Next Article in Journal
Supportive Management of Mucositis and Metabolic Derangements in Head and Neck Cancer Patients
Next Article in Special Issue
The Tip of the Iceberg: Clinical Implications of Genomic Sequencing Projects in Head and Neck Cancer
Previous Article in Journal
Analysis of Pre-Analytic Factors Affecting the Success of Clinical Next-Generation Sequencing of Solid Organ Malignancies
Previous Article in Special Issue
Cytokine and Adipokine Levels in Patients with Premalignant Oral Lesions or in Patients with Oral Cancer Who Did or Did Not Receive 1α,25-Dihydroxyvitamin D3 Treatment upon Cancer Diagnosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Feasibility of Primary Tumor Culture Models and Preclinical Prediction Assays for Head and Neck Cancer: A Narrative Review

by
Amy J. C. Dohmen
1,2,*,
Justin E. Swartz
3,
Michiel W. M. Van Den Brekel
1,
Stefan M. Willems
4,
René Spijker
5,6,
Jacques Neefjes
2 and
Charlotte L. Zuur
1
1
Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
2
Department of Cell Biology, the Netherlands Cancer Institute—Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
3
Department of Otorhinolaryngology—Head and Neck Surgery, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3508 GA, The Netherlands
4
Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3508 GA, The Netherlands
5
Medical library, Academic Medical Center, Amsterdam 1100 DE, The Netherlands
6
Dutch Cochrane Centre, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3508 GA, The Netherlands
*
Author to whom correspondence should be addressed.
Cancers 2015, 7(3), 1716-1742; https://doi.org/10.3390/cancers7030858
Submission received: 2 July 2015 / Revised: 6 August 2015 / Accepted: 20 August 2015 / Published: 28 August 2015
(This article belongs to the Special Issue Head and Neck Cancer)

Abstract

:
Primary human tumor culture models allow for individualized drug sensitivity testing and are therefore a promising technique to achieve personalized treatment for cancer patients. This would especially be of interest for patients with advanced stage head and neck cancer. They are extensively treated with surgery, usually in combination with high-dose cisplatin chemoradiation. However, adding cisplatin to radiotherapy is associated with an increase in severe acute toxicity, while conferring only a minor overall survival benefit. Hence, there is a strong need for a preclinical model to identify patients that will respond to the intended treatment regimen and to test novel drugs. One of such models is the technique of culturing primary human tumor tissue. This review discusses the feasibility and success rate of existing primary head and neck tumor culturing techniques and their corresponding chemo- and radiosensitivity assays. A comprehensive literature search was performed and success factors for culturing in vitro are debated, together with the actual value of these models as preclinical prediction assay for individual patients. With this review, we aim to fill a gap in the understanding of primary culture models from head and neck tumors, with potential importance for other tumor types as well.

1. Introduction

Seventy percent of all patients with head and neck squamous cell carcinoma (HNSCC) present with advanced stage disease and are characterized by an overall 5-year survival rate of approximately 35%–60% in the case of surgical treatment with or without chemotherapy (CT) and radiotherapy (RT) [1,2,3]. From around 1980 onward the addition of high-dose cisplatin to RT (CCRT) has become the routine treatment for locally advanced disease [4]. Nevertheless, a meta-analysis of randomized trials in 2009 indicated that there is only a moderate absolute overall survival benefit of 6.5% at 5 years when adding chemotherapy to loco-regional treatment [5]. A subgroup of head and neck cancer (HNC) patients with HPV-positive oropharynx carcinomas usually shows better prognosis following CCRT [6]. A similar analysis in laryngeal cancer patients also described no survival benefit from the addition of chemotherapy to radiotherapy [7]. Moreover, the addition of high-dose cisplatin to RT is accompanied with a substantial increase in grade three or worse toxicity of 52% to 89% [8]. A more personalized patient selection for this treatment should improve the quality-of-life of the non-responding patient population.
More effective and less toxic targeted therapies have not (yet) penetrated in the treatment of patients with HNC. In recent years, only cetuximab has been registered as a radiosensitizer to improve treatment for advanced HNSCC. Literature, however, shows inconclusive results for survival benefit of this treatment compared to CCRT [9,10]. Unfortunately, this leaves CCRT the mainstream of therapy with rather variable individual clinical outcome. It therefore remains a major challenge in HNSCC to develop novel drugs for improved survival and to reveal patients prior to therapy that will actually benefit from the intended treatment regimen. Consequently, there is a strong need for a preclinical model to identify those tumors of patients that will respond to a particular treatment. One of such models is the technique of culturing primary tumor tissue and testing drugs prior to treatment. In order for a culture model to be feasible as a preclinical treatment prediction tool, it should be a short-term culture technique, resembling the patient’s tumor as closely as possible and it should be low in costs. Xenograft mouse models can be used to assess therapy response as well. However, they are in fact long-term assays in which the patient’s tumor cells adjust to the murine environment, leading to genetic drift of the tumor cells. These models are not optimal, expensive and difficult for multiple drug testing. For these reasons, we excluded xenograft mouse models from our literature search. With this review, we aim to study feasibility and success percentages of previously described fresh primary HNSCC culturing techniques and their preclinical chemo- and radiosensitivity assays.

