Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Pre-treatment tumour perfusion parameters and initial RECIST response do not predict long-term survival outcomes for patients with head and neck squamous cell carcinoma treated with induction chemotherapy

  • Natalie M. Lowe,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Lucy E. Kershaw,

    Roles Data curation, Formal analysis, Methodology, Writing – review & editing

    Affiliations Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom, Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Jonathan M. Bernstein,

    Roles Data curation, Investigation, Methodology, Project administration, Validation, Writing – review & editing

    Affiliations Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom, Department of Otolaryngology—Head & Neck Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom

  • Stephanie B. Withey,

    Roles Formal analysis

    Affiliation Medical Physics, University Hospitals Birmingham, Birmingham, United Kingdom

  • Kathleen Mais,

    Roles Investigation

    Affiliation Head and Neck Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Jarrod J. Homer,

    Roles Conceptualization, Writing – review & editing

    Affiliations Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom, University Department of Otolaryngology—Head & Neck Surgery, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom

  • Nicholas J. Slevin,

    Roles Conceptualization, Funding acquisition, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    Affiliations Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom, Head and Neck Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Suzanne C. Bonington,

    Roles Conceptualization, Formal analysis, Methodology, Resources, Validation, Writing – review & editing

    Affiliation Department of Radiology, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Bernadette M. Carrington,

    Roles Conceptualization, Formal analysis, Methodology, Project administration, Validation, Writing – review & editing

    Affiliation Department of Radiology, The Christie NHS Foundation Trust, Manchester, United Kingdom

  • Catharine M. West

    Roles Conceptualization, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    Catharine.West@manchester.ac.uk

    Affiliations Division of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom

Abstract

Objectives

Previously, we showed that pre-treatment tumour plasma perfusion (Fp) predicts RECIST response to induction chemotherapy (ICT) in locoregionally advanced head and neck squamous cell carcinoma (HNSCC). The aim here was to determine whether the pre-treatment tumour Fp estimate, changes in tumour Fp or RECIST response post 2 cycles of ICT were prognostic for long-term survival outcomes.

Methods

A prospective study enrolled patients with high stage HNSCC treated with docetaxel (T), cisplatin (P) and 5-fluorouracil (F) (ICT) followed by synchronous cisplatin and intensity modulated radiotherapy. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) before and after two cycles of ICT was used to measure Fp and RECIST response.

Results

Forty-two patients were recruited and 37 underwent two scans. The median follow-up was 36 (range 23–49) months. Pre-treatment tumour Fp (stratified by median) was not prognostic for overall survival (p = 0.42), disease specific survival (p = 0.20) and locoregional control (p = 0.64). Neither change in tumour Fp nor RECIST response post two cycles of ICT was prognostic for any outcome (p>0.21).

Conclusion

DCE-MRI parameters do not predict long-term survival outcomes following ICT and RECIST response to ICT may not be an appropriate endpoint to determine early efficacy of a treatment in HNSCC patients.

Introduction

Head and neck cancer is one of the world’s leading malignancies with an estimated global incidence of over 686,000 cases in 2012. In Europe in 2013, head and neck cancer contributed 135,400 new oncology diagnoses and 61,300 deaths.[1] Concurrent chemoradiotherapy is the non-surgical standard of care for patients who present with high stage disease.[24] Despite advances in chemoradiotherapy, unlike some other cancer sites where survival rates rose substantially in recent decades, the improvement in head and neck cancer survival rates has been modest. As chemotherapy primarily acts as a radiosensitiser for locoregional treatment in head and neck squamous cell carcinoma (HNSCC),[5] the use of induction chemotherapy (ICT) has been explored to tackle distant metastases.[6] The preferred ICT regimen comprises a taxane (T), platinum agent (P) and 5-fluorouracil (F) (TPF) [710] and has been shown in several studies to lower distant metastasis rates compared with CRT alone.[5, 7, 1113] Despite this, there is controversy as to whether this translates to an improved overall survival outcome.[14, 15] It has been suggested, however, that as well as several problems with poor methodology undermining the applicability of trials comparing ICT to CRT alone; trials have included patients that are unlikely to benefit from the potential advantages of ICT such as those with a low risk of distant metastases, hence diluting any positive effects.[14] Tumour heterogeneity also affects response to treatment.[16] The key to extracting the benefits of ICT may be meticulous patient and tumour selection.

Three cycles of TPF take nine weeks to complete. Approximately 30% of patients do not respond to ICT and hence may have their definitive treatment delayed for little if any benefit.[8, 17] Methods to detect prior to or early in the course of ICT which patients are unlikely to respond would identify patients who should be directed immediately to CRT to prevent delays in definitive locoregional treatment.

