1
|
Jasim SA, Al-Hawary SIS, Kaur I, Ahmad I, Hjazi A, Petkov I, Ali SHJ, Redhee AH, Shuhata Alubiady MH, Al-Ani AM. Critical role of exosome, exosomal non-coding RNAs and non-coding RNAs in head and neck cancer angiogenesis. Pathol Res Pract 2024; 256:155238. [PMID: 38493725 DOI: 10.1016/j.prp.2024.155238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/13/2024] [Accepted: 03/02/2024] [Indexed: 03/19/2024]
Abstract
Head and neck cancer (HNC) refers to the epithelial malignancies of the upper aerodigestive tract. HNCs have a constant yet slow-growing rate with an unsatisfactory overall survival rate globally. The development of new blood vessels from existing blood conduits is regarded as angiogenesis, which is implicated in the growth, progression, and metastasis of cancer. Aberrant angiogenesis is a known contributor to human cancer progression. Representing a promising therapeutic target, the blockade of angiogenesis aids in the reduction of the tumor cells oxygen and nutrient supplies. Despite the promise, the association of existing anti-angiogenic approaches with severe side effects, elevated cancer regrowth rates, and limited survival advantages is incontrovertible. Exosomes appear to have an essential contribution to the support of vascular proliferation, the regulation of tumor growth, tumor invasion, and metastasis, as they are a key mediator of information transfer between cells. In the exocrine region, various types of noncoding RNAs (ncRNAs) identified to be enriched and stable and contribute to the occurrence and progression of cancer. Mounting evidence suggest that exosome-derived ncRNAs are implicated in tumor angiogenesis. In this review, the characteristics of angiogenesis, particularly in HNC, and the impact of ncRNAs on HNC angiogenesis will be outlined. Besides, we aim to provide an insight on the regulatory role of exosomes and exosome-derived ncRNAs in angiogenesis in different types of HNC.
Collapse
Affiliation(s)
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Iliya Petkov
- Medical University - Sofia, Department of Neurology, Sofia, Bulgaria
| | - Saad Hayif Jasim Ali
- Department of medical laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | | | | |
Collapse
|
2
|
den bossche VV, Zaryouh H, Vara-Messler M, Vignau J, Machiels JP, Wouters A, Schmitz S, Corbet C. Microenvironment-driven intratumoral heterogeneity in head and neck cancers: clinical challenges and opportunities for precision medicine. Drug Resist Updat 2022; 60:100806. [DOI: 10.1016/j.drup.2022.100806] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
|
3
|
Willenbrock F, Cox CM, Parkes EE, Wilhelm-Benartzi CS, Abraham AG, Owens R, Sabbagh A, Jones CM, Hughes DLI, Maughan T, Hurt CN, O'Neill EE, Mukherjee S. Circulating biomarkers and outcomes from a randomised phase 2 trial of gemcitabine versus capecitabine-based chemoradiotherapy for pancreatic cancer. Br J Cancer 2021; 124:581-586. [PMID: 33100327 PMCID: PMC7851394 DOI: 10.1038/s41416-020-01120-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Phase 2 SCALOP trial compared gemcitabine with capecitabine-based consolidation chemoradiotherapy (CRT) in locally advanced pancreatic cancer (LAPC). METHODS Thirty-five systematically identified circulating biomarkers were analysed in plasma samples from 60 patients enroled in SCALOP. Each was measured in triplicate at baseline (prior to three cycles of gemcitabine-capecitabine induction chemotherapy) and, for a subset, prior to CRT. Association with overall survival (OS) was determined using univariable Cox regression and optimal thresholds delineating low to high values identified using time-dependent ROC curves. Independence from known prognostic factors was assessed using Spearman correlation and the Wilcoxon rank sum test prior to multivariable Cox regression modelling including independent biomarkers and known prognostic factors. RESULTS Baseline circulating levels of C-C motif chemokine ligand 5 (CCL5) were significantly associated with OS, independent of other clinicopathological characteristics. Patients with low circulating CCL5 (CCL5low) had a median OS of 18.5 (95% CI 11.76-21.32) months compared to 11.3 (95% CI 9.86-15.51) months in CCL5high; hazard ratio 1.95 (95% CI 1.04-8.65; p = 0.037). CONCLUSIONS CCL5 is an independent prognostic biomarker in LAPC. Given the known role of CCL5 in tumour invasion, metastasis and the induction of an immunosuppressive micro-environment, targeting of CCL5-mediated pathways may offer therapeutic potential in pancreatic cancer. CLINICAL TRIAL REGISTRATION The SCALOP trial was registered with ISRCTN, number 96169987 (registered 29 May 2008).
Collapse
Affiliation(s)
| | | | | | | | | | - Robert Owens
- Oxford University Hospital NHS Trust, Oxford, UK
| | - Ahmad Sabbagh
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | | | - Tim Maughan
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Eric E O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | | |
Collapse
|
4
|
Jannier S, Kemmel V, Sebastia Sancho C, Chammas A, Sabo AN, Pencreach E, Farace F, Chenard MP, Lhermitte B, Geoerger B, Aerts I, Frappaz D, Leblond P, André N, Ducassou S, Corradini N, Bertozzi AI, Guérin E, Vincent F, Velten M, Entz-Werle N. SFCE-RAPIRI Phase I Study of Rapamycin Plus Irinotecan: A New Way to Target Intra-Tumor Hypoxia in Pediatric Refractory Cancers. Cancers (Basel) 2020; 12:cancers12103051. [PMID: 33092063 PMCID: PMC7656302 DOI: 10.3390/cancers12103051] [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: 08/21/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary More and more relapsing or refractory pediatric cancers are described to present hypoxic features linked to a worse outcome. Therefore, the aim of our phase I study RAPIRI was the targeting of the central node mTor/HIF-1α with rapamycin plus irinotecan and determine the appropriated dose of this combination. As expected, the tolerance was optimal across all dose levels and no maximum tolerated dose of both drugs was reached. The pharmacokinetics (PK) helped us to refine the doses to use in the future phase II trial and the importance of PK follow-up in such combination. We also confirmed in almost half of the interpretable patients for tumor response a non-progressive disease. All those observations additionally to the ancillary’s studies provide strong evidence to propose a next trial focusing on brain tumors and sarcomas and using biweekly 125 mg/m2 irinotecan dose with a PK follow-up and a rapamycin dose of 1.5 mg/m2/day, reaching a blood concentration above 10 µg/L. Abstract Hypoxic environment is a prognostic factor linked in pediatric cancers to a worse outcome, favoring tumor progression and resistance to treatments. The activation of mechanistic Target Of Rapamycin (mTor)/hypoxia inducible factor (HIF)-1α pathway can be targeted by rapamycin and irinotecan, respectively. Therefore, we designed a phase I trial associating both drugs in pediatric refractory/relapsing solid tumors. Patients were enrolled according to a 3 + 3 escalation design with ten levels, aiming to determine the MTD (maximum tolerated dose) of rapamycin plus irinotecan. Rapamycin was administered orally once daily in a 28-day cycle (1 to 2.5 mg/m2/day), associating biweekly intravenous irinotecan (125 to 240 mg/m2/dose). Toxicities, pharmacokinetics, efficacy analyses, and pharmacodynamics were evaluated. Forty-two patients, aged from 2 to 18 years, were included. No MTD was reached. Adverse events were mild to moderate. Only rapamycin doses of 1.5 mg/m2/day reached over time clinically active plasma concentrations. Tumor responses and prolonged stable disease were associated with a mean irinotecan area under the curve of more than 400 min.mg/L. Fourteen out of 31 (45.1%) patients had a non-progressive disease at 8 weeks. Most of them were sarcomas and brain tumors. For the phase II trial, we can then propose biweekly 125 mg/m2 irinotecan dose with a pharmacokinetic (PK) follow-up and a rapamycin dose of 1.5 mg/m2/day, reaching a blood concentration above 10 µg/L.
Collapse
Affiliation(s)
- Sarah Jannier
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
| | - Véronique Kemmel
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
- Laboratory of Pharmacology and Toxicology in Neurocardiology-EA7296, University of Strasbourg, 67000 Strasbourg, France
| | - Consuelo Sebastia Sancho
- Radiology Department, Pediatric Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (C.S.S.); (A.C.)
| | - Agathe Chammas
- Radiology Department, Pediatric Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (C.S.S.); (A.C.)
| | - Amelia-Naomie Sabo
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
- Laboratory of Pharmacology and Toxicology in Neurocardiology-EA7296, University of Strasbourg, 67000 Strasbourg, France
| | - Erwan Pencreach
- Oncobiology Platform, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Françoise Farace
- «Circulating Tumor Cells» Translational Platform, Gustave Roussy, University of Paris-Saclay, 94800 Villejuif, France;
| | - Marie Pierre Chenard
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France; (M.P.C.); (B.L.)
- Centre de Ressources Biologiques, University Hospital of Strasbourg, 67098 Strasbourg, France
| | - Benoit Lhermitte
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France; (M.P.C.); (B.L.)