2. Materials and Methods

A narrative review was performed via a systematic literature search in Pubmed searching for primary HNSCC tumor culturing techniques (research Question 1) and their in vitro sensitivity assays with clinical correlation (research Question 2) (Supplementary Materials). We screened titles and abstracts of the identified literature using preformulated criteria (Figure 1a,b). Thereafter, a full text screen of the selected articles was done. Included were studies that described any technique for culturing fresh primary tumor tissue of HNSCC patients, except for techniques involving only cultures using xenografts models. The search includes papers using cell lines. Only papers considering primary tumor tissue to establish fresh cell lines were included. Studies describing the use of purchased or already established cell lines, while not reporting the technique of its establishment, were excluded. Also included were fresh HNSCC culture studies regarding in vitro versus in vivo chemosensitivity or radiosensitivity assays. Additionally, references of the included studies were screened and added to the literature list when relevant. Final selection was based on consensus of all authors.
Figure 1. Flow diagram of the systematic review process for the (a) search of various culturing techniques used in head and neck cancer; (b) search for chemo- and radiosensitivity assays.
Figure 1. Flow diagram of the systematic review process for the (a) search of various culturing techniques used in head and neck cancer; (b) search for chemo- and radiosensitivity assays.
Cancers 07 00858 g001aCancers 07 00858 g001b

3. Results

An overview of key publications for fresh primary tumor cell culture of HNSCC is presented in Table 1, representing the culturing techniques, and Table 2, showing the clinical correlation.

3.1. HNSCC Cell Lines

The first attempts to establish HNSCC monolayer cell lines were performed in the 1950’s on a variety of tumors (sarcoma, leukemia, Hodgkin, myeloma, kidney, breast, lung, pharynx, larynx, rectum, melanoma and ovary) [11]. From the 1980s onward, several groups, including those of Carey and Grenman [12], Rheinwald and Beckett [13], and Easty [14], were among the earliest to be able to establish HNSCC cell lines, achieving success rates of around 30% [12]. Nowadays, a myriad of HNSCC cell lines are available for in vitro experiments, as summarized by Carey in 1994 [12], Sacks in 1996 [15] and Lin in 2007 [16]. Also, tumor cell lines from particular patient cases are available, such as very young patients or patients with Fanconi anemia-associated HNSCC [17,18]. It is not exactly known why certain tumors may be cultured indefinitely, while others cannot, although culture success rates have improved by taking biopsies under aseptic conditions from non-necrotic and uninfected tumor areas.
In all studies, HNSCC cell lines were established through the “explant technique”, described extensively in 1994 by Carey [12]. In this technique, fresh tumor specimens were mechanically minced into fragments. Samples may be further dispersed enzymatically using trypsin, DNase, collagenase or a combination thereof [19]. The cell suspension was then placed into a rich culture medium, such as DMEM or RPMI-1640 with additional fetal bovine serum (FBS) and transferred to petri-dishes or culture flasks [20,21,22]. A combination of antibiotics and antimycotics was added to prevent bacterial or fungal overgrowth, and fibroblast overgrowth was managed through selective trypsinisation or cell scraping [12]. The cells were then cultured at 37 °C in an air mixture with 5% CO2. When cells grew to confluency, they were passaged. According to Carey, a cell line may be considered established after the 20th passage (e.g., several months, depending on growth rate), as about 15% of tumor cells initially show growth but then stop growing or die. Success percentages of 11%–33% have been described for establishment of cell lines from HNSCC in this fashion [13,23,24]. Recently, Owen et al. described higher success rates of 50%. They used fluorescence associated cell sorting to separate fibroblasts from tumor cells. This appeared to be a promising technique to reduce fibroblast overgrowth and to improve the success rate of cell line establishment [25].
Regarding in vitro to in vivo correlation, no significant difference was found between radiosensitivity of HNSCC cell lines established from 7 patients with recurrent disease after RT, and cell lines derived from 13 patients without prior irradiation. Moreover, two patients with unfavorable clinical response to RT, provided cell lines with good RT response in vitro [26]. However, these preclinical assays did not consider fresh tumor specimens but cell regrowth from previously established cell lines up to 22 passages, conditions that may have selected cells with reasonable radiosensitivity.
Unfortunately, while assays using HNSCC cell lines have been proven essential for experiments concerning molecular biology, they seem not useful as a preclinical prediction model for the individual cancer patient. It is critical to establish cells in culture that best resemble the patient’s tumor. This implies that the selection on the fast growing stable cells, surviving under culture conditions, should be prevented. Short tissue cultures, where various cells are still present and not out-selected, would be critical to arrive at patient-relevant culture conditions for the testing of various treatment conditions.