Zima et al [18, 19] showed that HNSCCs with elevated blood volume and blood flow detected by pre-treatment computed tomography (CT) perfusion imaging had a good response to ICT. Petralia et al [20] also found baseline tumour blood volume in patients with upper aerodigestive tract squamous cell carcinomas was significantly lower in non-responders to ICT as demonstrated by perfusion CT. Our group set up a study to investigate whether similar findings were seen with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), and showed that pre-treatment tumour plasma perfusion (Fp) predicts response to ICT.[17] Whether the prediction of early response to ICT relates to long-term survival outcomes, however, is not known. This study therefore investigated whether the pre-treatment tumour Fp estimate, plus changes in tumour Fp or RECIST response post 2 cycles of ICT were prognostic for long-term survival outcomes.

Materials and methods

Patients

Ethical approval was granted by The North West 1 Research Ethics Committee (ref: 11/H1017/5) and The Christie Research and Development Department for a prospective open cohort study to recruit 50 patients. Recruitment started in March 2011 and the 50th patient was recruited in July 2013. All patients gave written informed consent. Patients were eligible if they had histologically or cytologically proven stage IV HNSCC (staged according to American Joint Committee on Cancer (AJCC) using the tumour, node, metastases (TNM) system [21]) and had been referred for treatment with three cycles of ICT followed by CRT as decided by The Christie Head and Neck multidisciplinary team. ICT was a modified version of the TAX 323 doses [7] planned as three cycles of: docetaxel (75 mg/m2 IV on day 1), cisplatin (75 mg/m2 IV on day 1) and 5-FU (750 mg/m2 IV on days 2–5) followed by two weeks of rest. Following the two-week rest period after the third cycle of TPF, patients were given concurrent CRT involving intensity-modulated radiation therapy (IMRT) with 66 Gy in 30 fractions plus concurrent cisplatin (100 mg/m2 IV) at the beginning of week 1 and week 4 (day 1 and 22). It has been suggested that ICT and CRT provide different benefits for advanced HNSCC with CRT mainly producing locoregional control and ICT controlling distant metastases hence complimenting each other’s strengths.[5] As part of this study was to understand which patients may or may not benefit from ICT as an neoadjuvant to the current gold standard CRT treatment and as efforts to determine a standard CRT regimen post ICT are ongoing separately [6]; we used this well recognised chemoradiotherapy regimen suitable for use as a primary therapy without ICT as per Pignon 2009.[5] Patients were excluded from the trial if they had undergone any previous treatment for a head and neck carcinoma. P16 status was determined as described elsewhere.[17]

Imaging protocol

Baseline imaging was acquired in the three weeks before ICT. The imaging protocol is described in detail elsewhere.[17] Briefly, the examination consisted of conventional staging scans followed by high resolution T2-weighted (w) imaging, a saturation-recovery measurement of T1, and dynamic T1w imaging for a total of 7 minutes during which a bolus injection of 0.1 mmol/kg Gadobutrol (Gd) was administered in an antecubital vein. DCE-MRI analysis was on a whole-tumour region of interest basis (outlined on high resolution T2w images by two radiologists in consensus). The two compartment exchange model [22] was fitted to the Gd concentration vs time curves, which were converted from signal intensity vs time curves using a precontrast measurement of T1.[23] Arterial input functions were obtained from the internal carotid artery on a patient by patient basis using an automated procedure.[24]

A further DCE-MRI examination was acquired during the two-week window between the second and third cycles of ICT. Fp of the primary tumour and largest regional lymph node was determined on the second scan. Patients were divided into complete responders, partial responders and those with stable disease according to the Response Evaluation Criteria in Solid Tumours (RECIST 1.1).[25]

Study design

The prospective study was designed and powered to detect early response to ICT based on published data. The power calculation indicated a need for 38 patients to have a pre-treatment DCE-MRI scan to detect a one standard deviation difference in microvascular parameters with 77% power. Fifty patients were recruited to the trial and 42 completed the baseline DCE-MRI scan. Thirty-seven patients completed both the baseline DCE-MRI scan and the follow-up scan after two cycles of ICT. Fig 1 shows a flow diagram indicating the reasons for exclusion from the trial. The results of the primary analysis of the study have been published.[17] Secondary endpoints of the study were to assess relationships with long-term outcomes for: baseline Fp of the primary lesion and / or largest regional lymph node; the change in Fp of the primary lesion and / or largest regional lymph node between the baseline DCE-MRI scan and the second DCE-MRI scan (post two cycles of ICT); and RECIST response between the baseline and second DCE-MRI scan (post two cycles of ICT) using target lesions from both the primary tumour and nodal metastases as per RECIST guidelines version 1.1. [25]

thumbnail
Fig 1. Flow diagram showing patient inclusion and exclusion from the trial.