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, Université Paris-Saclay, INSERM U1015, 94800 Villejuif, France;
| | - Isabelle Aerts
- Oncology Center SIREDO, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Didier Frappaz
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
| | - Pierre Leblond
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
- Pediatric Oncology Unit, Oscar Lambret Center, 59020 Lille, France
| | - Nicolas André
- Pediatric Onco-Hematology Unit, CHU La Timone, 13005 Marseille, France;
| | - Stephane Ducassou
- Pediatric Onco-Hematology Department, University Hospital of Bordeaux, 33000 Bordeaux, France;
| | - Nadège Corradini
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
- Pediatric Oncology Unit, University Hospital of Nantes, 44093 Nantes, France
| | - Anne Isabelle Bertozzi
- Pediatric Onco-Hematology Department, University Hospital of Toulouse, 31059 Toulouse, France;
| | - Eric Guérin
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
| | - Florence Vincent
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
| | - Michel Velten
- Clinical Research Department, ICANS, 67200 Strasbourg, France;
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets, Faculty of Pharmacy, 67401 Illkirch, France
- Correspondence: ; Tel.: +33-3-88-12-83-96
| |
Collapse
|
5
|
Sørensen BS, Horsman MR. Tumor Hypoxia: Impact on Radiation Therapy and Molecular Pathways. Front Oncol 2020; 10:562. [PMID: 32373534 PMCID: PMC7186437 DOI: 10.3389/fonc.2020.00562] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/30/2020] [Indexed: 01/25/2023] Open
Abstract
Tumor hypoxia is a common feature of the microenvironment in solid tumors, primarily due to an inadequate, and heterogeneous vascular network. It is associated with resistance to radiotherapy and results in a poorer clinical outcome. The presence of hypoxia in tumors can be identified by various invasive and non-invasive techniques, and there are a number of approaches by which hypoxia can be modified to improve outcome. However, despite these factors and the ongoing extensive pre-clinical studies, the clinical focus on hypoxia is still to a large extent lacking. Hypoxia is a major cellular stress factor and affects a wide range of molecular pathways, and further understanding of the molecular processes involved may lead to greater clinical applicability of hypoxic modifiers. This review is a discussion of the characteristics of tumor hypoxia, hypoxia-related molecular pathways, and the role of hypoxia in treatment resistance. Understanding the molecular aspects of hypoxia will improve our ability to clinically monitor hypoxia and to predict and modify the therapeutic response.
Collapse
Affiliation(s)
- Brita Singers Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael R Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
6
|
Randomized phase II trial of cixutumumab alone or with cetuximab for refractory recurrent/metastatic head and neck squamous cell carcinoma. Oral Oncol 2018; 82:83-90. [PMID: 29909907 DOI: 10.1016/j.oraloncology.2018.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Cixutumumab (CIX) and cetuximab (CET) monoclonal antibodies block ligand-binding to insulin-like growth factor-1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) respectively. The objective of this study was to assess the efficacy of CIX alone or combined with CET in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) patients. METHODS In this open-label phase II trial, 91 R/M HNSCC patients who progressed within 90 days of platinum-based chemotherapy, were randomized to CIX 10 mg/kg alone or with CET 500 mg/m2 every 2 weeks. Patients were stratified by prior CET use. The primary endpoint was median progression-free survival (PFS). Exploratory biomarker assessments included relevant markers on archival tumor and serial cytokine/angiogenic-factor profiles in blood. RESULTS Forty-seven patients were treated with CIX monotherapy and 44 with combination. The median PFS was 1.9 and 2.0 months and clinical benefit rate (complete or partial responses and stable disease) was 5.9% and 15.3%, respectively. There was no exacerbation of CET toxicity by concurrent CIX exposure. Higher tumor expression of IGF-1 was associated with improved PFS in the CIX + CET arm while increased p-EGFR expression correlated with shorter PFS in patients receiving single agent CIX. Higher serum baseline levels of IGF-1 and IGFBP-3 correlated with improved PFS and overall survival (OS) in the CIX arm. Neither regimen resulted in improved PFS or OS compared to historical data with CET alone. CONCLUSION The results of this study do not support the use of cixutumumab alone or with cetuximab in unselected patients with R/M HNSCC.
Collapse
|
7
|
Jiffar T, Yilmaz T, Lee J, Miller Y, Feng L, El-Naggar A, Kupferman ME. Brain derived neutrophic factor (BDNF) coordinates lympho-vascular metastasis through a fibroblast-governed paracrine axis in the tumor microenvironment. ACTA ACUST UNITED AC 2017; 4. [PMID: 28966935 PMCID: PMC5617346 DOI: 10.14800/ccm.1566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has long been known that the tumor microenvironment contributes to the proliferation and survival of neoplasms through the constant interaction with the stromal and immune compartments. In this investigation, we explored the role of cancer-associated fibroblasts (CAFs) in the regulation of the tumor microenvironment in head and neck squamous cell carcinoma (HNSCC) though a complex intercellular BDNF-TrkB signaling system. Our studies show that conditioned media derived from patient-derived CAFs promoted HNSCC cell proliferation, in vitro cell migration, cell invasion and chemotherapy resistance, compared to normal fibroblasts. Furthermore, examination of the in vivo impact of CAF pathophysiology in the tumor microenvironment in animal xenograft models revealed that HNSCC cell lines in combination with CAFs promoted tumor growth and increased incidence of lymphovascular metastasis as compared to injection of tumor cells or CAF cells alone. Using pharmacological and genetic alterations, we mechanistically demonstrate the critical importance of BDNF-TrkB signaling in the tumor microenvironment. These investigations further support the rationale for BDNF/TRKB targeted therapy against in the treatment of HNSCC.
Collapse
Affiliation(s)
- Tilahun Jiffar
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Turker Yilmaz
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Junegoo Lee
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Yair Miller
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Lei Feng
- Department of Biostatistics, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Adel El-Naggar
- Department of Pathology, MD Anderson Cancer Center, Houston TX 77030, USA
| | - Michael E Kupferman
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston TX 77030, USA
| |
Collapse
|
8
|
Weidhaas JB, Harris J, Schaue D, Chen AM, Chin R, Axelrod R, El-Naggar AK, Singh AK, Galloway TJ, Raben D, Wang D, Matthiesen C, Avizonis VN, Manon RR, Yumen O, Nguyen-Tan PF, Trotti A, Skinner H, Zhang Q, Ferris RL, Sidransky D, Chung CH. The KRAS-Variant and Cetuximab Response in Head and Neck Squamous Cell Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol 2017; 3:483-491. [PMID: 28006059 DOI: 10.1001/jamaoncol.2016.5478] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Importance There is a significant need to find biomarkers of response to radiotherapy and cetuximab in locally advanced head and neck squamous cell carcinoma (HNSCC) and biomarkers that predict altered immunity, thereby enabling personalized treatment. Objectives To examine whether the Kirsten rat sarcoma viral oncogene homolog (KRAS)-variant, a germline mutation in a microRNA-binding site in KRAS, is a predictive biomarker of cetuximab response and altered immunity in the setting of radiotherapy and cisplatin treatment and to evaluate the interaction of the KRAS-variant with p16 status and blood-based transforming growth factor β1 (TGF-β1). Design, Setting, and Participants A total of 891 patients with advanced HNSCC from a phase 3 trial of cisplatin plus radiotherapy with or without cetuximab (NRG Oncology RTOG 0522) were included in this study, and 413 patients with available samples were genotyped for the KRAS-variant. Genomic DNA was tested for the KRAS-variant in a CLIA-certified laboratory. Correlation of the KRAS-variant, p16 positivity, outcome, and TGF-β1 levels was evaluated. Hazard ratios (HRs) were estimated with the Cox proportional hazards model. Main Outcomes and Measures The correlation of KRAS-variant status with cetuximab response and outcome, p16 status, and plasma TGF-β1 levels was tested. Results Of 891 patients eligible for protocol analyses (786 male [88.2%], 105 [11.2%] female, 810 white [90.9%], 81 nonwhite [9.1%]), 413 had biological samples for KRAS-variant testing, and 376 had plasma samples for TGF-β1 measurement. Seventy patients (16.9%) had the KRAS-variant. Overall, for patients with the KRAS-variant, cetuximab improved both progression-free survival (PFS) for the first year (HR, 0.31; 95% CI, 0.10-0.94; P = .04) and overall survival (OS) in years 1 to 2 (HR, 0.19; 95% CI, 0.04-0.86; P = .03). There was a significant interaction of the KRAS-variant with p16 status for PFS in patients treated without cetuximab. The p16-positive patients with the KRAS-variant treated without cetuximab had worse PFS than patients without the KRAS-variant (HR, 2.59; 95% CI, 0.91-7.33; P = .07). There was a significant 3-way interaction among the KRAS-variant, p16 status, and treatment for OS (HR, for KRAS-variant, cetuximab and p16 positive, 0.22; 95% CI, 0.03-1.66; HR for KRAS-variant, cetuximab and p16 negative, 1.43; 95% CI, 0.48-4.26; HR for KRAS-variant, no cetuximab and p16 positive, 2.48; 95% CI, 0.64-9.65; and HR for KRAS-variant, no cetuximab and p16 negative, 0.61; 95% CI, 0.23-1.59; P = .02). Patients with the KRAS-variant had significantly elevated TGF-β1 plasma levels (median, 23 376.49 vs 18 476.52 pg/mL; P = .03) and worse treatment-related toxic effects. Conclusions and Relevance Patients with the KRAS-variant with HNSCC significantly benefit from the addition of cetuximab to radiotherapy and cisplatin, and there is a significant interaction between the KRAS-variant and p16 status. Elevated TGF-β1 levels in patients with the KRAS-variant suggests that cetuximab may help these patients by overcoming TGF-β1-induced suppression of antitumor immunity. Trial Registration clinicaltrials.gov Identifier: NCT00265941.