3.2. Single Cell Cultures

One essential way of culturing is by starting off with single cell suspensions from tumor biopsies. This is usually done by mechanical and enzymatic digestion. The first part of Table 1 describes studies using this technique.

3.2.1. The Cell-Adhesive Matrix (CAM) Assay

The cell adhesive matrix (CAM) assay is a monolayer culture system developed by Baker et al., that uses a fibronectin and fibrinopeptides coated dish for optimized cell adhesion [27]. Cell growth was stimulated through hormone- and growth factor-supplemented medium. Fresh primary tumor biopsies (melanoma, sarcoma, lung, colon, ovarian and renal) were mechanically and enzymatically digested and plated as single cells. After 24 h of incubation, RT or drugs were administered. After 2 weeks the cultures were fixed for quantification of cell growth and survival. Baker et al. successfully cultured 75%–90% of tumors using this technique. The articles reviewed, reaches culture success rates of 60%, within 14–21 days of culturing, in studies with a large number of patients (Table 1) [27].
The CAM assay in HNSCC has only been used to assess radiosensitivity. Brock et al. first reported on radiosensitivity using the CAM-assay in 1990, in which 72 of 121 HNSCC patients were evaluable (60% success rate) (Table 2) [28,29]. Radiosensitivity was determined by comparing the cell-covered surface to the total surface of 24-well plates after radiation with 2 Gray (surviving fraction at 2 Gray, SF2). The SF2 was 0.40 in 12 patients with recurrent disease and 0.30 in 60 patients with local tumor control (p > 0.05).
Table 1. Overview of the various culturing techniques from HNSCC tissue.
Table 1. Overview of the various culturing techniques from HNSCC tissue.
Authors, yearAssayRead-outResultCorrected for stromaDaysPatientSuccess
(n)(%)
Single cell cultures after enzyme digest
Brock 1990CAM monolayerSF2SF2 0.33 (0.11–0.91)No1412160
Girinsky 1993CAM monolayerSF2, alphaSF2 0.39 (0.37–0.42), alpha 0.18 (0.13–0.24)No14–219660
Girinsky 1994CAM monolayerSF2, alphaSF2 0.39 (0.37–0.41), alpha 0.19 (0.14–0.25)No14–2115660
Eschwege 1997CAM monolayerSF2SF2 0.41 (0.21–0.88)--92-
Mattox 1980aSoft-agar clonogenicCE (>20 cells), 3-ThCE 0.006 (0.001–0.08)No10–143664
Mattox 1980bSoft-agar clonogenicCE (>20 cells)CE 0.001–0.19-14–217345
Johns 1982Soft-agar clonogenicCE (≥30 cells, ≥5 colonies)CE 0.005No7–147349
Mattox 1984Soft-agar clonogenicCE (≥6 colonies)CE -No14–2115836
Cobleigh 1984Soft-agar clonogenicCE (≥30 cells, >50 µm, >5 colonies)Growth observationNo7–14510
Schiff 1984Soft-agar and agaroseCE (>20 cells)CE 0.002–0.08No7–211956/90 **
Rofstad 1987Soft-agar clonogenicPE (>50 cells), SF2SF2 ±0.18–0.45. PE 0.6–2.2No28–35433
Stausbøl-Grøn 1995Soft-agar clonogenicPE (>50 cells, >60 µm), SF2SF2 0.36 (0.19–0.88). PE 0.02–0.75Yes2815-
Stausbøl-Grøn 1999aSoft-agar clonogenicPE (>60 µm), SF2SF2 0.50 (0.11–1.00). PE 0.052 (0.005–1.60)Yes2810570
Stausbøl-Grøn 1999bSoft-agar clonogenicPE (>50 cells/>60 µm), SF2SF2 0.50 (0.19–1.00). PE 0.043 (0.005–1.03)Yes2810568
