therapy, and Division of Hematology and Oncology; UCSD Moores Cancer Center ; La Jolla , CA , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Massarelli E, Ferrarotto R, Glisson BS. New Strategies in Human Papillomavirus–Related Oropharynx Cancer: Effecting Advances in Treatment for a Growing Epidemic. Clin Cancer Res 2015; 21:3821-8. [DOI: 10.1158/1078-0432.ccr-14-1329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Schwaederle M, Elkin SK, Tomson BN, Carter JL, Kurzrock R. Squamousness: Next-generation sequencing reveals shared molecular features across squamous tumor types. Cell Cycle 2015; 14:2355-61. [PMID: 26030731 PMCID: PMC4613537 DOI: 10.1080/15384101.2015.1053669] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In order to gain a better understanding of the underlying biology of squamous cell carcinoma (SCC), we tested the hypothesis that SCC originating from different organs may possess common molecular alterations. SCC samples (N = 361) were examined using clinical-grade targeted next-generation sequencing (NGS). The most frequent SCC tumor types were head and neck, lung, cutaneous, gastrointestinal and gynecologic cancers. The most common gene alterations were TP53 (64.5% of patients), PIK3CA (28.5%), CDKN2A (24.4%), SOX2 (17.7%), and CCND1 (15.8%). By comparing NGS results of our SCC cohort to a non-SCC cohort (N = 277), we found that CDKN2A, SOX2, NOTCH1, TP53, PIK3CA, CCND1, and FBXW7 were significantly more frequently altered, unlike KRAS, which was less frequently altered in SCC specimens (all P < 0.05; multivariable analysis). Therefore, we identified “squamousness” gene signatures (TP53, PIK3CA, CCND1, CDKN2A, SOX2, NOTCH 1, and FBXW7 aberrations, and absence of KRAS alterations) that were significantly more frequent in SCC versus non-SCC histologies. A multivariable co-alteration analysis established 2 SCC subgroups: (i) patients in whom TP53 and cyclin pathway (CDKN2A and CCND1) alterations strongly correlated but in whom PIK3CA aberrations were less frequent; and (ii) patients with PIK3CA alterations in whom TP53 mutations were less frequent (all P ≤ 0 .001, multivariable analysis). In conclusion, we identified a set of 8 genes altered with significantly different frequencies when SCC and non-SCC were compared, suggesting the existence of patterns for “squamousness.” Targeting the PI3K-AKT-mTOR and/or cyclin pathway components in SCC may be warranted.
Collapse
Affiliation(s)
- Maria Schwaederle
- a Center for Personalized Cancer Therapy; University of California San Diego Moores Cancer Center ; La Jolla , CA USA
| | | | | | | | | |
Collapse
|
14
|
Moulder S, Helgason T, Janku F, Wheler J, Moroney J, Booser D, Albarracin C, Morrow PK, Atkins J, Koenig K, Gilcrease M, Kurzrock R. Inhibition of the phosphoinositide 3-kinase pathway for the treatment of patients with metastatic metaplastic breast cancer. Ann Oncol 2015; 26:1346-52. [PMID: 25878190 DOI: 10.1093/annonc/mdv163] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 03/16/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Mesenchymal/metaplastic breast cancers (MpBCs) are often triple-negative (TNBC), and chemo-refractory, and can harbor phosphoinositide 3-kinase (PI3kinase) alterations; thus, therapy with mTor inhibitors may demonstrate activity. PATIENTS AND METHODS Patients with mesenchymal/MpBC treated with temsirolimus-based regimens were evaluated. Mutational analyses [polymerase chain reaction (PCR)-based DNA sequencing method, mass spectrometric detection (Sequenom MassARRAY), or next-generation sequencing] as well as loss of phosphatase and tensin homolog (PTEN) (immunohistochemistry) were performed (archived tissue when available). RESULTS Twenty-three patients (one of whom was on two separate trials) were treated using temsirolimus-containing regimens: temsirolimus alone (n = 1 patient) or combined with the following: liposomal doxorubicin and bevacizumab (DAT, n = 18); liposomal doxorubicin (DT, n = 1); paclitaxel and bevacizumab (TAT, n = 2); paclitaxel (TT, n = 1); carboplatin and bevacizumab (CAT, n = 1). Response rate [complete response (CR) + partial response (PR)] was 25% across all regimens; 32% in the anthracycline-based regimens [DAT and DT (CR = 2, PR = 4; N = 19)]. An additional two patients achieved stable disease (SD) ≥6 months [total SD ≥6 months/CR/PR = 8 (33%)]. Molecular aberrations in the PI3K pathway were common: PIK3CA mutation = 6/15 (40%), PTEN mutation = 3/11 (27%), and PTEN loss = 2/11 (18%). A point mutation in the NF2 gene (K159fs*16; NF2 alterations can activate mTor) was found in one patient who attained CR (3+ years). Of the eight patients who achieved SD ≥6 months/CR/PR, all 4 patients with available tissue had a molecular aberration that activate the PIK3CA/Akt/mTOR axis: PIK3CA mutation = 2; PTEN loss = 1; NF2 aberration = 1. CONCLUSIONS DAT has activity in MpBCs including complete CRs. Molecular aberrations that can activate the PI3 K/Akt/mTOR axis are common in MpBC.