Collapse
Affiliation(s)
- Joanne B Weidhaas
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA (University of California, Los Angeles), Los Angeles, California
| | - Jonathan Harris
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA (University of California, Los Angeles), Los Angeles, California
| | - Allen M Chen
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA (University of California, Los Angeles), Los Angeles, California
| | - Robert Chin
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA (University of California, Los Angeles), Los Angeles, California
| | - Rita Axelrod
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adel K El-Naggar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | | | | | - David Raben
- Department of Radiation Oncology, University of Colorado at Denver, Aurora
| | - Dian Wang
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee
| | - Chance Matthiesen
- Department of Radiation Oncology, Oklahoma University Health Sciences Center, Oklahoma City
| | - Vilija N Avizonis
- Department of Radiation Oncology, Intermountain Medical Center, Salt Lake City, Utah
| | - Rafael R Manon
- University of Florida Health Cancer Center, Orlando Health, Orlando
| | - Omar Yumen
- Department of Radiation Oncology, Geisinger Medical Center CCOP, Danville, Pennsylvania
| | - Phuc Felix Nguyen-Tan
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montreal, Montreal, Quebec, Canada
| | - Andy Trotti
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Heath Skinner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Qiang Zhang
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Robert L Ferris
- Cancer Immunology Program and Tumor Microvenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
9
|
Brøndum L, Eriksen JG, Singers Sørensen B, Mortensen LS, Toustrup K, Overgaard J, Alsner J. Plasma proteins as prognostic biomarkers in radiotherapy treated head and neck cancer patients. Clin Transl Radiat Oncol 2017; 2:46-52. [PMID: 29658000 PMCID: PMC5893530 DOI: 10.1016/j.ctro.2017.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/27/2016] [Accepted: 01/03/2017] [Indexed: 01/03/2023] Open
Abstract
Background Blood-based protein biomarkers can be a useful tool as pre-treatment prognostic markers, as they can reflect both variations in the tumor microenvironment and the host immune response. We investigated the influence of a panel of plasma proteins for the development of any failure defined as recurrent disease in the T-, N-, or M-site in HNSCC. Methods We used a multiplex bead-based approach to analyze 19 proteins in 86 HNSCC patients and 15 healthy controls. We evaluated the associations between the biomarkers, loco-regional failure, failure in the T-, N-, or M-site, overall survival (OS), p16 status, and hypoxia. Results In 41 p16 positive oropharynx cancer patients we identified a profile of biomarkers consisting of upregulation of IL-2, IL-4, IL-6, IL-8, eotaxin, GRO-a, and VEGF and downregulation of VEGFR-1 and VEGFR-2 with a significantly reduced risk of failure (p < 0.01). None of the individual proteins were associated with outcome. Conclusion The identified plasma profile potentially reflects an activated immune response in a subgroup of the p16 positive patients.
Collapse
Affiliation(s)
- Line Brøndum
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Brita Singers Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Kasper Toustrup
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
10
|
Welsh L, Panek R, Riddell A, Wong K, Leach MO, Tavassoli M, Rahman D, Schmidt M, Hurley T, Grove L, Richards T, Koh DM, Nutting C, Harrington K, Newbold K, Bhide S. Blood transfusion during radical chemo-radiotherapy does not reduce tumour hypoxia in squamous cell cancer of the head and neck. Br J Cancer 2017; 116:28-35. [PMID: 27884018 PMCID: PMC5220150 DOI: 10.1038/bjc.2016.386] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/08/2016] [Accepted: 10/25/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Patients with head and neck squamous cell carcinoma (HNSCC) undergoing radical chemo-radiation (CRT) frequently receive transfusion with packed red cells (PRCT) during radiotherapy on the basis that PRCT increases tumour oxygenation and overcomes hypoxia-induced radio-resistance. This is likely to be a significant oversimplification given the fact that tumour hypoxia is the result of several intrinsic and extrinsic factors, including many that are not directly related to serum haemoglobin (Hb). Therefore, we have studied the effect of PRCT on tumour oxygenation in a prospective cohort of patients who developed low Hb during radical CRT for HNSCC. METHODS This was a prospective study of 20 patients with HNSCC receiving radical CRT undergoing PRCT for Hb<11.5 g dl-1. Patients underwent pretransfusion and posttransfusion intrinsic susceptibility-weighted (SWI) MRI and dynamic contrast-enhanced (DCE) MRI. Blood samples were obtained at the time of MRI scanning and two further time points for measuring Hb and a panel of serum cytokine markers of tumour hypoxia. 3D T2* and Ktrans maps were calculated from the MRI data for primary tumours and cervical lymph node metastases. RESULTS PRCT produced no change (11 patients) or reduced (1 patient) T2* (tumour oxygenation) in 12 of the 16 (75%) evaluable primary tumours. Three of the four patients with improved tumour oxygenation progressed or had partial response following treatment completion. There were variable changes in Ktrans (tumour perfusion or vessel permeability) following PRCT that were of small magnitude for most tumours. Pre- and Post-PRCT levels of measured cytokines were not significantly different. CONCLUSIONS This study suggests that PRCT during radical CRT for HNSCC does not improve tumour oxygenation. Therefore, oncologists should consider changing practice according to NICE and American Association of Blood Banks guidelines on PRCT for anaemia.
Collapse
Affiliation(s)
- Liam Welsh
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Rafal Panek
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Angela Riddell
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
| | - Kee Wong
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Martin O Leach
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Mahvash Tavassoli
- King's College, Floor 2, Hodgkin Building, Guy's, London SE1 9RT, UK
| | - Durdana Rahman
- King's College, Floor 2, Hodgkin Building, Guy's, London SE1 9RT, UK
| | - Maria Schmidt
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Tara Hurley
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
| | - Lorna Grove
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
| | - Thomas Richards
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Dow-Mu Koh
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Christopher Nutting
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Kevin Harrington
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Kate Newbold
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Shreerang Bhide
- The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Royal Marsde Hospital, Downs Road, Sutton SM2 5PT, UK
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| |
Collapse
|
11
|
Signature of cytokines and angiogenic factors (CAFs) defines a clinically distinct subgroup of gastric cancer. Gastric Cancer 2017; 20:164-174. [PMID: 26681196 DOI: 10.1007/s10120-015-0583-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Little is known about cytokine and angiogenic factors (CAFs) in gastric cancer (GC) in terms of tumor classification and prognostic value. Here, we aimed to correlate CAF signature with overall survival (OS) in GC. METHODS We measured pretreatment serum levels of 52 kinds of CAFs in 68 GC patients who were treated with fluoropyrimidine and platinum combination chemotherapy using multiplex bead immunoassays and enzyme-linked immunosorbent assay. We evaluated correlations between CAF levels and pathological features and OS. RESULTS Three distinct patient groups were identified: one with high levels of proangiogenic factors, another with high levels of proinflammatory factors, and the other with high levels of both factors. Eleven CAFs [interleukin (IL)-2 receptor-alpha, growth-regulated alpha protein, hepatocyte growth factor, macrophage colony-stimulating factor, stromal cell-derived factor, IL-6, IL-8, IL-10, interferon-gamma, vascular endothelial growth factor, and osteopontin] were independently correlated with poor OS. Clustering analysis of these 11 CAFs revealed distinct high and low 11-CAF signature groups. High 11-CAF signature was associated with shorter OS (10.1 vs. 17.9 months, p = 0.026) along with poor performance status, and the presence of signet ring cell components in multivariate analysis of OS (HR 1.76, p = 0.029). The patients' traditional clinicopathological characteristics were not significantly different between the high and low 11-CAF signature groups. CONCLUSION Serum CAF profiling differentiated GC patient groups. A high 11-CAF signature could identify GC patients with a poor prognosis when treated with standard chemotherapy who need urgent new treatment strategies.
Collapse
|
12
|
Brøndum L, Sørensen BS, Eriksen JG, Mortensen LS, Lønbro S, Overgaard J, Alsner J. An evaluation of multiplex bead-based analysis of cytokines and soluble proteins in archived lithium heparin plasma, EDTA plasma and serum samples. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:601-611. [PMID: 27666533 DOI: 10.1080/00365513.2016.1230882] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To assess the usability of archived plasma and serum by multiplex (Luminex) analysis of circulating proteins (analytes) by evaluating the day to day variation, the effect of several freeze-thaw cycles, and the influence of the media and choice of anticoagulant. METHODS Nineteen analytes in plasma and serum from 86 head and neck cancer patients and 33 controls were evaluated: EGFR, leptin, OPN, VEGFR-1, VEGFR-2, IL-2, IL-13, PDGF-bb, TNF, PAI-1, SDF-1a, IL-4, IL-6, IL-8, eotaxin, G-CSF, VEGF, GRO-a, and HGF. RESULTS The correlation between measurements of the same samples analyzed on different dates was reasonable. However, samples run on different dates could exhibit different absolute values. The 75th percentile of the fold differences for samples run on different dates was 2.2. No significant difference was found between one and four freeze-thaw cycles (except for HGF), and the correlation was high. We found significant differences in mean concentrations of the majority of analytes in different media and with different anticoagulants. Only the following analytes did not show difference in mean concentrations: EDTA plasma vs. serum: leptin and VEGFR-2, LH plasma vs. serum: IL-2, IL-13, and VEGF, LH plasma levels vs. EDTA plasma: IL-2 and IL-4. CONCLUSION Stored serum, LH plasma, and EDTA plasma from clinical trials can be used for analysis of circulating cytokines and proteins. Variations in measurements occur, but are within reasonable ranges. The optimal type of media depends on the analytes, as different analytes have low number of measurements below the lower limit of quantification and higher dynamic ranges in different media.