https://doi.org/10.1371/journal.pone.0194841.g001

Statistical analysis

IBM SPSS Statistics 22 (IBM, Armonk, USA) was used for all statistical analyses. Patients were stratified by median Fp of the primary tumour and by the largest regional lymph node. Kaplan-Meier curves were plotted for these groups with respect to overall survival (OS), disease-specific survival (DSS) and locoregional control (LRC) measured from date of first day of treatment. Fp values were also determined in patients undergoing the second DCE-MRI scan (after two cycles of ICT). Pre-treatment values were then subtracted to obtain a value for the change in Fp. Patients were again stratified by median Fp of the primary tumour and largest regional lymph node. Patients were also stratified by RECIST response defined as complete or partial (CR or PR) versus stable or progressive disease (SD or PD). Cox regression was used to assess differences in outcome between all groups.

Results

Table 1 summarizes the patient characteristics for the group. Median follow-up in surviving patients was 36 months (range 23–49 months). ICT was stopped and treatment changed to palliation in one patient following identification of pre-treatment lung metastases. This patient was included in the OS analyses (on an intention to treat basis) but was excluded from the DSS and LRC analyses.

Median survival was not calculated because a 50% event rate was not reached for any group. At a median 3-year follow up OS for the whole group was 74.1%, DSS 87.6% and LRC 87.4%. Median tumour Fp was 43.8 ml/100ml/min, interquartile range 25.5–81.5 ml/100ml/min. As shown in Table 2, there were no statistically significant differences in OS (p = 0.42), DSS (p = 0.20) or LRC (p = 0.64) for patients with low versus high pre-treatment tumour Fp. Median nodal Fp was 28.19ml/100ml/min, interquartile range 18.01–55.20 ml/100ml/min. There were no statistically significant differences in OS (p = 0.88), DSS (p = 0.30) or LRC (p = 0.92) for patients with low versus high nodal Fp. Fig 2 shows the Kaplan-Meier curves for tumour and nodal Fp versus OS, DSS and LRC.

thumbnail
Fig 2.

Tumour plasma perfusion (Fp) in relation to overall survival (a), disease specific survival (b) and locoregional control (c). Nodal plasma perfusion in relation to survival (d) disease specific survival (e) and locoregional control (f).

https://doi.org/10.1371/journal.pone.0194841.g002

thumbnail
Table 2. Results from univariable Cox regression analyses.

https://doi.org/10.1371/journal.pone.0194841.t002

Change in primary tumour Fp was available for 37 patients and change in nodal Fp for 32 patients. Using Cox regression analysis, changes in Fp between the pre-treatment DCE-MRI and the DCE-MRI post two cycles of ICT had no prognostic significance (Table 2).

RECIST response between the pre-treatment MRI and the MRI post two cycles of ICT was available in 37 patients. One patient had CR, 23 patients had PR, 13 patients had SD and no patients had PD. The outcomes for patients with a CR or PR versus SD were not statistically significant for any outcome measures as shown in Table 2. Fig 3 shows Kaplan-Meier curves for RECIST response versus outcome.

thumbnail
Fig 3.

RECIST response to induction chemotherapy in relation to overall survival (a), disease specific survival (b) and locoregional control (c).

https://doi.org/10.1371/journal.pone.0194841.g003

Results from univariable analysis of smoking status, performance status,[26] p16 status, tumour differentiation and nodal grade with regards to survival outcome are beyond the scope of this article but can be found in supporting information S1 Table.

Discussion

The primary analysis of the study showed that high pre-treatment tumour Fp evaluated using pre-treatment DCE-MRI imaging in high stage HNSCC predicts initial response to ICT.[17] The secondary analysis reported here showed that this early prediction of good response to ICT did not correlate with good long term outcomes. Also, neither change in plasma perfusion post two cycles of ICT nor RECIST response were prognostic for long-term survival outcomes.