Collapse
Affiliation(s)
- S Moulder
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston Department of Breast Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - T Helgason
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston
| | - F Janku
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston
| | - J Wheler
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston
| | - J Moroney
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston
| | - D Booser
- Department of Breast Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - C Albarracin
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - P K Morrow
- Department of Breast Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - J Atkins
- Department of Investigational Cancer Therapeutics (Phase I), The University of Texas, M.D. Anderson Cancer Center, Houston
| | - K Koenig
- Department of Breast Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - M Gilcrease
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston
| | - R Kurzrock
- Department of Hematology and Oncology, UCSD Moores Cancer Center, San Diego, USA
| |
Collapse
|
15
|
Bowles DW, McDermott JD, Jimeno A. Novel treatments for head and neck squamous cell carcinoma: preclinical identification and clinical investigation. Future Oncol 2015; 10:1065-80. [PMID: 24941990 DOI: 10.2217/fon.14.18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common cancer worldwide. Classically, it is a disease related to tobacco and alcohol use; an increasing number of patients are being diagnosed with HNSCC caused by infection with the human papillomavirus. New deep-sequencing techniques have confirmed the importance of p53 and EGF receptor in HNSCC development, and have identified pathways of critical importance, such as PI3K/mTOR and NOTCH. Increasing knowledge of key molecular features has lead to new therapeutic avenues for HNSCC. Novel therapies under investigation in HNSCC include antibody and small molecule inhibitors of EGF receptor and its family members, PI3K inhibitors, antiangiogenic agents, immunotherapies and agents interacting with early developmental pathways such as Hedgehog.
Collapse
Affiliation(s)
- Daniel W Bowles
- Division of Medical Oncology, University of Colorado School of Medicine, CO, USA
| | | | | |
Collapse
|
16
|
Grünwald V, Keilholz U, Boehm A, Guntinas-Lichius O, Hennemann B, Schmoll HJ, Ivanyi P, Abbas M, Lehmann U, Koch A, Karch A, Zörner A, Gauler TC. TEMHEAD: a single-arm multicentre phase II study of temsirolimus in platin- and cetuximab refractory recurrent and/or metastatic squamous cell carcinoma of the head and neck (SCCHN) of the German SCCHN Group (AIO). Ann Oncol 2014; 26:561-7. [PMID: 25527417 DOI: 10.1093/annonc/mdu571] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Squamous cell carcinoma of the head and neck (SCCHN) is a common disease, which has a poor prognosis after failure of therapy. Activation of the PI3K-AKT-mTOR axis is commonly detected in recurrent or metastatic SCCHN, and provided the rationale for the clinical phase II trial in pretreated SCCHN. PATIENTS AND METHODS The primary end point was the progression-free survival rate (PFR) at 12 weeks. Forty eligible patients have been recruited after failure of platinum chemotherapy and cetuximab. A preplanned futility analysis was successfully passed after ≥1 success was detected in 20 patients. Secondary objectives consisted of progression-free survival (PFS), disease control rate (DCR), overall survival (OS), safety and tolerability, and predictive biomarkers for KRAS, BRAF, PIK3CA mutations, and HPV status. Archived tumor tissue was analyzed for DNA sequence. RESULTS A total of 40 patients were eligible. The PFR at 12 weeks was 40% (95% CI 25.0-54.6). The median PFS and OS were 56 days (95% CI 36-113 days) and 152 days (76-256 days), respectively. In 33 assessable patients, disease stabilization occurred in 57.6%, with tumor shrinkage in 13 patients (39.4%). Overall, the treatment was well tolerated. Fatigue (47.5%), anemia (25.0%), nausea (20.0%), and pneumonia (20.0%) were the most common adverse events. Neither PIK3CA mutations, nor HPV status were predictive for success with temsirolimus treatment. No mutations were found for KRAS or BRAF. CONCLUSION Tumor shrinkage and efficacy parameter indicate that inhibition of the PI3K-AKT-mTOR axis was a putative novel treatment paradigm for SCCHN. We could not identify parameters predictive for treatment success of temsirolimus, which underscores the need for refinement of the molecular analysis in future studies. CLINICAL TRIALS NUMBER NCT01172769.