Collapse
Affiliation(s)
- Line Brøndum
- a Department of Experimental Clinical Oncology , Aarhus University Hospital , Aarhus , Denmark
| | - Brita Singers Sørensen
- a Department of Experimental Clinical Oncology , Aarhus University Hospital , Aarhus , Denmark
| | | | | | - Simon Lønbro
- d Department of Public Health, Section for Sport Science , Aarhus University , Aarhus , Denmark
| | - Jens Overgaard
- a Department of Experimental Clinical Oncology , Aarhus University Hospital , Aarhus , Denmark
| | - Jan Alsner
- a Department of Experimental Clinical Oncology , Aarhus University Hospital , Aarhus , Denmark
| |
Collapse
|
13
|
Lv PC, Putt KS, Low PS. Evaluation of Nonpeptidic Ligand Conjugates for SPECT Imaging of Hypoxic and Carbonic Anhydrase IX-Expressing Cancers. Bioconjug Chem 2016; 27:1762-9. [PMID: 27362480 DOI: 10.1021/acs.bioconjchem.6b00271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As tumors grow, vasculature is often deficient or malformed, resulting in many localized areas of hypoxia. Cells located in these hypoxic regions exhibit an altered gene expression pattern that can significantly alter resistance to conventional anticancer treatments such as ionizing radiation and chemotherapeutic drugs. A priori knowledge of the level of hypoxia within a tumor may better guide clinical care. In an effort to create a hypoxia specific imaging agent, a ligand for the tissue hypoxia marker, carbonic anhydrase IX (CA IX), was synthesized and used as a targeting ligand to deliver an attached (99m)Tc-chelating agent. Binding of the resulting conjugates to hypoxic cancer cells was first characterized in vitro. Whole animal imaging and biodistribution studies then were performed to determine tumor specificity in vivo. Several conjugates were found to bind selectively to CA IX expressing tumors in a receptor-dependent manner. We suggest that such conjugates could prove useful in identifying hypoxic cancers and/or quantitating the level of hypoxia within a tumor.
Collapse
Affiliation(s)
- Peng-Cheng Lv
- Institute for Drug Discovery and ‡Department of Chemistry, Purdue University , West Lafayette, Indiana 47907 United States
| | - Karson S Putt
- Institute for Drug Discovery and ‡Department of Chemistry, Purdue University , West Lafayette, Indiana 47907 United States
| | - Philip S Low
- Institute for Drug Discovery and ‡Department of Chemistry, Purdue University , West Lafayette, Indiana 47907 United States
| |
Collapse
|
14
|
Boeckx C, Van den Bossche J, De Pauw I, Peeters M, Lardon F, Baay M, Wouters A. The hypoxic tumor microenvironment and drug resistance against EGFR inhibitors: preclinical study in cetuximab-sensitive head and neck squamous cell carcinoma cell lines. BMC Res Notes 2015; 8:203. [PMID: 26032726 PMCID: PMC4467624 DOI: 10.1186/s13104-015-1197-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/20/2015] [Indexed: 11/10/2022] Open
Abstract
Background Increased expression of the epidermal growth factor receptor (EGFR) is observed in more than 90% of all head and neck squamous cell carcinomas (HNSCC). Therefore, EGFR has emerged as a promising therapeutic target. Nevertheless, drug resistance remains a major challenge and an important potential mechanism of drug resistance involves the hypoxic tumor microenvironment. Therefore, we investigated the cytotoxic effect of the EGFR-targeting agents cetuximab and erlotinib under normoxia versus hypoxia. Findings Three cetuximab-sensitive HNSCC cell lines (SC263, LICR-HN2 and LICR-HN5) were treated with either cetuximab or erlotinib. Cells were incubated under normal or reduced oxygen conditions (<0.1% O2) for 24 or 72 h immediately after drug addition. Cell survival was assessed with the sulforhodamine B assay. Cetuximab and erlotinib established a dose-dependent growth inhibition under both normal and prolonged reduced oxygen conditions in all three HNSCC cell lines. However, a significantly increased sensitivity to cetuximab was observed in SC263 cells exposed to hypoxia for 72 h (p = 0.05), with IC50 values of 2.38 ± 0.59 nM, 0.64 ± 0.38 nM, and 0.10 ± 0.05 nM under normoxia, hypoxia for 24 h and hypoxia for 72 h, respectively. LICR-HN5 cells showed an increased sensitivity towards erlotinib when cells were incubated under hypoxia for 24 h (p = 0.05). Conclusions Our results suggest that both EGFR-inhibitors cetuximab and erlotinib maintain their growth inhibitory effect under hypoxia. These results suggest that resistance to anti-EGFR therapy in HNSCC is probably not the result of hypoxic regions within the tumor and other mechanisms are involved.
Collapse
Affiliation(s)
- Carolien Boeckx
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Jolien Van den Bossche
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Ines De Pauw
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Marc Peeters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium. .,Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.
| | - Filip Lardon
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Marc Baay
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - An Wouters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| |
Collapse
|
15
|
Welsh L, Panek R, McQuaid D, Dunlop A, Schmidt M, Riddell A, Koh DM, Doran S, Murray I, Du Y, Chua S, Hansen V, Wong KH, Dean J, Gulliford S, Bhide S, Leach MO, Nutting C, Harrington K, Newbold K. Prospective, longitudinal, multi-modal functional imaging for radical chemo-IMRT treatment of locally advanced head and neck cancer: the INSIGHT study. Radiat Oncol 2015; 10:112. [PMID: 25971451 PMCID: PMC4438605 DOI: 10.1186/s13014-015-0415-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/30/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Radical chemo-radiotherapy (CRT) is an effective organ-sparing treatment option for patients with locally advanced head and neck cancer (LAHNC). Despite advances in treatment for LAHNC, a significant minority of these patients continue to fail to achieve complete response with standard CRT. By constructing a multi-modality functional imaging (FI) predictive biomarker for CRT outcome for patients with LAHNC we hope to be able to reliably identify those patients at high risk of failing standard CRT. Such a biomarker would in future enable CRT to be tailored to the specific biological characteristics of each patients' tumour, potentially leading to improved treatment outcomes. METHODS/DESIGN The INSIGHT study is a single-centre, prospective, longitudinal multi-modality imaging study using functional MRI and FDG-PET/CT for patients with LAHNC squamous cell carcinomas receiving radical CRT. Two cohorts of patients are being recruited: one treated with, and another treated without, induction chemotherapy. All patients receive radical intensity modulated radiotherapy with concurrent chemotherapy. Patients undergo functional imaging before, during and 3 months after completion of radiotherapy, as well as at the time of relapse, should that occur within the first two years after treatment. Serum samples are collected from patients at the same time points as the FI scans for analysis of a panel of serum markers of tumour hypoxia. DISCUSSION The primary aim of the INSIGHT study is to acquire a prospective multi-parametric longitudinal data set comprising functional MRI, FDG PET/CT, and serum biomarker data from patients with LAHNC undergoing primary radical CRT. This data set will be used to construct a predictive imaging biomarker for outcome after CRT for LAHNC. This predictive imaging biomarker will be used in future studies of functional imaging based treatment stratification for patients with LAHNC. Additional objectives are: defining the reproducibility of FI parameters; determining robust methods for defining FI based biological target volumes for IMRT planning; creation of a searchable database of functional imaging data for data mining. The INSIGHT study will help to establish the role of FI in the clinical management of LAHNC. TRIAL REGISTRATION NCRI H&N CSG ID 13860.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Chemoradiotherapy/mortality
- Female
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/therapy
- Humans
- Longitudinal Studies
- Magnetic Resonance Imaging/methods
- Male
- Middle Aged
- Multimodal Imaging/methods
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm Staging
- Positron-Emission Tomography/methods
- Prognosis
- Prospective Studies
- Radiotherapy Planning, Computer-Assisted/methods
- Radiotherapy, Intensity-Modulated/methods
- Tomography, X-Ray Computed/methods
- Young Adult
Collapse
Affiliation(s)
- Liam Welsh
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
- Clinical Research Fellow, Head and Neck Unit, Royal Marsden Hospital, Sutton, Surrey, SM2 5PT, UK.
| | - Rafal Panek
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Dualta McQuaid
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Alex Dunlop
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Maria Schmidt
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Angela Riddell
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Dow-Mu Koh
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Simon Doran
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Iain Murray
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Yong Du
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Sue Chua
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Vibeke Hansen
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Kee H Wong
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Jamie Dean
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Sarah Gulliford
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Shreerang Bhide
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Martin O Leach
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Christopher Nutting
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Kevin Harrington
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Kate Newbold
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| |
Collapse
|
16
|
Zeitlin R, Nguyen HP, Rafferty D, Tyring S. Advancements in the Management of HPV-Associated Head and Neck Squamous Cell Carcinoma. J Clin Med 2015; 4:822-31. [PMID: 26239449 PMCID: PMC4470200 DOI: 10.3390/jcm4050822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 02/04/2023] Open
Abstract
Head and neck carcinomas have long been linked to alcohol and tobacco abuse; however, within the last two decades, the human papillomavirus (HPV) has emerged as a third etiology and is specifically associated with head and neck squamous cell carcinomas (HNSCC). In this anatomical region, the oncogenic HPV-16 mediates transformation and immortalization of epithelium, most commonly in the oropharynx. Nevertheless, the recent identification of novel HPV mechanisms thought to be specific to oropharyngeal carcinogenesis has coincided with observations that HPV-associated HNSCC has differing clinical behavior-in terms of natural history, therapeutic response, and prognosis-than HPV-negative head and neck tumors. Taken together with the growing incidence of HPV transmission in younger populations, these discoveries have sparked a rapid expansion in both laboratory and clinical studies on the infection and disease. Herein, we review the clinical characteristics of HPV-associated HNSCC, with particular emphasis on recent advancements in our understanding of the management of this infectious malignancy.