Björk-Eriksson 1998Soft-agar clonogenicCE (>50 cells/>60 µm), SF2SF2 0.48 (0.10–1.00). CE 0.093 (0.002–1.30)Yes2814074
Björk-Eriksson 2000Soft-agar clonogenicCE (>50 cells), SF2SF2 0.40 (0.10–1.00). CE -Yes2815670
Dollner 2004aColony forming (flavin free)CE (> 16 cells); C100Yes41392
Dollner 2004bColony forming (flavin free)CE (> 16 cells); C100Yes41989
Dollner 2006aColony forming (flavin free)CE (> 16 cells); C100Yes413-
Dollner 2006bColony forming (flavin free)CE (> 16 cells); C100Yes412-
Histocultures
Robbins 1994HDRA3-ThSensitivity: ≥84% IRYes3–152688
Singh 2002HDRAMTT, DNASensitivity: >30% IRNo24298
Ariyoshi 2003HDRAMTTSensitivity: >40%–60% IR, depending on drugNo719100
Hasegawa 2007HDRAMTTSensitivity: >40%–60% IRNo749-
Pathak 2007HDRAMTTSensitivity: >50% IRNo85791
Gerlach 2014Slice culture on membraneIHCCytotoxic effectNo5 h–7 days12-
Heimdal 2000aFragment spheroidsIHCViability, cytokineNo10–281890
Kross 2005Fragment spheroidsELISA, IHCIL-6, MCP-1, TNF-α *Yes/No>731-
Kross 2008Fragment spheroidsELISAIL-6, MCP-1 *Yes/No10–2865-
Lim 2011SquamospheresTumor differentiation, stemcell traitsPCR, IHC, FACS, xenograftNo>14476
Lim 2012SquamospheresTumor differentiation, stemcell traitsPCR, IHC, FACS, western blot, xenograftNo>14--
CAM = cell adhesive matrix; ELISA = enzyme-linked immuno sorbent assay; HDRA = histoculture drug response assay; IR = inhibition rate; SF2 = surviving fraction at 2 Gray; PCR = polymerase chain reaction; CE or PE = cloning or plating efficiency; FACS = fluorescence-activated cell sorting; C100 = complete suppression of colony formation; * cytokines and chemokine; MTT = yellow tetrazole is reduced to purple formazan in living cells; ** 56% soft-agar, 90% agarose; IHC = immunohistochemistry.
Table 2. Overview of the various assays and their chemo- and radiosensitivity correlations.
Table 2. Overview of the various assays and their chemo- and radiosensitivity correlations.
Authors, yearAssayIn vitro treatmentIn vivo treatmentRead-outCorrelationOutcome correlationFU (months)
Single cell cultures after enzyme digest
Brock 1990CAM monolayerRTPost-op RTSF2YesLocal control. SF2: recurrent 0.40 (n = 12), not yet recurred 0.30 (n = 60). Not significant.24
Girinsky 1993CAM monolayerRT70% RT, 30% post-op RTSF2, alphaYesLocal control: alpha value. Not for survival15 (1–29)
Girinsky 1994CAM monolayerRT62% RT, 38% post-op RTSF2, alphaYesLocal control: alpha value24 (9–47)
Eschwege 1997CAM monolayerRTRTSF2NoLocal control, survival68 (45–80)
Mattox 1980aSoft-agar clonogenicCT-CE, 3-Th---
Mattox 1980bSoft-agar clonogenicCT-CEYesEarly mortality: CE > 0.02%-
Johns 1982Soft-agar clonogenicCT-CEYesStage, N-class and survival: high CE (n = 29)-
Mattox 1984Soft-agar clonogenicCT-CENoNo correlation positive culture with stage, N-class, recurrence. No difference in survival for high (>0.02%) and low (<0.02%) CE 24
Cobleigh 1984Soft-agar clonogenic--CE---
Schiff 1984Soft-agar and agarose--CE---
Rofstad 1987Soft-agar clonogenicRT-PE, SF2---
Stausbøl-Grøn 1995Soft-agar clonogenicRTRTPE, SF2---
Stausbøl-Grøn 1999aSoft-agar clonogenicRT-PE, SF2NoOverall/tumor SF2 were not correlated with T/N and stage-