Collapse
Affiliation(s)
- V Grünwald
- Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover
| | - U Keilholz
- Charité Comprehensive Cancer Center, Berlin
| | - A Boehm
- Clinic and Policlinic for Ear, Nose and Throat, University Hospital, Leipzig
| | | | - B Hennemann
- Department for Hematology and Oncology, Ev. Bethesda- Johanniter Hospital, Duisburg
| | - H J Schmoll
- Clinic for Internal Medicine IV, University Hospital, Halle
| | - P Ivanyi
- Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover
| | | | | | | | | | - A Zörner
- Clinical Pharmacology, Hannover Medical School, Hannover
| | - T C Gauler
- West-German Cancer Center, University Hospital, Essen, Germany
| |
Collapse
|
17
|
Simpson DR, Mell LK, Cohen EEW. Targeting the PI3K/AKT/mTOR pathway in squamous cell carcinoma of the head and neck. Oral Oncol 2014; 51:291-8. [PMID: 25532816 DOI: 10.1016/j.oraloncology.2014.11.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/27/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023]
Abstract
Despite recent advances in novel therapies, the prognosis for patients with squamous cell carcinoma of the head and neck (SCCHN) remains poor. Progress in understanding the biology of cancer has led to the development of personalized therapy targeted at blocking defective signaling pathways of cancer cells. These drugs aim to act selectively to reduce the adverse effects associated with systemic therapy. Cetuximab (Erbitux®), an anti-epidermal growth factor receptor gene (EGFR)-targeted agent, is the only approved targeted therapy for patients with SCCHN. However, resistance to EGFR therapy remains a major obstacle to achieving a positive clinical outcome with cetuximab. Other therapies that offer better clinical outcomes in patients with advanced SCCHN are urgently needed. The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin pathway, which is downstream of EGFR, has also been implicated in SCCHN development and progression, and therefore, targeting this pathway offers another rational treatment approach. This review discusses the potential role of PI3K pathway inhibitors in the treatment of patients with advanced SCCHN, both alone and in combination with other therapies.
Collapse
Affiliation(s)
- Daniel R Simpson
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Ezra E W Cohen
- Department of Internal Medicine, Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, United States.
| |
Collapse
|
18
|
Cortelazzi B, Verderio P, Ciniselli CM, Pizzamiglio S, Bossi P, Gloghini A, Gualeni AV, Volpi CC, Locati L, Pierotti MA, Licitra L, Pilotti S, Perrone F. Receptor tyrosine kinase profiles and human papillomavirus status in oropharyngeal squamous cell carcinoma. J Oral Pathol Med 2014; 44:734-45. [DOI: 10.1111/jop.12301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Barbara Cortelazzi
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Paolo Verderio
- Unit of Medical Statistics Biometry and Bioinformatics Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Chiara Maura Ciniselli
- Unit of Medical Statistics Biometry and Bioinformatics Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Sara Pizzamiglio
- Unit of Medical Statistics Biometry and Bioinformatics Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Paolo Bossi
- Head and Neck Cancer Medical Oncology Unit Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Annunziata Gloghini
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Ambra V. Gualeni
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Chiara C. Volpi
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Laura Locati
- Head and Neck Cancer Medical Oncology Unit Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | | | - Lisa Licitra
- Head and Neck Cancer Medical Oncology Unit Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Federica Perrone
- Laboratory of Experimental Molecular Pathology Department of Pathology Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| |
Collapse
|
19
|
Abstract
Doublet chemotherapy with cisplatin is the reference for the treatment of recurrent cervical cancer. However, those tumors are little chemo-sensitive and overall survival remains poor. Moreover, because of pelvic irradiation, toxicities, especially hematologic toxicities, are increased and require a drug dose reduction. Finally, these treatments are rarely effective in radiation areas. Given all these elements, the development of new therapies is a prominent issue. This article reviews the results of the major targeted therapies in cervical cancer. Anti-EGFRs are disappointing despite of a strong biological rational. On the other hand, bevacizumab is the first targeted therapy to show a significant increase of overall survival. A major effort must be made in translational research for a better understanding of tumor biology of these tumors.