The tumour microenvironment affects the delivery and penetration of chemotherapy to malignant cells and so impaired tumour perfusion leads to a poor treatment response.[2729] It is therefore reasonable to expect that Fp (Fp is equivalent to blood flow [BF] adjusted for haematocrit) might be prognostic for long term outcomes. Indeed, several HNSCC studies show pre-treatment tumour hypoxia and haemodynamic imaging parameters such as blood volume (BV) and BF are prognostic for survival outcomes [3037] although there are conflicting reports.[38] However, these studies were performed on patients receiving definitive locoregional treatment such as XRT or CRT rather than ICT. Published trials assessing perfusion parameters relating to ICT predominantly report initial response to ICT treatment only.[1820] Hence, whilst there is evidence that tumour perfusion parameters can predict immediate response to ICT, ours is one of the first studies to assess long term outcomes. Despite pre-treatment Fp predicting initial response to ICT,[17] it does not equate to long term survival benefit. This result was echoed by Bisdas et al who found that baseline BV and BF predicted initial ICT response. However, it was noted that progression free survival in the responder group was not significantly different to the non-responders group (p = 0.80).[39] Nevertheless, stratifying patients to different treatments based on initial ICT response is currently performed in clinical practice. The RTOG 91–11 study [40] designed a trial modelled on the Department of Veterans Affairs Laryngeal Cancer Study Group trial of 1991 where patients in the ICT arm received either a further cycle of ICT then radiotherapy or salvage surgery depending on their response post 2 cycles of ICT. Urba et al 2006 [41] also suggested that single cycle ICT selects patients for organ sparing treatment rather than total laryngectomy depending on initial ICT response. There are differences in what constitutes a good or poor response to treatment ranging from “no response” to “50% response” to a “major response” and varying numbers of preliminary cycles of ICT given. In clinical practice, initial response to ICT is thought to suggest a tumour’s inherent treatment responsiveness and is considered an aid for subsequent treatment decisions. Our study suggests, however, that this practice of using response to ICT as a tool to stratify patients to different locoregional treatments may need to be revisited as an inadequate response to ICT may not translate into a poor outcome post CRT.[41] Larger trials are needed to verify our results.

A possible explanation for the lack of prognostic significance might be intra-tumoural heterogeneity. Tumours can have sub-regions with variable blood flow, architecture, metabolism, cell proliferation, genotypes and phenotypes.[4245] Gerlinger et al reported that “gene expression signatures of poor and good prognosis were detected in different regions of the same tumour”.[44] ICT is used predominantly to tackle distant micrometastases as an adjunct to definitive treatment and although potentially producing considerable tumour and nodal shrinkage, there is only a complete response in 0–40% of patients.[17, 20] It may be that the prognostic features of the remaining tissue are what ultimately determine the response to locoregional treatment and survival outcome. This may explain some of the discrepancies between findings based on patients treated solely with locoregional treatment of (chemo)radiotherapy and those treated with ICT first which best tackles distant metastases.

In 1996 the Food and Drug Administration stated that more cancer drugs would be granted accelerated approval if their benefit could be demonstrated by objective evidence of tumour shrinkage.[46] It was noted that evidence of better survival could now be demonstrated later implying that tumour shrinkage and survival benefit were related. There is evidence that in several cancer sites that this is the case.[47, 48] Studies including a meta-analysis by El-Maraghi and Eisenhauer showed that objective tumour response was a useful endpoint in phase II trials for several solid tumours as its observation predicted for eventual success in phase III trials.[49, 50] However, HNSCC were not included in these trials and the result was not seen in all subsites studied.

The relationship between objective tumour shrinkage (RECIST response) and long term outcomes has not been extensively studied in HNSCC but, like our study, those performed in HNSCC suggest RECIST response cannot be used a surrogate marker for long term survival outcomes. Passero et al found no relationship between a complete RECIST response measured by CT and progression free survival (PFS) in 53 HNSCC patients treated with concurrent CRT +/- ICT.[51] Matoba et al found no relationship between RECIST response measured using MR prior to and eight weeks post CRT on locoregional control or survival.[52] Studies have shown that RECIST response measured by CT does not correlate with pathological response in HNSCC, which may explain why RECIST response is an unreliable predictor of long-term outcome.[53, 54] Hence, although a widely used method of measuring change in tumour size in regular clinical use and as part of the reporting of trial outcomes, RECIST response appears to have no value as a prognostic factor for long-term survival outcomes in HNSCC. This finding has practical implications as RECIST response is used frequently in clinical practice and trial reporting despite a lack of evidence supporting its use for HNSCC. There is, therefore, a need for larger clinical trials investigating initial RECIST response to treatment in relation to long term survival outcomes, which need to investigate the effect of the treatment regimen used, i.e. CRT alone or with additional ICT.