Stausbøl-Grøn 1999bSoft-agar clonogenicRTRTPE, SF2NoOverall/tumor SF2 and PE did not predict local-regional control (n = 38)42 (16–70)
Björk-Eriksson 1998Soft-agar clonogenicRT-CE, SF2NoSF2 did not correlate with tumor grade, T/N class
Björk-Eriksson 2000Soft-agar clonogenicRTRT/CT/SurgeryCE, SF2YesTumor SF2 (0.40) prognostic for local control, not for overall survival. SF2: recurrent 0.53 (n = 14), not yet recurrent 0.38 (n = 70)25 (7–65)
Dollner 2004aColony forming (flavin free)CT-CE, C100---
Dollner 2004bColony forming (flavin free)CT-CE, C100---
Dollner 2006aColony forming (flavin free)CT-CE, C100---
Dollner 2006bColony forming (flavin free)CT-CE, C100---
Histocultures
Robbins 1994HDRACTCT3-ThYesClinical response. PPV 83%, NPV 64%. Sensitivity 71%, specificity 78%-
Singh 2002HDRACTSurgery/(C)RT/CTMTT, DNAYesClinical response. Chemosensitivity is a significant prognostic variable for 2 year cause specific survival30
Ariyoshi 2003HDRACTCT and CRTMTTYesClinical response. CRT: PPV 87%, NPV 50%. Sensitivity 87%, specificity 50% (patients received RT, in vitro no RT)-
""CT: PPV 90%, NPV 100%. Sensitivity 100%, specificity 67%
Hasegawa 2007HDRACTCT then surgeryMTTYesClinical response. PPV 77%, NPV 80%. Sensitivity 91%, specificity 57%.> 4 weeks
""Significant correlation between cisplatin sensitivity in vitro (50% cut-off) and clinical response. No correlation for 5-FU.
Pathak 2007HDRACTSurgery/(C)RT/CTMTTYesClinical response. PPV 69%, NPV 80%. Sensitivity 79%, specificity 71%.2 weeks
""Significant correlation between in vitro chemosensitivity and clinical response
Gerlach 2014Slice culture on membraneCT-IHC---
Heimdal 2000aFragment spheroids--IHC---
Kross 2005Fragment spheroids--ELISA, IHC---
Kross 2008Fragment spheroids--ELISAYesIncreased IL-6 levels predict recurrence and survival30
Lim 2011SquamospheresCT-Differentiation---
Lim 2012SquamospheresCT-Differentiation---
CAM = cell adhesive matrix; C100 = complete suppression of colony formation; HDRA = histoculture drug response assay; MTT = a yellow tetrazole, is reduced to purple formazan in living cells; RT = radiotherapy/irradiation; ELISA = enzyme-linked immuno sorbent assay; CT = chemotherapy; IHC = immunohistochemistry; SF2 = surviving fraction at 2 Gray; PPV = positive predictive value; CE or PE = cloning or plating efficiency; NPV = negative predictive value.
In 1994, Girinsky et al. described the CAM assay in 156 HNSCC biopsies. SF2 data were available for 76 HNSCC patients [30,31]. SF2 values were not predictive for long-term local control (cut-off 0.50; 66% versus 63%). On the other hand, a significantly higher local control rate (p = 0.04) was obtained for patients with higher alpha values (which illustrates the rate of cell kill by a single dose of RT; cut-off 0.07 Gy−1; 69% versus 38% at 2 years).
The third group to work with the CAM assay was Eschwege et al. [32]. They studied 92 HNSCC patients with mainly oropharyngeal carcinomas treated with RT and found both SF2 and alpha value not to be prognostic factors for local control and overall survival.

