• 6/30/2008
  • web-based article
  • Bhuvanesh Singh and David G. Pfister
  • Journal of Clinical Oncology, Vol 26, No 19 (July 1), 2008: pp. 3114-3116

Integrated chemotherapy and radiation therapy have become the standard of care for most patients with advanced stage laryngopharyngeal cancers. The concurrent administration of these two modalities is the approach recommended by most experts at present.1 This strategy achieves higher rates of locoregional control compared to when chemotherapy is given as induction treatment before, or as an adjuvant after, radiation therapy.2-6 However, the benefits from concomitant chemoradiotherapy treatment are tempered by higher rates of treatment-related sequelae, especially in the short term. This issue is of particular concern in patients that fail to respond and who have to endure the adverse effects of treatment.

Based on observations that response to chemotherapy predicts radiation response, initial combined modality trials used response to induction chemotherapy to select patients for subsequent organ preservation treatment with definitive radiation, reserving laryngectomy for chemoresistant patients.5,6 However, with a shift to concomitant administration of chemotherapy and radiation therapy, the opportunity for treatment selection based on initial response was lost.3 To maximize the potential value of induction chemotherapy in terms of patient selection while minimizing the delay in the start of concomitant treatment, Urba et al has promulgated an approach in larynx cancer in which one cycle of neoadjuvant chemotherapy is delivered to select patients for subsequent concomitant chemoradiotherapy.7 Their published work suggests that this approach offers advantages for survival improvement over historical controls. The group recently expanded their observations to oropharyngeal cancers, reporting 3-year overall survival of 67% (95% CI, 53.5% to 80.6%) and disease-free survival of 80.5% (95% CI, 68.9% to 92.1%) that was superior to historical controls.8

The use of induction chemotherapy primarily in select patients for subsequent therapy is not without issues. For example, the results of Radiation Therapy Oncology Group trial 91-11 demonstrated that among patients who had less than a partial response at the primary site to induction chemotherapy but refused recommended total laryngectomy, durable disease control was still possible with radiation therapy alone. Similarly, it can be difficult to assess response after just one cycle of chemotherapy, raising the possibility that patients who would have done well with radiation-based treatment alone are triaged to unnecessary surgery.9 Finally, newer triplet induction chemotherapy regimens combining cisplatin and fluorouracil with a taxane have proven to be more efficacious than cisplatin and fluorouracil alone.10-12 By focusing on a short course of induction therapy for patient selection purposes, the potential therapeutic benefit of a more prolonged course on distant control may be compromised.

Not surprisingly, there has been a growing interest in defining molecular subgroups that predict disease behavior to select treatment. The work from Kumar and colleagues13 reported in this issue of the Journal aims to define pretreatment molecular markers that predict response to chemotherapy in a clinical trials data set. This well designed study has the clear benefit of identifying markers that may be clinically applicable. The investigators chose to look at established markers individually and in combination based on biologic principles, leading to the identification of prognostically significant molecular predictors.

The genetic analysis of cancers reached a crescendo with the completion of the Human Genome Project along with the development of high throughput genome-wide analytic techniques. These analyses have helped to shape our understanding of human malignancies generally, and head and neck cancers specifically. Global genomic analyses have identified molecular subsets of head and neck squamous cell carcinoma (HNSCC), which may have prognostic implications.14-21 The challenge has been to reproduce prognostic gene signatures. For example, two independent groups have reported gene signatures that predict outcome in breast cancers using gene arrays, one including 70 genes and the other 76 genes.22-25 Interestingly, although both sets of genes have been validated in independent patient cohorts, there is relatively little overlap between the gene sets. Similarly, the use of individual and combined markers to predict outcome in HNSCC has shown conflicting results, as is well illustrated by the variable correlation between p53 status and outcome reported by different investigators.26-32