Limitations of this study are that it is single-centre and non-randomized. Also, the study was powered to assess the relationship between pre-treatment DCE-MRI parameters and RECIST response rather than relationships with long-term outcomes. Strengths are that it is prospective and focuses on the patient group most likely to benefit from TPF ICT, i.e., those with stage IV disease (and hence high risk of distant metastases). This cohort, therefore, is a representative group of patients that is most likely to be offered this treatment regimen.

Regarding future studies, larger trials are required to validate our study results in relation to using initial ICT response and RECIST response as an aid to treatment stratification. Further studies into intra-tumour heterogeneity and how it correlates with outcomes following ICT are also required.

Conclusions

Pre-treatment tumour Fp, change in tumour Fp measured using DCE-MRI and RECIST response post two cycles of ICT are not prognostic for long-term survival outcome in HNSCC patients treated with ICT followed by chemotherapy and IMRT. Intra-tumour heterogeneity post ICT may explain the inability to predict long-term outcomes prior to treatment. RECIST response to ICT may not be an appropriate endpoint to determine efficacy of treatment in a clinical or phase II trial setting for HNSCC patients.

Supporting information

S1 Table. Results from univariable analyses using Cox regression.

https://doi.org/10.1371/journal.pone.0194841.s001

(DOCX)

Acknowledgments

The authors thank Helen Valentine for help with p16 staining; Farag Shuweidi and Clare Hodgson for help with statistical analysis and Jason Kennedy for database management. The work was funded by the Christie Hospital Charitable Trust.