3.2.2. Soft-Agar Clonogenic Assays

Clonogenic assays, in which single tumor cells were cultured on agar-coated plates, were first described by Puck and Marcus on HeLa cervical tumors [33,34]. In 1977, Salmon and Hamburger utilized an adaptation of this technique as an in vitro clonogenic assay of anticancer drugs on tumor cells (myeloma, lymphoma, leukemia, lung, ovary, melanoma and neuroblastoma) [35]. Later, it was used for human pancreatic and colon tumor cells grown in immune-suppressed mice, popularized by Courtenay and Mills and referred to as the Courtenay-Mills clonogenic assay [36]. The main feature of this agar method is its selection for stem cells or transformed cells [37,38]. Although agar cultures also support benign tumors and anchorage-dependent cells, if supplemented with high serum levels or transforming growth factors, soft-agar is still a broadly accepted method for tumor cell selection based on their anchorage-independent growth behavior.
The successful use of the Courtenay-Mills soft-agar clonogenic assay with biopsies of HNSCC was first described by Mattox and Von Hoff [39,40,41], Johns [37] and Schiff [42] (Table 1). Primary HNSCC samples were washed, minced with scalpels and further disaggregated, as in the “explant” technique described by Carey [12]. The cell suspensions were placed in culture plates covered with a feeding layer containing agar, culture medium, FBS and a variety of other nutrients, and then incubated with a chemotherapeutic drug for one hour [39]. After that, the cells were washed, plated and incubated, along with untreated controls. After 7–21 days, the cultures can be evaluated for colony formation (clumps of more than 20–40 cells). Plating efficiency (number of colonies compared to number of plated cells) was generally low, around 0.005, meaning only 1 in 200 cells will grow out as a colony. Cultures were regarded successful if six or more colonies form in untreated control plates [37,39,40,41]. The survival fraction was calculated from the amount of colonies formed in treated, compared to untreated plates. Survival rates of 30% or less, compared to untreated controls, were considered an in vitro indicator of chemosensitivity [37,39,40,41]. We reviewed several studies using soft-agar clonogenic assays, showing overall success rates of 50% (0%–74%), where colonies of 20 to 50 cells form within a time span of 1–5 weeks [37,39,40,41,42,43,44,45,46,47,48,49]. These studies were done on a reasonably amount of patients (Table 1). More poorly differentiated tumors had higher overall culture success rates than well-differentiated tumors [41,42].
These authors also did in vivo correlations with this assay (Table 2). However, chemosensitivity testing was often not possible due to low tumor cell count. Mattox [40] and Johns [37] showed that a higher cloning efficiency (>0.02% and >0.05%) was associated with a higher likelihood of recurrence [37] and early mortality [37,40]. However, a follow-up study of 158 attempted fresh HNSCC cultures did not confirm this correlation [39]. Cobleigh attempted a soft-agar assay on HNSCC in 1984 as well, with no success [43]. Finally, Schiff tried to culture tumors from 19 HNSCC patients [42]. Samples from nine patients were cultured in agar and 10 in agarose. Culture success was higher in agarose-cultured samples (56% versus 90%).
With respect to radiosensitivity correlations done with this clonogenic assay, Rofstad, in 1987, studied various tumors (including four head and neck tumors) with a 33% culture success rate [44]. The SF2 differed considerably among individual tumors of the same histological type. In 1995, Stausbøl-Grøn cultured biopsies of 15 HNSCC patients prior to radiotherapy [45]. In 12 tumor biopsies 2%–33% of the colonies were tumor and 83%–100% of the colonies were fibroblasts. The overall SF2 correlated significantly to the fibroblast SF2 but not to tumor cell SF2. In 1999, the same group assessed radiosensitivity in 105 HNSCC patients. Culture was successful in 70%. Data were described from 38 patients who were treated with radiotherapy [46,47]. The majority of the colonies obtained from the biopsies were again fibroblast-marker positive. No significant correlations were found between overall or tumor SF2 and T/N-class and disease stage. Neither tumor cell SF2, overall SF2, nor plating efficiency predicted the locoregional tumor control probability.
Björk-Eriksson determined the intrinsic radiosensitivity of primary HNSCC on data collected over 5 years for 140 patients using a soft-agar clonogenic assay [48]. Care was taken to ensure that only colonies from malignant cells were scored by morphology and staining. Colonies with a radius of more than 60 µm (>50 cells) after 4 weeks of culture were quantified. They reached a culture success rate of 74% (104/140) with a colony-forming efficiency (CFE) of 0.093 and obtained SF2 data from 63% of the patients with a mean of 0.48 (0.10–1.00). Interestingly, these authors observed that approximately 0%–10% of cultured colonies were of a non-malignant cell type. In 2000, the same group reported on 156 previously untreated HNC patients (70% culture success rate, 110/156) and evaluated in 54% (84/156) of the patients the prognostic value of SF2 prospectively [49]. Eighty-four patients were mainly treated with neoadjuvant chemotherapy plus radiotherapy, with or without final surgery. For prognostic analyses, patients were divided in radioresistant (SF2 > 0.40) and radiosensitive (SF2 < 0.40) tumors. After multivariate analysis, tumor SF2 was found an independent prognostic factor for local control (p = 0.036), but not for overall survival (p = 0.20).
Dollner used a colony forming assay without soft-agar, in a 96-well plate format. In 2000, they used monochromatic light sources to avoid flavin-mediated photo-oxidative effects (termed “Flavino-assay”) especially during chemosensitivity testing. Fresh tumor biopsies were digested and after 3 days of exposure to various drugs adherent colonies were fixed and counted to determine the IC-50 [50,51]. The overall chemoresponse was dominated by stromal cell multidrug resistance [52,53,54]. Stromal cells were resistant to drug combinations in 98% of the experiments, whereas epithelial colonies were sensitive to cisplatin/5-FU in 16%, to carboplatin/5-FU in 8.3%, to cisplatin/docetaxel in 33% and to carboplatin/docetaxel in 8.3%. In 2010, the assay was correlated to clinical outcome in 18 cultures receiving neoadjuvant TPF (docetaxel, cisplatin and 5-FU) prior to RT [55]. Twelve tumors could be successfully cultured (66.7%) The in vivo tumor response to induction chemotherapy was correctly predicted by the tumor culture assay in 10 patients (83.3%). However, an in vitro prediction of clinical tumor response to the complete treatment regimen was disregarded [55]. These data were only published in a meeting abstract; the full article was not published and thus not fully evaluable. In recent years, this group has used this assay to test chemotherapy response to several drugs in vitro [56]. However, no reports have been published reporting a proper correlation between predicted outcome based on the Flavino assay and the actual patient outcome in the clinic.