Collapse
|
20
|
Mountzios G, Rampias T, Psyrri A. The mutational spectrum of squamous-cell carcinoma of the head and neck: targetable genetic events and clinical impact. Ann Oncol 2014; 25:1889-1900. [PMID: 24718888 DOI: 10.1093/annonc/mdu143] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Squamous-cell cancer of the head and neck (SCCHN) represents a heterogeneous disease entity, with various etiological factors implicated in the genesis of distinct molecular subsets of tumors, which exhibit different biological and clinical behavior. Treatment of SCCHN is expected to change in the next decade as targeted therapies continue to make strides. Recently, next-generation sequencing studies conducted on ∼190 SCCHN specimens shed light into the molecular pathogenesis of the disease. These studies discovered mutations in genes involved in the differentiation program of squamous epithelium and the Notch/p63 axis (such as NOTCH1, TP63 and FBXW7), and validated genetic alterations derived from previous studies (such as mutations in TP53, CDKN2A, PIK3CA, CCND1 and HRAS) as driver genetic events in SCCHN neoplastic transformation. More recently, comprehensive data from The Cancer Genome Atlas (TCGA) project on 306 SCCHN specimens provided further insight into SCCHN inherent molecular complexity, identifying novel significantly mutated genes, including FAT1, MLL2, TGFRBR2, HLA-A, NFE2l2 and CASP8. In this article, we provide an overview of the mutational spectrum of SCCHN, with emphasis on the clinical implementation of this knowledge. We also discuss the potential integration of new data within the framework of precision cancer medicine.
Collapse
Affiliation(s)
- G Mountzios
- Department of Medical Oncology, University of Athens School of Medicine, Athens, Greece
| | - T Rampias
- Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven,USA
| | - A Psyrri
- Second Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, Athens, Greece; Department of Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven,USA.
| |
Collapse
|
21
|
Iglesias-Bartolome R, Martin D, Gutkind JS. Exploiting the head and neck cancer oncogenome: widespread PI3K-mTOR pathway alterations and novel molecular targets. Cancer Discov 2014; 3:722-5. [PMID: 23847349 DOI: 10.1158/2159-8290.cd-13-0239] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SUMMARY Two studies published in this issue of Cancer Discovery describe the emerging mutational landscape of head and neck squamous cell carcinomas (HNSCC) and their genomic and epigenetic alterations, thus identifying novel actionable cancer drivers and predictive biomarkers for targeted therapies. Most genomic alterations in HNSCC converge in a handful of molecular pathways, resulting in cell-cycle deregulation, genomic instability, cell differentiation defects, and persistent mitogenic signaling, the latter involving aberrant phosphoinositide 3-kinase (PI3K)/mTOR pathway activation, thereby rendering HNSCC responsive to PI3K/mTOR inhibitors. Cancer Discov; 3(7); 722-5. ©2013 AACR.
Collapse
Affiliation(s)
- Ramiro Iglesias-Bartolome
- Oral and Pharyngeal Cancer Branch, National Institute of Dental Research, NIH, Bethesda, MD 20892-4330, USA
| | | | | |
Collapse
|
22
|
TRAIL combinations: The new 'trail' for cancer therapy (Review). Oncol Lett 2014; 7:1327-1332. [PMID: 24765133 PMCID: PMC3997674 DOI: 10.3892/ol.2014.1922] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 01/21/2014] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) therapy is anticipated to be one of the most effective cancer treatments. However, resistance to TRAIL therapy remains a challenge facing the development of anticancer strategies. To circumvent this problem, TRAIL combinations have been experimented with for over ten years to induce synergism or sensitize resistant cancer cells. By analyzing the signaling pathways triggered by these combinations, this review has defined a set of core targets for novel combinatorial treatments. The review suggests specific pathways to be targeted together with TRAIL for more efficient treatment, including cellular FLICE inhibitory protein and its downstream survival factors, the Bcl-2 family and other prominent targets. The suggested pathways provide new avenues for more effective TRAIL-based cancer therapy.