References

  1. 1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer. 2014:n/a-n/a. pmid:25220842
  2. 2. Blanchard P, Hill C, Guihenneuc-Jouyaux C, Baey C, Bourhis J, Pignon JP. Mixed treatment comparison meta-analysis of altered fractionated radiotherapy and chemotherapy in head and neck cancer. Journal of clinical epidemiology. 2011;64(9):985–92. pmid:21330105
  3. 3. Nguyen-Tan PF, Zhang Q, Ang KK, Weber RS, Rosenthal DI, Soulieres D, et al. Randomized Phase III Trial to Test Accelerated Versus Standard Fractionation in Combination With Concurrent Cisplatin for Head and Neck Carcinomas in the Radiation Therapy Oncology Group 0129 Trial: Long-Term Report of Efficacy and Toxicity. Journal of Clinical Oncology. 2014. pmid:25366680
  4. 4. Bourhis J, Sire C, Graff P, Gregoire V, Maingon P, Calais G, et al. Concomitant chemoradiotherapy versus acceleration of radiotherapy with or without concomitant chemotherapy in locally advanced head and neck carcinoma (GORTEC 99–02): an open-label phase 3 randomised trial. The Lancet Oncology. 2012;13(2):145–53. Epub 2012/01/21. pmid:22261362.
  5. 5. Pignon J-P, Maître Al, Maillard E, Bourhis J. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): An update on 93 randomised trials and 17,346 patients. Radiotherapy and Oncology. 2009;92(1):4–14. pmid:19446902
  6. 6. Ghi M, Paccagnella A, Ferrari D, Foa P, Alterio D, Codecà C, et al. Induction TPF followed by concomitant treatment versus concomitant treatment alone in locally advanced Head and Neck Cancer. A phase II-III trial. Annals of Oncology. 2017.
  7. 7. Vermorken JB, Remenar E, van Herpen C, Gorlia T, Mesia R, Degardin M, et al. Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. The New England journal of medicine. 2007;357(17):1695–704. Epub 2007/10/26. pmid:17960012.
  8. 8. Posner MR, Hershock DM, Blajman CR, Mickiewicz E, Winquist E, Gorbounova V, et al. Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. The New England journal of medicine. 2007;357(17):1705–15. Epub 2007/10/26. pmid:17960013.
  9. 9. Lorch JH, Goloubeva O, Haddad RI, Cullen K, Sarlis N, Tishler R, et al. Induction chemotherapy with cisplatin and fluorouracil alone or in combination with docetaxel in locally advanced squamous-cell cancer of the head and neck: long-term results of the TAX 324 randomised phase 3 trial. The Lancet Oncology. 2011;12(2):153–9. Epub 2011/01/15. pmid:21233014.
  10. 10. Hitt R, Grau JJ, Lopez-Pousa A, Berrocal A, Garcia-Giron C, Irigoyen A, et al. A randomized phase III trial comparing induction chemotherapy followed by chemoradiotherapy versus chemoradiotherapy alone as treatment of unresectable head and neck cancer. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2014;25(1):216–25. Epub 2013/11/22. pmid:24256848.
  11. 11. Ma J, Liu Y, Huang XL, Zhang ZY, Myers JN, Neskey DM, et al. Induction chemotherapy decreases the rate of distant metastasis in patients with head and neck squamous cell carcinoma but does not improve survival or locoregional control: a meta-analysis. Oral oncology. 2012;48(11):1076–84. Epub 2012/07/18. pmid:22800881.
  12. 12. Cohen EW, Karrison T, Kocherginsky M, al e. Docetaxel based chemoradiotherapy plus or minus induction chemotherapy to decrease events in head and neck cancer (DeCIDE) Journal of Clinical Oncology. 2012;30(s).
  13. 13. Cohen EE, Karrison TG, Kocherginsky M, Mueller J, Egan R, Huang CH, et al. Phase III randomized trial of induction chemotherapy in patients with N2 or N3 locally advanced head and neck cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(25):2735–43. Epub 2014/07/23. pmid:25049329.
  14. 14. Stokes WA, Amini A, Jones BL, McDermott JD, Raben D, Ghosh D, et al. Survival impact of induction chemotherapy in advanced head and neck cancer: A National Cancer Database analysis. Head & neck. 2017;39(6):1113–21. Epub 2017/03/17. pmid:28301079.
  15. 15. Ghi MG, Paccagnella A, Ferrari D, Foa P, Alterio D, Codeca C, et al. Induction TPF followed by concomitant treatment versus concomitant treatment alone in locally advanced head and neck cancer. A phase II-III trial. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2017;28(9):2206–12. Epub 2017/09/16. pmid:28911070.
  16. 16. Wang P, Popovtzer A, Eisbruch A, Cao Y. An approach to identify, from DCE MRI, significant subvolumes of tumors related to outcomes in advanced head-and-neck cancer. Medical physics. 2012;39(8):5277–85. Epub 2012/08/17. pmid:22894453; PubMed Central PMCID: PMCPmc3422362.
  17. 17. Bernstein JM, Kershaw LE, Withey SB, Lowe NM, Homer JJ, Slevin NJ, et al. Tumor plasma flow determined by dynamic contrast-enhanced MRI predicts response to induction chemotherapy in head and neck cancer. Oral oncology. 2015;51(5):508–13. Epub 2015/02/24. pmid:25700703.
  18. 18. Zima A, Carlos R, Gandhi D, Case I, Teknos T, Mukherji SK. Can pretreatment CT perfusion predict response of advanced squamous cell carcinoma of the upper aerodigestive tract treated with induction chemotherapy? Am J Neuroradiol. 2007;28(2):328–34. pmid:17297007