3.3. Histocultures

Another way of culturing is to leave tumor tissue intact by using only mechanical mincing. This maintains the normal and (largely) unaffected tumor-tumor environment interactions as occurring in vivo. The second part of Table 1 depicts studies using this technique.

3.3.1. The Histoculture Drug Response Assay (HDRA)

In an effort to preserve the 3-dimensional (3D) histological structure of the tumor, a method was developed to culture (mouse) breast tumor fragments without further dispersal [57], thereby maintaining cell heterogeneity and cell-cell interactions [58]. These models became the cornerstone of the “histoculture drug response assay” (HDRA), further developed by the group of Hoffman for gastric and colorectal cancers [59,60].
Primary tumor material was minced into fragments of about 0.5 mm diameter and placed on 1 × 1 cm collagen sponge gels in a 24-well plate. One mL of RPMI-1640 medium supplemented with FBS was added and the plate was incubated. RPMI medium was selected rather than (D) MEM, for better preservation of phenotypic heterogeneity [61]. For chemosensitivity assessment, drugs were added to the culture medium and cultured for 7 days. Viability was determined using the MTT assay that measures metabolic activity by a spectrophotometer. When the inhibition rate (absorbance in treated, compared to untreated samples) was 50% or more, tumors were regarded as chemosensitive [62].
Robbins and colleagues were the first to describe the HDRA in HNSCC [63]. They investigated inhibition of tumor proliferation by cisplatin using radioactive 3H-thymidine incorporation in tumor cells as an endpoint. In a group of 26 patients with HNC (21 SCC, five with other histological types), 23 (88%) specimens were evaluable. The authors described a positive predictive value (PPV) of 83% and a negative predictive value (NPV) of 64% for partial or complete clinical response in patients treated with cisplatin chemoradiation. Singh observed a correlation between in vitro chemosensitivity and 2-year cause-specific survival, for cisplatin, 5-FU and both agents [64]. However, the 41 patients included endured various treatment modalities including chemotherapy, surgery and RT. Therefore, no conclusion could be drawn considering chemosensitivity in this study. Another study concerning patients treated for oral cavity SCC showed a PPV of 87% and a NPV of 50% for sensitivity testing with 5-FU, cisplatin, adriamycin, bleomycin and docetaxel [65]. In 2007, Hasegawa et al. assessed both primary tumors and lymph node metastases and found a significant correlation between in vitro cisplatin sensitivity and clinical response. There was no correlation for 5-FU [62]. Pathak studied a rather homogenous group of oral cavity SCC patients receiving chemotherapy regimens resulting in comparable predictive values [66].
The efficacy and utility of the HDRA as a useful predictor for chemotherapy response in patients is described in a number of studies of various human solid tumors, including gastric and esophageal cancer [67,68], colorectal cancer [59] and ovarian cancer [69,70,71].
Recently, Gerlach and colleagues described an adaptation of the HDRA [72]. In this assay, HNSCC fragments of 12 tumors were sliced with a vibratome or tissue chopper and were placed on membranes, rather than a collagen sponge. Tumor slices were incubated with docetaxel, cisplatin, or no drugs and cultured for 5 h to 7 days in a flavin-free culture medium. The slices were then fixed, embedded in paraffin and examined using Ki-67 (a proliferation marker), caspase-3 staining (an apoptosis marker) and γH2AX (a marker for double-strand DNA breaks). After 7 days of culture, tissue quality was decreased in some tumor slices. Increased apoptosis was observed in the slices exposed to drug, compared to controls. In their publication on this culture method, no correlations to clinical outcomes were done [72]. Recently, more groups have started to generate histocultures of HNSCC to investigate the effect of existing or novel, more targeted drug-based, therapies, such as the PI3K inhibitor LY294002, to investigate the effect of molecular signaling in tumor growth [73,74].