Collapse
Affiliation(s)
- Ross Zeitlin
- University of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32603, USA.
| | | | - David Rafferty
- Paul L. Foster School of Medicine, 5001 El Paso Dr., El Paso, TX 79905, USA.
| | - Stephen Tyring
- Department of Dermatology, University of Texas Medical School at Houston, 6655 Travis Street, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Ramchandani D, Weber GF. Interactions between osteopontin and vascular endothelial growth factor: Implications for cancer. Biochim Biophys Acta Rev Cancer 2015; 1855:202-22. [PMID: 25732057 DOI: 10.1016/j.bbcan.2015.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 02/10/2015] [Accepted: 02/22/2015] [Indexed: 12/12/2022]
Abstract
For this comprehensive review, 257 publications with the keywords "osteopontin" or "OPN" and "vascular endothelial growth factor" or "VEGF" in PubMed were screened (time frame from year 1996 to year 2014). 37 articles were excluded because they were not focused on the interactions between these molecules, and papers relevant for transformation-related phenomena were selected. Osteopontin (OPN) and vascular endothelial growth factor (VEGF) are characterized by a convergence in function for regulating cell motility and angiogenesis, the response to hypoxia, and apoptosis. Often, they are co-expressed or one molecule induces the other, however, in some settings OPN-associated pathways and VEGF-associated pathways are distinct. Their relationships affect the pathogenesis in cancer, where they contribute to progression and angiogenesis and serve as markers for poor prognosis. The inhibition of OPN may reduce VEGF levels and suppress tumor progression. In vascular pathologies, these two cytokines mediate remodeling, but may also perpetuate inflammation and narrowing of the arteries. OPN and VEGF are elevated and contribute to vascularization in inflammatory diseases.
Collapse
Affiliation(s)
| | - Georg F Weber
- James L. Winkle College of Pharmacy, University of Cincinnati, USA.
| |
Collapse
|
18
|
Vassilakopoulou M, Psyrri A, Argiris A. Targeting angiogenesis in head and neck cancer. Oral Oncol 2015; 51:409-15. [PMID: 25680863 DOI: 10.1016/j.oraloncology.2015.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/06/2015] [Accepted: 01/08/2015] [Indexed: 10/24/2022]
Abstract
Angiogenesis is a crucial step in tumor growth and metastasis. Head and neck squamous cell carcinomas (HNSCC) highly express angiogenesis factors, such as vascular endothelial growth factor (VEGF), which are associated with patient prognosis. Antiangiogenesis agents can potentially modulate tumor microenvironment and induce radiosensitivity and chemosensitivity. In this review, we discuss the molecular mechanisms underlying angiogenesis involved in HNSCC, preclinical data with antiangiogenesis agents as well as potential predictive biomarkers. We also review novel therapies under investigation and summarize the results of clinical trials using antiangiogenesis agents alone or in combination with conventional therapies in HNSCC.
Collapse
Affiliation(s)
| | - Amanda Psyrri
- Attikon Hospital and Medical School of Athens, Athens, Greece
| | - Athanassios Argiris
- Hygeia Hospital, Athens, Greece; University of Texas Health Science Center at San Antonio, TX, USA.
| |
Collapse
|
19
|
Gilbert J, Schell MJ, Zhao X, Murphy B, Tanvetyanon T, Leon ME, Neil Hayes D, Haigentz M, Saba N, Nieva J, Bishop J, Sidransky D, Ravi R, Bedi A, Chung CH. A randomized phase II efficacy and correlative studies of cetuximab with or without sorafenib in recurrent and/or metastatic head and neck squamous cell carcinoma. Oral Oncol 2015; 51:376-82. [PMID: 25593015 DOI: 10.1016/j.oraloncology.2014.12.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 12/15/2014] [Indexed: 01/15/2023]
Abstract
INTRODUCTION A combination of cetuximab and sorafenib in patients with recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) were assessed for potential benefit. MATERIAL AND METHODS In a randomized phase II study, R/M HNSCC patients were treated with cetuximab 400mg/m(2) IV on day 1 followed by 250mg/m(2) IV weekly (Arm A), or cetuximab at the same dose/schedule plus sorafenib 400mg PO twice-a-day (Arm B). Each cycle was 21days. Tumor p16 and HPV status, and plasma immunomodulatory cytokine levels were assessed. RESULTS Of 55 patients enrolled (Arm A-27, Arm B-28), 52 patients received assigned treatments and 43 were evaluable for response. Overall response rate was 8% for both arms. Median overall survival (OS) and progression-free survival (PFS) were 9.0 and 3.0months in Arm A, and 5.7 and 3.2months in Arm B, respectively. Forty-four patients had tumors available for p16 staining (35-negative, 9-positive). Three of nine p16-positive tumors were also HPV positive. The p16-negative patients had significantly better PFS compared to the p16-positive patients (3.7 vs. 1.6months; p-value: 0.03), regardless of study arms. Twenty-four plasma samples were tested for 12 cytokine levels and patients with higher TGFβ1 levels had inferior PFS compared to lower levels (1.9 vs. 4.7months; adjusted p-value: 0.015), regardless of study arms. CONCLUSIONS A subset of R/M patients with p16-negative tumors or lower plasma TGFβ1 levels had longer PFS given the cetuximab-based therapy. However, both arms showed only modest response and sorafenib given with cetuximab did not demonstrate clinical benefit.
Collapse
Affiliation(s)
- Jill Gilbert
- Vanderbilt University, Nashville, TN, United States.
| | | | - Xiuhua Zhao
- H. Lee Moffitt Cancer Center, Tampa, FL, United States.
| | | | | | - Marino E Leon
- H. Lee Moffitt Cancer Center, Tampa, FL, United States.
| | - D Neil Hayes
- UNC Lineberger Cancer Center, Chapel Hill, NC, United States.
| | - Missak Haigentz
- Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States.
| | - Nabil Saba
- Emory Winship Cancer Institute, Atlanta, GA, United States.
| | - Jorge Nieva
- University of Southern California, Los Angeles, CA, United States.
| | - Justin Bishop
- Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - David Sidransky
- Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Rajani Ravi
- Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Atul Bedi
- Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | | |
Collapse
|
20
|
Gold KA, Kim ES, Liu DD, Yuan P, Behrens C, Solis LM, Kadara H, Rice DC, Wistuba II, Swisher SG, Hofstetter WL, Lee JJ, Hong WK. Prediction of survival in resected non-small cell lung cancer using a protein expression-based risk model: implications for personalized chemoprevention and therapy. Clin Cancer Res 2013; 20:1946-54. [PMID: 24366692 DOI: 10.1158/1078-0432.ccr-13-1959] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with resected non-small cell lung cancer (NSCLC) are at risk for recurrence of disease, but we do not have tools to predict which patients are at highest risk. We set out to create a risk model incorporating both clinical data and biomarkers. EXPERIMENTAL DESIGN We assembled a comprehensive database with archival tissues and clinical follow-up from patients with NSCLC resected between 2002 and 2005. Twenty-one proteins identified from our preclinical studies as related to lung carcinogenesis were investigated, including pathways related to metabolism, DNA repair, inflammation, and growth factors. Expression of proteins was quantified using immunohistochemistry. Immunohistochemistry was chosen because it is widely available and can be performed on formalin-fixed paraffin-embedded specimens. Cox models were fitted to estimate effects of clinical factors and biomarkers on recurrence-free survival (RFS) and overall survival (OS). RESULTS A total of 370 patients are included in our analysis. With median follow-up of 5.3 years, median OS is 6.4 years. A total of 209 cases with recurrence or death were observed. Multicovariate risk models for RFS and OS were developed including relevant biomarkers, age, and stage. Increased expression of phospho-adenosine monophosphate-activated protein kinase (pAMPK), phospho-mTOR (pmTOR), epithelial cell adhesion molecule (EpCAM), and calcium/calmodulin-dependent serine protein kinase were significant (P < 0.05) predictors for favorable RFS; insulin receptor, chemokine (C-X-C motif) receptor 2 (CXCR2), and insulin-like growth factor-1 receptor predicted for unfavorable RFS. Significant (P < 0.05) predictors for favorable OS include pAMPK, pmTOR, and EpCAM; CXCR2 and flap structure-specific endonuclease-1 predicted unfavorable OS. CONCLUSION We have developed a comprehensive risk model predictive for recurrence in our large retrospective database, which is one of the largest reported series of resected NSCLC.