  19. 19. Gandhi D, Chepeha DB, Miller T, Carlos RC, Bradford CR, Karamchandani R, et al. Correlation between initial and early follow-up CT perfusion parameters with endoscopic tumor response in patients with advanced squamous cell carcinomas of the oropharynx treated with organ-preservation therapy. Am J Neuroradiol. 2006;27(1):101–6. pmid:16418366
  20. 20. Petralia G, Preda L, Giugliano G, Jereczek-Fossa BA, Rocca A, D'Andrea G, et al. Perfusion computed tomography for monitoring induction chemotherapy in patients with squamous cell carcinoma of the upper aerodigestive tract: correlation between changes in tumor perfusion and tumor volume. Journal of computer assisted tomography. 2009;33(4):552–9. Epub 2009/07/30. pmid:19638848.
  21. 21. Edge SE, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC Cancer Staging Manual. 7th edition ed. New York, USA: Spinger; 2009.
  22. 22. Brix G, Kiessling F, Lucht R, Darai S, Wasser K, Delorme S, et al. Microcirculation and microvasculature in breast tumors: pharmacokinetic analysis of dynamic MR image series. Magnetic resonance in medicine. 2004;52(2):420–9. Epub 2004/07/30. pmid:15282828.
  23. 23. Brookes JA, Redpath TW, Gilbert FJ, Murray AD, Staff RT. Accuracy of T1 measurement in dynamic contrast-enhanced breast MRI using two- and three-dimensional variable flip angle fast low-angle shot. Journal of magnetic resonance imaging: JMRI. 1999;9(2):163–71. Epub 1999/03/17. pmid:10077009.
  24. 24. Donaldson SB, Bonington SC, Kershaw LE, Cowan R, Lyons J, Elliott T, et al. Dynamic contrast-enhanced MRI in patients with muscle-invasive transitional cell carcinoma of the bladder can distinguish between residual tumour and post-chemotherapy effect. European journal of radiology. 2013;82(12):2161–8. Epub 2013/09/17. pmid:24034835.
  25. 25. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). European journal of cancer (Oxford, England: 1990). 2009;45(2):228–47. Epub 2008/12/23. pmid:19097774.
  26. 26. Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. American journal of clinical oncology. 1982;5(6):649–55. Epub 1982/12/01. pmid:7165009.
  27. 27. Pascal J, Bearer EL, Wang Z, Koay EJ, Curley SA, Cristini V. Mechanistic patient-specific predictive correlation of tumor drug response with microenvironment and perfusion measurements. Proceedings of the National Academy of Sciences. 2013;110(35):14266–71. pmid:23940372
  28. 28. Sinek J, Frieboes H, Zheng X, Cristini V. Two-dimensional chemotherapy simulations demonstrate fundamental transport and tumor response limitations involving nanoparticles. Biomedical microdevices. 2004;6(4):297–309. Epub 2004/11/19. pmid:15548877.
  29. 29. Jain RK. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science (New York, NY). 2005;307(5706):58–62. pmid:15637262
  30. 30. Hermans R, Meijerink M, Van den Bogaert W, Rijnders A, Weltens C, Lambin P. Tumor perfusion rate determined noninvasively by dynamic computed tomography predicts outcome in head-and-neck cancer after radiotherapy. International journal of radiation oncology, biology, physics. 2003;57(5):1351–6. Epub 2003/11/25. pmid:14630273.
  31. 31. Brizel DM, Dodge RK, Clough RW, Dewhirst MW. Oxygenation of head and neck cancer: changes during radiotherapy and impact on treatment outcome. Radiother Oncol. 1999;53(2):113–7. Epub 2000/02/09. pmid:10665787.
  32. 32. Nordsmark M, Bentzen SM, Rudat V, Brizel D, Lartigau E, Stadler P, et al. Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiotherapy and oncology: journal of the European Society for Therapeutic Radiology and Oncology. 2005;77(1):18–24. Epub 2005/08/16. pmid:16098619.
  33. 33. Bisdas S, Rumboldt Z, Surlan-Popovic K, Baghi M, Koh TS, Vogl TJ, et al. Perfusion CT in squamous cell carcinoma of the upper aerodigestive tract: long-term predictive value of baseline perfusion CT measurements. AJNR Am J Neuroradiol. 2010;31(3):576–81. Epub 2009/10/31. pmid:19875471.
  34. 34. Stadler P, Becker A, Feldmann HJ, Hansgen G, Dunst J, Wurschmidt F, et al. Influence of the hypoxic subvolume on the survival of patients with head and neck cancer. Int J Radiat Oncol Biol Phys. 1999;44(4):749–54. Epub 1999/07/01. pmid:10386631.
  35. 35. Popovic KS, Lukic S, Popovic P. Pretreatment perfusion CT and CT volumetry in squamous cell carcinoma of the head and neck region. Journal of BUON: official journal of the Balkan Union of Oncology. 2014;19(4):937–43. Epub 2014/12/24. pmid:25536599.
  36. 36. Ursino S, Faggioni L, Fiorica F, Delishaj D, Seccia V, Pasqualetti F, et al. Role of perfusion CT in the evaluation of metastatic nodal tumor response after radiochemotherapy in head and neck cancer: preliminary findings. European review for medical and pharmacological sciences. 2017;21(21):4882–90. Epub 2017/11/23. pmid:29164573.
  37. 37. Truong MT, Saito N, Ozonoff A, Wang J, Lee R, Qureshi MM, et al. Prediction of locoregional control in head and neck squamous cell carcinoma with serial CT perfusion during radiotherapy. AJNR American journal of neuroradiology. 2011;32(7):1195–201. Epub 2011/07/16. pmid:21757530.