Divergent findings are not uncommon when assessing individual molecular markers, reflecting the genetic complexity of HNSCC. To overcome these issues, several studies have attempted to define molecular prognostic groups based on global genomic profiles. Several studies have used comparative genomic hybridization (CGH) to categorize prognostic subgroups in HNSCC. Three of these studies define individual prognostic markers among a background of complex genetic aberrations identified by CGH using well defined statistical methods.14,21,33 These studies identified amplifications at 11q13 and 3q26-27 as markers of outcome. The 11q13 amplification has been well studied, with well characterized oncogenes including cyclin D1, EMS1, and TAOS1 as putative targets for 11q13 amplification.34,35 Amplification at 3q26.3 has also been a topic of significant analysis, with several putative genes (including PIK3CA, PKC-, LAMP3, and eIF-5A2) identified at this locus.36-40 Similarly, gene array studies have delineated putative prognostic subsets that predict outcome.15-17,29 Overall, studies using DNA-based assessment have been more reproducible than those that profile using mRNA-based gene arrays. Moreover, even though they are provocative, none of these studies have been validated sufficiently to allow use in routine clinical practice.

Given the significance of response to chemotherapy in HNSCC, several studies have used this as a surrogate end point for defining molecular markers. Predictably, the literature on genome-based assessment of factors predicting response to platinum-based treatment is limited. Studies using CGH suggest that increased chromosomal instability may contribute to tumors containing or acquiring genetic changes that confer treatment resistance in HNSCC, but do not reproducibly identify specific events that predict treatment response. Gene expression array analyses have identified factors that may predict response to chemotherapy. In an interesting study, 45 genes were identified that predicted cisplatin response.18 The accuracy of these markers was 83%, with a positive predictive value of 78% and a negative predictive value of 100% in ovarian and lung cancers. Other studies have also identified prognostic signatures, but there is limited overlap between studies in the gene sets, calling their significance into question.41,42 Moreover, limited information is available for gene sets predicating chemoresponse in HNSCC.15,29

In contrast to specific or combined molecular markers, the definition of patient subgroups by biologic parameters has been much more reliable in predicting outcome. In vitro chemosensitivity testing has been shown by some investigators to be an independent predictor of outcome,43-45 although a technology assessment done by the American Society of Clinical Oncology would suggest that chemopredictive assays are not ready for routine use.46 The presence of human papillomavirus (HPV) in oropharyngeal cancers also predicts outcome. Like cervical cancers, HPV appears to play a causative role in the development of HNSCC.47 A meta-analysis of published trials, including 5,046 HNSCC cancer specimens, shows a 26% prevalence of HPV, with the vast majority being HPV type 16 (HPV-16).48 The predominant location of HPV-associated tumors is in the oropharynx, with a predilection for nonsmokers (up to 50% of cases). Similar to cervical cancers, detection of HPV in HNSCC is associated with sexual history, implicating direct exposure as a cause for infection.49 In addition, immunosuppression has been suggested to increase the risk for infection and development of HPV-related HNSCC.50 Of specific interest, HPV-positive tumors have improved outcomes relative to HPV-negative cases.51 The outcome advantage is maintained in patients treated with chemoradiotherapy, suggesting that HPV status may be a predictor of treatment response.52 This assertion is validated by the studies from Worden et al54 and Kumar et al,13 suggesting that HPV status should be considered in the selection of patients for concomitant chemoradiotherapy. Recent work indicates that the genetic composition of HPV-positive and HPV-negative cancers may be different, suggesting putative molecular markers that may have predictive value.20,53 Kumar and colleagues report an association between p16 and HPV status, suggesting that p16 may serve as a surrogate marker for HPV infection.13,54 Weinberg and colleagues combined the HPV status with the p16 expression level to develop a prognostic categorization for oropharyngeal tumors and showed that it was a treatment response predictor.52,55

Despite these promising findings, the key question that remains is whether any of the putative predictors can be used to individualize treatment selection. Unfortunately, unlike the example of kinase mutations in other solid tumors, the predictive value of individual or combined molecular markers remains insufficient for routine clinical use in HNSCC. Even more importantly, the inherent genetic differences that predict response to chemotherapy remain unidentified, which may not only serve as treatment selectors, but also as therapeutic targets. The current work suggests that combining the epidermal growth factor receptor (EGFR) expression status with HPV status may predict chemoradiotherapy treatment response. Given the success of EGFR targeting in combination with radiation in HNSCC, could targeting EGFR in HPV-negative, EGFR overexpressing cases improve control? These and other possibilities merit investigation.