Collapse
Affiliation(s)
- Kathryn A Gold
- Authors' Affiliations: Departments of Thoracic/Head and Neck Medical Oncology, Biostatistics, Pathology, and Thoracic and Cardiovascular Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; Levine Cancer Center; Philips Digital Pathology; and Department of Pathology, University of San Francisco Xavier de Chuquisaca Sucre-Bolivia, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ostheimer C, Bache M, Güttler A, Kotzsch M, Vordermark D. A pilot study on potential plasma hypoxia markers in the radiotherapy of non-small cell lung cancer. Osteopontin, carbonic anhydrase IX and vascular endothelial growth factor. Strahlenther Onkol 2013; 190:276-82. [PMID: 24322994 DOI: 10.1007/s00066-013-0484-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/16/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hypoxic radioresistance plays a critical role in the radiotherapy of cancer and adversely impacts prognosis and treatment response. This prospective study investigated the interrelationship and the prognostic significance of several hypoxia-related proteins in non-small cell lung cancer (NSCLC) patients treated by radiotherapy ± chemotherapy. MATERIAL AND METHODS Pretreatment osteopontin (OPN), vascular endothelial growth factor (VEGF) and carbonic anhydrase IX (CA IX) plasma levels were determined by ELISA in 55 NSCLC (M0) patients receiving 66 Gy curative-intent radiotherapy or chemoradiation. Marker correlation, association with clinicopathological parameters and the prognostic value of a biomarker combination was evaluated. RESULTS All biomarkers were linearly correlated and linked to different clinical parameters including lung function, weight loss (OPN), gross tumor volume (VEGF) and T stage (CA IX). High OPN (p = 0.03), VEGF (p = 0.02) and CA IX (p = 0.04) values were significantly associated with poor survival. Double marker combination additively increased the risk of death by a factor of 2 and high plasma levels of the triple combination OPN/VEGF/CA IX yielded a 5.9-fold risk of death (p = 0.009). The combined assessment of OPN/VEGF/CA IX correlated independently with prognosis (p = 0.03) in a multivariate Cox regression model including N stage, T stage and GTV. CONCLUSION This pilot study suggests that a co-detection augments the prognostic value of single markers and that the integration of OPN, VEGF and CA IX into a hypoxic biomarker profile for the identification of patients with largely hypoxic and radioresistant tumors should be further evaluated.
Collapse
Affiliation(s)
- C Ostheimer
- Department of Radiation Oncology, Martin-Luther-University Halle-Wittenberg, Dryanderstr. 4, 06110, Halle (Saale), Germany,
| | | | | | | | | |
Collapse
|
22
|
Al-Taei S, Banner R, Powell N, Evans M, Palaniappan N, Tabi Z, Man S. Decreased HPV-specific T cell responses and accumulation of immunosuppressive influences in oropharyngeal cancer patients following radical therapy. Cancer Immunol Immunother 2013; 62:1821-30. [PMID: 24146146 PMCID: PMC11028645 DOI: 10.1007/s00262-013-1488-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 10/08/2013] [Indexed: 12/19/2022]
Abstract
Oropharyngeal cancer (OPC) is a type of squamous cell head and neck cancer that is often associated with human papillomavirus (HPV) infection, suggesting the potential for immunotherapeutic targeting of HPV antigens. This study aimed to determine the effect of radical therapy on HPV-specific T cells and other immune parameters in 20 OPC patients, as a prelude to future immunotherapy studies. HPV DNA could be detected in 9/12 available tissue samples (8/9 HPV(+) samples were also p16(+)). HPV-specific T cell responses against HPV16 E6 and E7 peptides were detected by enzyme-linked immunoSPOT in 10/13 and 8/13 evaluable patients, respectively, but did not appear to correlate with HPV status. Post-treatment, both HPV E6 and E7 T cell responses were decreased (4/13 and 2/13 patients, respectively). These reductions in T cell response could not be explained by a concurrent decrease in memory T cells whose absolute numbers were relatively unaffected by radical therapy (27,975 vs. 25,661/10(5) PBMC) despite a significant decrease in overall lymphocyte counts (1.74 vs. 0.69 × 10(9)/L). Instead, there were significant increases in regulatory T cells (3.7 vs. 6.8 %) and a population of myeloid-derived suppressor cells (CD14(-)HLA-DR(-)CD15(hi), 12.38 vs. 21.92 %). This suggests that immunosuppression may contribute to the reduction in HPV-specific T cell responses post-treatment, although study of larger patient cohorts will be required to test whether this affects clinical outcome. Overall these findings suggest that HPV-targeted immunotherapy in post-therapy OPC patients will require multiple strategies to boost T cell immunity and to overcome the influence of immunosuppressive cells.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/therapy
- Carcinoma, Squamous Cell/virology
- Cell Proliferation
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Human papillomavirus 16/genetics
- Human papillomavirus 16/immunology
- Humans
- Immunoenzyme Techniques
- Immunologic Memory
- Immunotherapy
- Interferon-gamma/metabolism
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/virology
- Male
- Middle Aged
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Myeloid Cells/virology
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Oropharyngeal Neoplasms/immunology
- Oropharyngeal Neoplasms/therapy
- Oropharyngeal Neoplasms/virology
- Papillomavirus E7 Proteins/genetics
- Papillomavirus E7 Proteins/immunology
- Papillomavirus Infections/immunology
- Papillomavirus Infections/therapy
- Papillomavirus Infections/virology
- Real-Time Polymerase Chain Reaction
- Repressor Proteins/genetics
- Repressor Proteins/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/virology
Collapse
Affiliation(s)
| | | | - Ned Powell
- Institute of Cancer and Genetics, Cardiff University School of Medicine, Cancer and Genetics Building, Heath Park, Cardiff, CF14 4XN UK
| | | | | | | | - Stephen Man
- Institute of Cancer and Genetics, Cardiff University School of Medicine, Cancer and Genetics Building, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|
23
|
Boeckx C, Baay M, Wouters A, Specenier P, Vermorken JB, Peeters M, Lardon F. Anti-epidermal growth factor receptor therapy in head and neck squamous cell carcinoma: focus on potential molecular mechanisms of drug resistance. Oncologist 2013; 18:850-64. [PMID: 23821327 DOI: 10.1634/theoncologist.2013-0013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeted therapy against the epidermal growth factor receptor (EGFR) is one of the most promising molecular therapeutics for head and neck squamous cell carcinoma (HNSCC). EGFR is overexpressed in a wide range of malignancies, including HNSCC, and initiates important signal transduction pathways in HNSCC carcinogenesis. However, primary and acquired resistance are serious problems and are responsible for low single-agent response rate and tumor recurrence. Therefore, an improved understanding of the molecular mechanisms of resistance to EGFR inhibitors may provide valuable indications to identify biomarkers that can be used clinically to predict response to EGFR blockade and to establish new treatment options to overcome resistance. To date, no predictive biomarker for HNSCC is available in the clinic. Therapeutic resistance to anti-EGFR therapy may arise from mechanisms that can compensate for reduced EGFR signaling and/or mechanisms that can modulate EGFR-dependent signaling. In this review, we will summarize some of these molecular mechanisms and describe strategies to overcome that resistance.
Collapse
Affiliation(s)
- Carolien Boeckx
- Center for Oncological Research Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of Antwerp, Wilrijk, Belgium
| | | | | | | | | | | | | |
Collapse
|
24
|
Agarwal M, He C, Siddiqui J, Wei J, Macoska JA. CCL11 (eotaxin-1): a new diagnostic serum marker for prostate cancer. Prostate 2013; 73:573-81. [PMID: 23059958 PMCID: PMC3594486 DOI: 10.1002/pros.22597] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 09/10/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND The recent recommendation of the U.S. Preventive Services Task Force against PSA-based screening for prostate cancer was based, in part, on the lack of demonstrated diagnostic utility of serum PSA values in the low, but detectable range to successfully predict prostate cancer. Though controversial, this recommendation reinforced the critical need to develop, validate, and determine the utility of other serum and/or urine transcript and protein markers as diagnostic markers for PCa. The studies described here were intended to determine whether inflammatory cytokines might augment serum PSA as a diagnostic marker for prostate cancer. METHODS Multiplex ELISA assays were performed to quantify CCL1, CCL2, CCL5, CCL8, CCL11, CCL17, CXCL1, CXCL5, CXCL8, CXCL10, CXCL12, and IL-6 protein levels in the serum of 272 men demonstrating serum PSA values of <10 ng/ml and undergoing a 12 core diagnostic needle biopsy for detection of prostate cancer. Logistic regression was used to identify the associations between specific chemokines and prostate cancer status adjusted for prostate volume, and baseline PSA. RESULTS Serum levels for CCL1 (I-309) were significantly elevated among all men with enlarged prostates (P < 0.04). Serum levels for CCL11 (Eotaxin-1) were significantly elevated among men with prostate cancer regardless of prostate size (P < 0.01). The remaining 10 cytokines examined in this study did not exhibit significant correlations with either prostate volume or cancer status. CONCLUSIONS Serum CCL11 values may provide a useful diagnostic tool to help distinguish between prostatic enlargement and prostate cancer among men demonstrating low, but detectable, serum PSA values.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Chang He
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Javed Siddiqui
- Center for Translational Pathology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - John Wei
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
- Center for Translational Pathology, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Jill A. Macoska
- Department of Urology, The University of Michigan School of Medicine, Ann Arbor, MI
- Address Correspondence To: Jill A. Macoska, Ph.D., Department of Urology, The University of Michigan, 6217 Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0944, (734) 647-8121 TEL, (734) 647-9271 FAX,
| |
Collapse
|
25
|
Bauman JE, Arias-Pulido H, Lee SJ, Fekrazad MH, Ozawa H, Fertig E, Howard J, Bishop J, Wang H, Olson GT, Spafford MJ, Jones DV, Chung CH. A phase II study of temsirolimus and erlotinib in patients with recurrent and/or metastatic, platinum-refractory head and neck squamous cell carcinoma. Oral Oncol 2013; 49:461-7. [PMID: 23384718 PMCID: PMC3805493 DOI: 10.1016/j.oraloncology.2012.12.016] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The epidermal growth factor receptor (EGFR) is a validated target in head and neck squamous cell carcinoma (HNSCC). In recurrent and/or metastatic (R/M) HNSCC, resistance to anti-EGFR therapy inevitably occurs. Downstream activation of the PI3K/Akt/mTOR pathway is an established resistance mechanism. Concurrent mTOR blockade may improve efficacy of anti-EGFR therapy. MATERIALS AND METHODS Erlotinib 150 mg daily and temsirolimus 15 mg weekly were administered to patients with platinum-refractory R/M HNSCC and ECOG performance status 0-2. The primary endpoint was progression-free survival (PFS). Correlative studies determined PIK3CA and HRAS mutation status; p16, EGFR, pS6K, pAkt and PTEN expression; and pre- and post-treatment plasma levels of 20 immunomodulatory cytokines. RESULTS Twelve patients enrolled; six withdrew within 6 weeks due to toxicity or death, prompting early closure of the trial. Grade ≥ 3 toxicities included fatigue, diarrhea, gastrostomy tube infection, peritonitis, pneumonia, dyspnea, and HN edema. Median PFS was 1.9 months. Median overall survival was 4.0 months. Six/12 tumors were p16(+), 9/11 lacked measurable PTEN expression, and 1/12 harbored a PIK3CA mutation. On exploratory analysis, high baseline plasma VEGF and interferon-gamma levels marginally associated with tumor progression. CONCLUSIONS The combination of erlotinib and temsirolimus was poorly tolerated. Low prevalence of PTEN expression and 8% incidence of PIK3CA mutations indicate biological relevance of this pathway in R/M disease. Investigation of more tolerable combinations of EGFR and PI3K/Akt/mTOR pathway inhibitors in selected HNSCC patients is warranted.