Collapse
|
23
|
Fury MG, Lee NY, Sherman E, Ho AL, Rao S, Heguy A, Shen R, Korte S, Lisa D, Ganly I, Patel S, Wong RJ, Shaha A, Shah J, Haque S, Katabi N, Pfister DG. A phase 1 study of everolimus + weekly cisplatin + intensity modulated radiation therapy in head-and-neck cancer. Int J Radiat Oncol Biol Phys 2013; 87:479-86. [PMID: 24074921 DOI: 10.1016/j.ijrobp.2013.06.2043] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/14/2013] [Accepted: 06/18/2013] [Indexed: 11/17/2022]
Abstract
PURPOSE Elevated expression of eukaryotic protein synthesis initiation factor 4E (eIF4E) in histologically cancer-free margins of resected head and neck squamous cell carcinomas (HNSCCs) is mediated by mammalian target of rapamycin complex 1 (mTORC1) and has been associated with increased risk of disease recurrence. Preclinically, inhibition of mTORC1 with everolimus sensitizes cancer cells to cisplatin and radiation. METHODS AND MATERIALS This was single-institution phase 1 study to establish the maximum tolerated dose of daily everolimus given with fixed dose cisplatin (30 mg/m(2) weekly × 6) and concurrent intensity modulated radiation therapy for patients with locally and/or regionally advanced head-and-neck cancer. The study had a standard 3 + 3 dose-escalation design. RESULTS Tumor primary sites were oral cavity (4), salivary gland (4), oropharynx (2), nasopharynx (1), scalp (1), and neck node with occult primary (1). In 4 of 4 cases in which resected HNSCC surgical pathology specimens were available for immunohistochemistry, elevated expression of eIF4E was observed in the cancer-free margins. The most common grade ≥3 treatment-related adverse event was lymphopenia (92%), and dose-limiting toxicities (DLTs) were mucositis (n=2) and failure to thrive (n=1). With a median follow up of 19.4 months, 2 patients have experienced recurrent disease. The maximum tolerated dose was everolimus 5 mg/day. CONCLUSIONS Head-and-neck cancer patients tolerated everolimus at therapeutic doses (5 mg/day) given with weekly cisplatin and intensity modulated radiation therapy. The regimen merits further evaluation, especially among patients who are status post resection of HNSCCs that harbor mTORC1-mediated activation of eIF4E in histologically negative surgical margins.
Collapse
Affiliation(s)
- Matthew G Fury
- Department of Medicine, Head and Neck Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Combination therapy with bevacizumab and temsirolimus in squamous cell carcinoma of the head and neck. Oral Oncol 2013; 49:e25. [DOI: 10.1016/j.oraloncology.2013.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 04/09/2013] [Indexed: 11/23/2022]
|
25
|
Lui VWY, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, Freilino M, Sauerwein S, Peyser ND, Xiao D, Diergaarde B, Wang L, Chiosea S, Seethala R, Johnson JT, Kim S, Duvvuri U, Ferris RL, Romkes M, Nukui T, Kwok-Shing Ng P, Garraway LA, Hammerman PS, Mills GB, Grandis JR. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov 2013; 3:761-9. [PMID: 23619167 DOI: 10.1158/2159-8290.cd-13-0103] [Citation(s) in RCA: 455] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Genomic findings underscore the heterogeneity of head and neck squamous cell carcinoma (HNSCC). Identification of mutations that predict therapeutic response would be a major advance. We determined the mutationally altered, targetable mitogenic pathways in a large HNSCC cohort. Analysis of whole-exome sequencing data from 151 tumors revealed the phosphoinositide 3-kinase (PI3K) pathway to be the most frequently mutated oncogenic pathway (30.5%). PI3K pathway-mutated HNSCC tumors harbored a significantly higher rate of mutations in known cancer genes. In a subset of human papillomavirus-positive tumors, PIK3CA or PIK3R1 was the only mutated cancer gene. Strikingly, all tumors with concurrent mutation of multiple PI3K pathway genes were advanced (stage IV), implicating concerted PI3K pathway aberrations in HNSCC progression. Patient-derived tumorgrafts with canonical and noncanonical PIK3CA mutations were sensitive to an mTOR/PI3K inhibitor (BEZ-235), in contrast to PIK3CA-wild-type tumorgrafts. These results suggest that PI3K pathway mutations may serve as predictive biomarkers for treatment selection.
Collapse
Affiliation(s)
- Vivian W Y Lui
- Departments of 1Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|