Notes:
1. published online ahead of print at www.jco.org on May 12, 2008
2. The author(s) indicated no potential conflicts of interest.

References:
1. National Comprehensive Cancer Network Practice Guidelines: Head and Neck Cancer (v.1.2007). http://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf

2. Pfister DG, Laurie SA, Weinstein GS, et al: American Society of Clinical Oncology clinical practice guideline for the use of larynx-preservation strategies in the treatment of laryngeal cancer. J Clin Oncol 24:3693-3704, 2006

3. Forastiere AA, Goepfert H, Maor M, et al: Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer. N Engl J Med 349:2091-2098, 2003

4. Pignon JP, Bourhis J, Domenge C, et al: Chemotherapy added to locoregional treatment for head and neck squamous-cell carcinoma: Three meta-analyses of updated individual data: MACH-NC Collaborative Group: Meta-Analysis of Chemotherapy on Head and Neck Cancer. Lancet 355:949-955, 2000

5. Wolf GT, Fisher SG: Effectiveness of salvage neck dissection for advanced regional metastases when induction chemotherapy and radiation are used for organ preservation. Laryngoscope 102:934-939, 1992

6. Lefebvre JL, Chevalier D, Luboinski B, et al: Larynx preservation in pyriform sinus cancer: Preliminary results of a European Organisation for Research and Treatment of Cancer phase III trial: EORTC Head and Neck Cancer Cooperative Group. J Natl Cancer Inst 88:890-899, 1996

7. Urba S, Wolf G, Eisbruch A, et al: Single-cycle induction chemotherapy selects patients with advanced laryngeal cancer for combined chemoradiation: A new treatment paradigm. J Clin Oncol 24:593-598, 2006

8. Worden FP, Urba S, Bradford C, et al: One cycle of induction chemotherapy (IC) in advanced oropharyngeal cancer (SCCOP) to select patients for organ preservation (OP). J Clin Oncol 23:503s, 2005 (abstr 5512)

9. Pfister DG, Ridge JA: Induction chemotherapy for larynx preservation: Patient selection or therapeutic effect? J Clin Oncol 24:540-543, 2006

10. Vermorken JB, Remenar E, van Herpen C, et al: Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med 357:1695-1704, 2007

11. Posner MR, Hershock DM, Blajman CR, et al: Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N Engl J Med 357:1705-1715, 2007

12. Hitt R, Lopez-Pousa A, Martinez-Trufero J, et al: Phase III study comparing cisplatin plus fluorouracil to paclitaxel, cisplatin, and fluorouracil induction chemotherapy followed by chemoradiotherapy in locally advanced head and neck cancer. J Clin Oncol 23:8636-8645, 2005

13. Kumar B, Cordell K, Lee DJ, et al: EGFR, p16, HPV titer, BCLXL and p53, sex and smoking as indicators of response to therapy and survival in oropharyngeal cancer. J Clin Oncol 26:3128-3137, 2008

14. Bockmuhl U, Schluns K, Kuchler I, et al: Genetic imbalances with impact on survival in head and neck cancer patients. Am J Pathol 157:369-375, 2000

15. Akervall J: Genomic screening of head and neck cancer and its implications for therapy planning. Eur Arch Otorhinolaryngol 263:297-304, 2006

16. Belbin TJ, Singh B, Barber I, et al: Molecular classification of head and neck squamous cell carcinoma using cDNA microarrays. Cancer Res 62:1184-1190, 2002