  38. 38. Cao Y, Popovtzer A, Li D, Chepeha DB, Moyer JS, Prince ME, et al. Early prediction of outcome in advanced head-and-neck cancer based on tumor blood volume alterations during therapy: a prospective study. International journal of radiation oncology, biology, physics. 2008;72(5):1287–90. Epub 2008/11/26. pmid:19028268; PubMed Central PMCID: PMCPmc3638953.
  39. 39. Bisdas S, Rumboldt Z, Wagenblast J, Baghi M, Koh TS, Hambek M, et al. Response and Progression-Free Survival in Oropharynx Squamous Cell Carcinoma Assessed by Pretreatment Perfusion CT: Comparison with Tumor Volume Measurements. American Journal of Neuroradiology. 2009;30(4):793. pmid:19351906
  40. 40. Weber RS, Berkey BA, Forastiere A, Cooper J, Maor M, Goepfert H, et al. Outcome of salvage total laryngectomy following organ preservation therapy: the Radiation Therapy Oncology Group trial 91–11. Archives of otolaryngology—head & neck surgery. 2003;129(1):44–9. Epub 2003/01/15. pmid:12525193.
  41. 41. Urba S, Wolf G, Eisbruch A, Worden F, Lee J, Bradford C, et al. Single-cycle induction chemotherapy selects patients with advanced laryngeal cancer for combined chemoradiation: a new treatment paradigm. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2006;24(4):593–8. Epub 2005/12/29. pmid:16380415.
  42. 42. O'Connor JP, Rose CJ, Waterton JC, Carano RA, Parker GJ, Jackson A. Imaging intratumor heterogeneity: role in therapy response, resistance, and clinical outcome. Clinical cancer research: an official journal of the American Association for Cancer Research. 2015;21(2):249–57. Epub 2014/11/26. pmid:25421725.
  43. 43. Balluff B, Frese CK, Maier SK, Schone C, Kuster B, Schmitt M, et al. De novo discovery of phenotypic intratumour heterogeneity using imaging mass spectrometry. The Journal of pathology. 2015;235(1):3–13. Epub 2014/09/10. pmid:25201776.
  44. 44. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. The New England journal of medicine. 2012;366(10):883–92. Epub 2012/03/09. pmid:22397650.
  45. 45. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nature reviews Cancer. 2012;12(5):323–34. pmid:22513401
  46. 46. Clinton B, Gore A, National Performance R. Reinventing the regulation of cancer drugs: accelerating approval and expanding access. [Washington, D.C.?]: National Performance Review; 1996. 12 p. p.
  47. 47. Paesmans M, Sculier JP, Libert P, Bureau G, Dabouis G, Thiriaux J, et al. Response to chemotherapy has predictive value for further survival of patients with advanced non-small cell lung cancer: 10 years experience of the european lung cancer working party. Eur J Cancer. 1997;33(14):2326–32. http://dx.doi.org/10.1016/S0959-8049(97)00325-0. pmid:9616276
  48. 48. Buyse M, Thirion P, Carlson RW, Burzykowski T, Molenberghs G, Piedbois P. Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis. The Lancet. 2000;356(9227):373–8. http://dx.doi.org/10.1016/S0140-6736(00)02528-9.
  49. 49. Goffin J, Baral S, Tu D, Nomikos D, Seymour L. Objective responses in patients with malignant melanoma or renal cell cancer in early clinical studies do not predict regulatory approval. Clin Cancer Res. 2005;11(16):5928–34. Epub 2005/08/24. pmid:16115935.
  50. 50. El-Maraghi RH, Eisenhauer EA. Review of Phase II Trial Designs Used in Studies of Molecular Targeted Agents: Outcomes and Predictors of Success in Phase III. J Clin Oncol. 2008;26(8):1346–54. pmid:18285606.
  51. 51. Passero VA, Branstetter BF, Shuai Y, Heron DE, Gibson MK, Lai SY, et al. Response assessment by combined PET-CT scan versus CT scan alone using RECIST in patients with locally advanced head and neck cancer treated with chemoradiotherapy. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2010;21(11):2278–83. Epub 2010/05/01. pmid:20430907.
  52. 52. Matoba M, Tuji H, Shimode Y, Kondo T, Oota K, Tonami H. Lesion regression rate based on RECIST: prediction of treatment outcome in patients with head and neck cancer treated with chemoradiotherapy compared with FDG PET-CT. Journal of radiation research. 2015;56(3):553–60. Epub 2015/04/02. pmid:25829531; PubMed Central PMCID: PMCPmc4426917.
  53. 53. Patil V, Noronha V, Joshi A, Muddu Krishna V, Juvekar S, Pantvaidya G, et al. Is There a Limitation of RECIST Criteria in Prediction of Pathological Response, in Head and Neck Cancers, to Postinduction Chemotherapy? ISRN oncology. 2013;2013:259154. Epub 2013/10/11. pmid:24109521; PubMed Central PMCID: PMCPmc3786467.
  54. 54. William WN Jr., Pataer A, Kalhor N, Correa AM, Rice DC, Wistuba II, et al. Computed tomography RECIST assessment of histopathologic response and prediction of survival in patients with resectable non-small-cell lung cancer after neoadjuvant chemotherapy. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer. 2013;8(2):222–8. Epub 2013/01/05. pmid:23287849; PubMed Central PMCID: PMCPmc3549050.