Collapse
Affiliation(s)
- Julie E Bauman
- Department of Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
A novel serum protein signature associated with resistance to epidermal growth factor receptor tyrosine kinase inhibitors in head and neck squamous cell carcinoma. Eur J Cancer 2013; 49:2512-21. [PMID: 23582742 DOI: 10.1016/j.ejca.2013.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/05/2013] [Accepted: 03/05/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND Acquired resistance to tyrosine kinase inhibitors (TKIs) is becoming a major challenge in the treatment of many cancers. Epidermal growth factor receptor (EGFR) is overexpressed in squamous carcinomas, notably those of the head and neck (HNSCC), and can be targeted with several TKIs. We aimed to identify soluble proteins suitable for development as markers of EGFR TKI resistance in cancer patients to aid in early and minimally invasive assessment of therapeutic responses. METHODS Resistant HNSCC cell lines were generated by exposure to an EGFR TKI, gefitinib, in vitro. Cell lines were characterised for their biological behaviour in vitro (using growth inhibition assays, flow cytometry, western blots, antibody arrays and/or immunoassays) and in vivo (using subcutaneous tumour xenografts). Sera from EGFR-treated and -untreated HNSCC patients were analysed by immunoassay. RESULTS Two independent sublines of CAL 27 and a PJ34 subline with acquired resistance to EGFR TKIs (gefitinib, erlotinib and afatinib) were developed. Resistant cells grew as highly aggressive xenografts leading to reduced host survival rates compared with EGFR-TKI sensitive cells. This suggested a link between resistance in vitro and poor prognosis in vivo. A significant upregulation of proteins linked to tumour angiogenesis and invasion was identified in resistant cells. This 'resistance-associated protein signature' (RAPS) was detected in the sera of a small cohort of HNSCC patients and was associated with reduced survival. CONCLUSION We have identified a protein signature associated with EGFR-TKI resistance that may also be linked to poor prognosis and warrants further investigation as a potential clinical biomarker.
Collapse
|
27
|
Bedi A, Chang X, Noonan K, Pham V, Bedi R, Fertig EJ, Considine M, Califano JA, Borrello I, Chung CH, Sidransky D, Ravi R. Inhibition of TGF-β enhances the in vivo antitumor efficacy of EGF receptor-targeted therapy. Mol Cancer Ther 2012; 11:2429-39. [PMID: 22927667 DOI: 10.1158/1535-7163.mct-12-0101-t] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
EGF receptor (EGFR)-targeted monoclonal antibodies (mAb), such as cetuximab, execute their antitumor effect in vivo via blockade of receptor-ligand interactions and engagement of Fcγ receptors on immune effector cells that trigger antibody-dependent cell-mediated cytotoxicity (ADCC). We show that tumors counteract the in vivo antitumor activity of anti-EGFR mAbs by increasing tumor cell-autonomous expression of TGF-β. We show that TGF-β suppresses the expression of key molecular effectors of immune cell-mediated cytotoxicity, including Apo2L/TRAIL, CD95L/FasL, granzyme B, and IFN-γ. In addition to exerting an extrinsic inhibition of the cytotoxic function of immune effectors, TGF-β-mediated activation of AKT provides an intrinsic EGFR-independent survival signal that protects tumor cells from immune cell-mediated apoptosis. Treatment of mice-bearing xenografts of human head and neck squamous cell carcinoma with cetuximab resulted in emergence of resistant tumor cells that expressed relatively higher levels of TGF-β compared with untreated tumor-bearing mice. Although treatment with cetuximab alone forced the natural selection of TGF-β-overexpressing tumor cells in nonregressing tumors, combinatorial treatment with cetuximab and a TGF-β-blocking antibody prevented the emergence of such resistant tumor cells and induced complete tumor regression. Therefore, elevated levels of TGF-β in the tumor microenvironment enable tumor cells to evade ADCC and resist the antitumor activity of cetuximab in vivo. Our results show that TGF-β is a key molecular determinant of the de novo and acquired resistance of cancers to EGFR-targeted mAbs, and provide a rationale for combinatorial targeting of TGF-β to improve anti-EGFR-specific antibody therapy of EGFR-expressing cancers.
Collapse
Affiliation(s)
- Atul Bedi
- Head and Neck Cancer Research Division, Department of Otolaryngology-Head and Neck Surgery, 1550 Orleans Street, Johns Hopkins University School of Medicine, Cancer Research Building II, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ralhan R. How will increasing our knowledge of the head and neck cancer cells' secretome benefit head and neck squamous cell carcinoma patients? Expert Rev Proteomics 2012; 9:115-8. [PMID: 22462781 DOI: 10.1586/epr.12.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
Abstract
PURPOSE OF REVIEW Despite advances in multimodality therapy, the overall 5-year survival rate is 40-50% in patients with head and neck squamous cell carcinoma (HNSCC) and current multimodality approaches impart significant toxicities. This review highlights promising targets with the potential to improve clinical outcomes in HNSCC. RECENT FINDINGS In addition to mutagenic exposure to tobacco and alcohol as risk factors, recent studies have shown that human papillomavirus is one of the main causes of HNSCC and as such is being investigated as a therapeutic target. Furthermore, recent data generated from whole exome sequencing of HNSCC, new insights into the biology of DNA damage repair, and increased understanding of tumor hypoxia responses are pointing to new therapeutic possibilities for treating HNSCC. SUMMARY HNSCC is a heterogeneous disease. Improved treatment will require a rapid translation of basic science research, and the simultaneous development of novel therapeutics and corresponding biomarkers to guide their application.
Collapse
|
30
|
Lucs A, Saltman B, Chung CH, Steinberg BM, Schwartz DL. Opportunities and challenges facing biomarker development for personalized head and neck cancer treatment. Head Neck 2012; 35:294-306. [PMID: 22287320 DOI: 10.1002/hed.21975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/08/2011] [Indexed: 12/25/2022] Open
Abstract
Head and neck oncologists have traditionally relied on clinical tumor features and patient characteristics to guide care of individual patients. As surgical, radiotherapeutic, and systemic treatments have evolved to become more anatomically precise and mechanistically specific, the opportunity for improved cure and functional patient recovery has never been more promising for this historically debilitating cancer. However, personalized treatment must be accompanied by sophisticated patient selection to triage the application of advanced therapies toward ideal patient candidates. In this monograph, we review current progress, investigative themes, and key challenges facing head and neck cancer biomarker development intended to make personalized head and neck cancer treatment a clinical reality.
Collapse
Affiliation(s)
- Alexandra Lucs
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Benjamin Saltman
- Department of Otolaryngology, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY, USA
| | - Christine H Chung
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bettie M Steinberg
- The Feinstein Institute for Medical Research, Manhasset, NY, USA.,Department of Otolaryngology, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY, USA
| | - David L Schwartz
- The Feinstein Institute for Medical Research, Manhasset, NY, USA.,Department of Otolaryngology, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY, USA.,Department of Radiation Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY, USA
| |
Collapse
|
31
|
Langer CJ. Exploring biomarkers in head and neck cancer. Cancer 2012; 118:3882-92. [PMID: 22281752 DOI: 10.1002/cncr.26718] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 09/09/2011] [Accepted: 10/28/2011] [Indexed: 02/04/2023]
Abstract
Personalized medicine based on predictive markers linked to drug response, it is hoped, will lead to improvements in outcomes and avoidance of unnecessary treatment in squamous cell carcinoma of the head and neck (SCCHN). Recent research has shown that expression of ERCC1 may predict resistance to treatment with platinum agents. Future testing for this marker may help select the optimal type of chemotherapy. Infection with human papillomavirus (HPV) is associated with less aggressive disease and better prognosis in locally advanced SCCHN treated with chemoradiation or radiation alone; HPV-positive patients may ultimately benefit from less intensive, less toxic therapy. K-RAS mutations, occurring in about 40% of colorectal cancers and associated with lack of benefit from epidermal growth factor receptor (EGFR) antibodies in this disease, are found in <5% of SCCHN patients, making routine testing for K-RAS mutations unwarranted at this time. Virtually all head and neck tumors overexpress EGFR, which limits the usefulness of EGFR expression as a marker for treatment selection. Although the incidence of EGFR tyrosine kinase domain mutations is very rare, a better understanding of the role of EGFR mutations, expression, amplification, and downstream effects in SCCHN may help define the role of EGFR in this setting. These observations caution against extrapolating results obtained with biomarkers in other types of cancer to SCCHN. Validation of each biomarker in the context of SCCHN clinical trials will be required before a specific marker can be incorporated into daily practice.