17. Chung CH, Parker JS, Karaca G, et al: Molecular classification of head and neck squamous cell carcinomas using patterns of gene expression. Cancer Cell 5:489-500, 2004

18. Hsu DS, Balakumaran BS, Acharya CR, et al: Pharmacogenomic strategies provide a rational approach to the treatment of cisplatin-resistant patients with advanced cancer. J Clin Oncol 25:4350-4357, 2007

19. O’Regan EM, Toner ME, Smyth PC, et al: Distinct array comparative genomic hybridization profiles in oral squamous cell carcinoma occurring in young patients. Head Neck 28:330-338, 2006

20. Smeets SJ, Braakhuis BJM, Abbas S, et al: Genome-wide DNA copy number alterations in head and neck squamous cell carcinomas with or without oncogene-expressing human papillomavirus. Oncogene 25:2558-2564, 2006

21. Wreesmann VB, Shi W, Thaler HT, et al: Identification of novel prognosticators of outcome in squamous cell carcinoma of the head and neck. J Clin Oncol 22:3965-3972, 2004

22. Foekens JA, Atkins D, Zhang Y, et al: Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol 24:1665-1671, 2006

23. Wang Y, Klijn JG, Zhang Y, et al: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 365:671-679, 2005

24. van de Vijver MJ, He YD, van’t Veer LJ, et al: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999-2009, 2002

25. van ‘t Veer LJ, Dai H, van de Vijver MJ, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:530-536, 2002

26. Bradford CR, Zhu S, Ogawa H, et al: P53 mutation correlates with cisplatin sensitivity in head and neck squamous cell carcinoma lines. Head Neck 25:654-661, 2003

27. Bradford CR, Zhu S, Poore J, et al: p53 mutation as a prognostic marker in advanced laryngeal carcinoma: Department of Veterans Affairs Laryngeal Cancer Cooperative Study Group. Arch Otolaryngol Head Neck Surg 123:605-609, 1997

28. Bradford CR, Zhu S, Wolf GT, et al: Overexpression of p53 predicts organ preservation using induction chemotherapy and radiation in patients with advanced laryngeal cancer: Department of Veterans Affairs Laryngeal Cancer Study Group. Otolaryngol Head Neck Surg 113:408-412, 1995

29. Ganly I, Talbot S, Carlson D, et al: Identification of angiogenesis/metastases genes predicting chemoradiotherapy response in patients with laryngopharyngeal carcinoma. J Clin Oncol 25:1369-1376, 2007

30. Homma AA, Furuta YY, Oridate NN, et al: Prognostic significance of clinical parameters and biological markers in patients with squamous cell carcinoma of the head and neck treated with concurrent chemoradiotherapy. Clin Cancer Res 5:801-806, 1999

31. Lavertu PP, Adelstein DDJ, Myles JJ, et al: P53 and Ki-67 as outcome predictors for advanced squamous cell cancers of the head and neck treated with chemoradiotherapy. The Laryngoscope 111:1878-1892, 2001

32. Osman I, Sherman E, Singh B, et al: Alteration of p53 pathway in squamous cell carcinoma of the head and neck: Impact on treatment outcome in patients treated with larynx preservation intent. J Clin Oncol 20:2980-2987, 2002

33. Ashman JN, Patmore HS, Condon LT, et al: Prognostic value of genomic alterations in head and neck squamous cell carcinoma detected by comparative genomic hybridisation. Br J Cancer 89:864-869, 2003

34. Huang X, Gollin SM, Raja S, et al: High-resolution mapping of the 11q13 amplicon and identification of a gene, TAOS1, that is amplified and overexpressed in oral cancer cells. Proc Natl Acad Sci U S A 99:11369-11374, 2002

35. Rodrigo JP, Garcia LA, Ramos S, et al: EMS1 gene amplification correlates with poor prognosis in squamous cell carcinomas of the head and neck. Clin Cancer Res 6:3177-3182, 2000