Collapse
Affiliation(s)
- Corey J Langer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
32
|
Abstract
Head and neck squamous cell cancer is the sixth most common cancer in the world. Despite advances in combined modality therapy, poor outcomes continue to be observed in the form of locoregional recurrence, metastasis, and development of second primary tumors. As tumors vary in their molecular and genetic etiology and because often there is already deregulation at the molecular level in otherwise histopathologically normal tissue, risk stratification using clinical and pathologic criteria alone has proved to be inadequate. In this article, the reader will gain an appreciation for the current advances in biomarker discovery using advanced technology and data interpretation in microarray analysis and proteomics. In addition, other molecular targets, aside from epidermal growth factor receptor, are discussed in the context of their promising role in predicting recurrence, response to therapy, survival, and overall prognosis.
Collapse
|
33
|
Argiris A, Lee SC, Feinstein T, Thomas S, Branstetter BF, Seethala R, Wang L, Gooding W, Grandis JR, Ferris RL. Serum biomarkers as potential predictors of antitumor activity of cetuximab-containing therapy for locally advanced head and neck cancer. Oral Oncol 2011; 47:961-6. [PMID: 21889392 DOI: 10.1016/j.oraloncology.2011.07.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/25/2011] [Accepted: 07/29/2011] [Indexed: 02/02/2023]
Abstract
We sought to identify biomarkers of antitumor activity in patients with locally advanced head and neck cancer treated with therapy containing cetuximab, an epidermal growth factor receptor (EGFR) inhibitor. Patients with stage III-IVB head and neck cancer received cisplatin, docetaxel, and cetuximab (TPE) followed by radiotherapy, cisplatin, and cetuximab (XPE) and maintenance cetuximab in a phase II clinical trial. Serum and tissue biomarkers were examined for treatment-related changes and for association with clinical outcomes. Concentrations of 31 cytokines, chemokines and growth factors were measured before and after 3 cycles (9 weeks) of induction TPE using multi-analyte immunobead-based profiling (Luminex Corp., Austin, TX), with selected analytes validated by a single analyte enzyme-linked immunosorbent assay. Tumor biomarkers included phosphorylated signal transducer and activator of transcription-3 (pSTAT3), EGFR and human papillomavirus (HPV). Thirty-one patients had baseline biomarkers and 25 had paired samples, pre- and post-TPE. Adjusting for false discovery, 14 analytes including MCP1c, IP-10, Leptin, interleukin (IL)-5, Eotaxin, IL-6, G-CSF, CXCL5 changed significantly post TPE induction. Serum vascular endothelial growth factor (VEGF) and IL-6 levels were associated with tumor response as assessed by positron emission tomography and progression-free survival, however, the association was not significant after adjustment for false discovery. Analytes were not associated with toxicities, smoking history, HPV status, EGFR amplification, or pSTAT3 tumor protein levels. Baseline serum biomarkers, in particular VEGF and IL-6, were identified as potentially useful prognostic markers of cetuximab-containing therapy. Validation is warranted in future studies specifically designed to detect biomarker associations.
Collapse
Affiliation(s)
- Athanassios Argiris
- Head and Neck Cancer Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Saddoughi SA, Garrett-Mayer E, Chaudhary U, O'Brien PE, Afrin LB, Day TA, Gillespie MB, Sharma AK, Wilhoit CS, Bostick R, Senkal CE, Hannun YA, Bielawski J, Simon GR, Shirai K, Ogretmen B. Results of a phase II trial of gemcitabine plus doxorubicin in patients with recurrent head and neck cancers: serum C₁₈-ceramide as a novel biomarker for monitoring response. Clin Cancer Res 2011; 17:6097-105. [PMID: 21791630 DOI: 10.1158/1078-0432.ccr-11-0930] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Here we report a phase II clinical trial, which was designed to test a novel hypothesis that treatment with gemcitabine (GEM)/doxorubicin (DOX) would be efficacious via reconstitution of C(18)-ceramide signaling in head and neck squamous cell carcinoma (HNSCC) patients for whom first-line platinum-based therapy failed. EXPERIMENTAL DESIGN Patients received GEM (1,000 mg/m²) and DOX (25 mg/m²) on days 1 and 8, every 21 days, until disease progression. After completion of 2 treatment cycles, patients were assessed radiographically, and serum samples were taken for sphingolipid measurements. RESULTS We enrolled 18 patients in the trial, who were evaluable for toxicity, and 17 for response. The most common toxicity was neutropenia, observed in 9 of 18 patients, and there were no major nonhematologic toxicities. Of the 17 patients, 5 patients had progressive disease (PD), 1 had complete response (CR), 3 exhibited partial response (PR), and 8 had stable disease (SD). The median progression-free survival was 1.6 months (95% CI: 1.4-4.2) with a median survival of 5.6 months (95% CI: 3.8-18.2). Remarkably, serum sphingolipid analysis revealed significant differences in patterns of C₁₈-ceramide elevation in patients with CR/PR/SD in comparison with patients with PD, indicating the reconstitution of tumor suppressor ceramide generation by GEM/DOX treatment. CONCLUSIONS Our data suggest that the GEM/DOX combination could represent an effective treatment for some patients with recurrent or metastatic HNSCC, and that serum C₁₈-ceramide elevation might be a novel serum biomarker of chemotherapy response.
Collapse
Affiliation(s)
- Sahar A Saddoughi
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Argiris A, Duffy AG, Kummar S, Simone NL, Arai Y, Kim SW, Rudy SF, Kannabiran VR, Yang X, Jang M, Chen Z, Suksta N, Cooley-Zgela T, Ramanand SG, Ahsan A, Nyati MK, Wright JJ, Van Waes C. Early tumor progression associated with enhanced EGFR signaling with bortezomib, cetuximab, and radiotherapy for head and neck cancer. Clin Cancer Res 2011; 17:5755-64. [PMID: 21750205 DOI: 10.1158/1078-0432.ccr-11-0861] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A phase I clinical trial and molecular correlative studies were conducted to evaluate preclinical evidence for combinatorial activity of the proteasome inhibitor bortezomib, the epidermal growth factor receptor (EGFR) inhibitor cetuximab, and radiation therapy. EXPERIMENTAL DESIGN Patients with radiotherapy-naive stage IV or recurrent squamous cell carcinoma of the head and neck (SCCHN) were studied. Escalating doses of bortezomib (0.7, 1.0, and 1.3 mg/m²) were given intravenously twice weekly on days 1, 4, 8, and 11, every 21 days, with weekly cetuximab beginning 1 week prior and concurrently with intensity-modulated radiotherapy, delivered in 2 Gy fractions to 70 to 74 Gy. Molecular effects were examined in serial serum and SCCHN tumor specimens and the cell line UMSCC-1. RESULTS Seven patients were accrued before the study was terminated when five of six previously untreated patients with favorable prognosis oropharyngeal SCCHN progressed within 1 year (progression-free survival = 4.8 months; 95% CI, 2.6-6.9). Three patients each received bortezomib 0.7 or 1.0 mg/m², without dose-limiting toxicities; one patient treated at 1.3 mg/m² was taken off study due to recurring cetuximab infusion reaction and progressive disease (PD). Expected grade 3 toxicities included radiation mucositis (n = 4), dermatitis (n = 4), and rash (n = 1). SCCHN-related cytokines increased in serial serum specimens of patients developing PD (P = 0.029). Bortezomib antagonized cetuximab- and radiation-induced cytotoxicity, degradation of EGFR, and enhanced prosurvival signal pathway activation in SCCHN tumor biopsies and UMSCC-1. CONCLUSIONS Combining bortezomib with cetuximab and radiation therapy showed unexpected early progression, evidence for EGFR stabilization, increased prosurvival signaling, and SCCHN cytokine expression, warranting avoidance of this combination.
Collapse
Affiliation(s)
- Athanassios Argiris
- Hematology-Oncology and Head and Neck Cancer Program, and Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Brennan DJ, O'Connor DP, Rexhepaj E, Ponten F, Gallagher WM. Antibody-based proteomics: fast-tracking molecular diagnostics in oncology. Nat Rev Cancer 2010; 10:605-17. [PMID: 20720569 DOI: 10.1038/nrc2902] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The effective implementation of personalized cancer therapeutic regimens depends on the successful identification and translation of informative biomarkers to aid clinical decision making. Antibody-based proteomics occupies a pivotal space in the cancer biomarker discovery and validation pipeline, facilitating the high-throughput evaluation of candidate markers. Although the clinical utility of these emerging technologies remains to be established, the traditional use of antibodies as affinity reagents in clinical diagnostic and predictive assays suggests that the rapid translation of such approaches is an achievable goal. Furthermore, in combination with, or as alternatives to, genomic and transcriptomic methods for patient stratification, antibody-based proteomics approaches offer the promise of additional insight into cancer disease states. In this Review, we discuss the current status of antibody-based proteomics and its contribution to the development of new assays that are crucial for the realization of individualized cancer therapy.
Collapse
Affiliation(s)
- Donal J Brennan
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | |
Collapse
|