36. Singh B, Reddy PG, Goberdhan A, et al: p53 regulates cell survival by inhibiting PIK3CA in squamous cell carcinomas. Genes Dev 16:984-993, 2002

37. Eder AM, Sui X, Rosen DG, et al: Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci U S A 102:12519-12524, 2005

38. Kanao H, Enomoto T, Kimura T, et al: Overexpression of LAMP3/TSC403/DC-LAMP promotes metastasis in uterine cervical cancer. Cancer Res 65:8640-8645, 2005

39. Regala RP, Weems C, Jamieson L, et al: Atypical protein kinase Ciota plays a critical role in human lung cancer cell growth and tumorigenicity. J Biol Chem 280:31109-31115, 2005

40. Brass N, Heckel D, Sahin U, et al: Translation initiation factor eIF-4gamma is encoded by an amplified gene and induces an immune response in squamous cell lung carcinoma. Hum Mol Genet 6:33-39, 1997

41. Kerley-Hamilton JS, Pike AM, Li N, et al: A p53-dominant transcriptional response to cisplatin in testicular germ cell tumor-derived human embryonal carcinoma. Oncogene 24:6090-6100, 2005

42. Dressman HK, Berchuck A, Chan G, et al: An integrated genomic-based approach to individualized treatment of patients with advanced-stage ovarian cancer. J Clin Oncol 25:517-525, 2007

43. Singh B, Li R, Xu L, et al: Prediction of survival in patients with head and neck cancer using the histoculture drug response assay. Head Neck 24:437-442, 2002

44. Ohie S, Udagawa Y, Aoki D, et al: Histoculture drug response assay to monitor chemoresponse. Methods Mol Med 110:79-86, 2005

45. Furukawa T, Kubota T, Hoffman RM: Clinical applications of the histoculture drug response assay. Clin Cancer Res 1:305-311, 1995

46. Schrag D, Garewal HS, Burstein HJ, et al: American Society of Clinical Oncology Technology Assessment: Chemotherapy sensitivity and resistance assays. J Clin Oncol 22:3631-3638, 2004

47. Gillison ML, Koch WM, Capone RB, et al: Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 92:709-720, 2000

48. Kreimer AR, Clifford GM, Boyle P, et al: Human papillomavirus types in head and neck squamous cell carcinomas worldwide: A systematic review. Cancer Epidemiol Biomarkers Prev 14:467-475, 2005

49. D’Souza G, Kreimer AR, Viscidi R, et al: Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356:1944-1956, 2007

50. Kutler DI, Wreesmann VB, Goberdhan A, et al: Human papillomavirus DNA and p53 polymorphisms in squamous cell carcinomas from Fanconi anemia patients.J Natl Cancer Inst 95:1718-1721, 2003

51. Fakhry C, Gillison ML: Clinical implications of human papillomavirus in head and neck cancers. J Clin Oncol 24:2606-2611, 2006

52. Weinberger PM, Yu Z, Haffty BG, et al: Molecular classification identifies a subset of human papillomavirus–associated oropharyngeal cancers with favorable prognosis. J Clin Oncol 24:736-747, 2006

53. Strati K, Pitot HC, Lambert PF: Identification of biomarkers that distinguish human papillomavirus (HPV)-positive versus HPV-negative head and neck cancers in a mouse model. Proc Natl Acad Sci U S A 103:14152-14157, 2006

54. Worden F, Kumar B, Lee J, et al: Chemoselection as a strategy for organ preservation in advanced oropharynx cancer: Response an survival positively associated with HPV16 copy number. J Clin Oncol 26:3138-3146, 2008

55. Weinberger P, Kountourakis P, Sasaki C, et al: Oropharyngeal squamous cell cancers (OSCC) bearing transcriptionally active human papillomavirus (HPV) display distinct protein expression profile. J Clin Oncol 24:, 2006 (abstr 10028)