3.3.2. Spheroids, Squamospheres and Organoids

The spheroid culture technique was developed as well to maintain tumor tissue heterogeneity and a 3D architecture (Table 1) [75]. Spheroids would ideally resemble the growth pattern of solid tumors in vivo as they are composed of an outer layer of proliferating cells closest to a nutrient and oxygen supply (capillaries) with inner layers of quiescent and -most central- necrotic cells. This was tested with a variety of cell lines [76,77]. Technically, they can either be grown from cells obtained from monolayer cell cultures after trypsinisation or grown from fresh tumor biopsy fragments [16].
In 2000, Heimdal described malignant and benign “fragment spheroids” in a nonadhesive system [78]. HNSCC fragments were cultured on agar-coated culture flasks and after 10–14 days rounded spheroid-like structures were selected for a 2-week co-culture with autologous monocytes derived from peripheral blood samples of the patients. Cytokine IL-6 production of the monocytes was significantly higher in case of direct cell-cell (i.e., tumor-monocyte) contact compared to co-cultures where tumor cells and monocytes were separated by a semi-permeable membrane. In 2005, Kross used the same model to study the cytokine secretion, and to describe the number of epithelial cells (cytokeratin positive), fibroblasts (vimentin-positive) and macrophages (CD68 positive) in both malignant HNSCC and benign spheroids [79]. In malignant spheroids, the proportion of epithelial cells during spheroid formation decreases from 28% to 13%. The density of macrophages (2%) and fibroblasts (13%) did not change. Monocytes secreted more IL-6 when co-cultured with malignant compared to benign spheroids. In 2008 they found increased IL-6 cytokine production in vitro to be predictive for recurrence and survival (Table 2) [80].
A few years later, Lim et al. described “squamospheres” resulting from culturing mechanically and enzymatically digested biopsies from 47 HNSCC patients [81]. Single cells were incubated for 2–4 weeks to assess sphere forming ability (self-renewal) and other cancer stem cell hallmarks like tumor-initiating capabilities and chemoresistance. A distinction was made between undifferentiated squamospheres (cultured in stem cell medium: serum-free, with N2, B27, EGF and bFGF) and differentiated squamospheres (medium with 10% FBS, without EGF and bFGF). Overall, the success rate of spheroid formation was 6%. Single cells from spheres were assessed for anchorage-independent growth ability as an indicator for cell transformation in vitro; undifferentiated cells that maintained sphere forming capability sustained and differentiated cells diminished in agar. In agreement, tumor formation in nude mice was significantly better for undifferentiated cells. This was later confirmed by Pozzi et al., who found better tumorigenicity in sphere forming cancer stem cell (CSC)-enriched cell populations than in unselected tumor cells [82]. To investigate whether HNSCC CSCs can be expanded in adherent cultures without loss of stem cell properties, Lim et al. tested different plate coatings [83]. HNSCC-CSCs grew much faster on type IV collagen-coated plates than in suspension. Adherent CSCs expressed stem cell markers, were chemoresistant, produced tumors in mice and showed less spontaneous apoptotic cell death.
Leong et al. described the establishment of three cell lines from primary HNSCC grown as spheroids or monolayers. They confirmed the improved chemoresistance of spheroids when treated with 5FU, cisplatin, etoposide or irradiation [84]. Unfortunately, correlation of the ex vivo results with the actual clinical outcome was not one of the aims of this study. On the other hand, while sphere formation or sphere formation capability of CSCs, may increase resistance to some drugs, another group has shown in primary HNSCC spheroid cultures that it is also possible to target these CSCs in particular [85].
Until now, only one group described an “organoid culture assay” of HNSCC [86,87]. Although the authors did not use fresh primary tumor, they aimed for 3D in vitro tumor growth allowing to form organized and differentiated structures such as those existing in the organism. After full digestion of a xenografted HNSCC in mice, single cell suspension droplets were seeded on a bridge-like filter in a petri-dish. In this model the tissue grows at the air-medium interface, as medium was added just until the bridge. After 4 weeks solid culture nodules were disaggregated again to assess viability of cells by Trypan Blue. Pathologic evaluation of the nodules showed histological characteristics similar to the original human hypopharyngeal carcinoma up to 3 weeks of culturing. After 3 weeks degeneration was seen.

3.4. Other Assays

Various other techniques to establish in vitro cultures of primary HNSCC were reported, but were only described by a single group and not further popularized. For completeness, these assays are briefly described below.

3.4.1. Flow-Cytometric Analysis

In 1989, Garozzo presented a different model for short-term culturing of HNSCC in which he acknowledged an equal contribution of all cell populations in the progression of neoplastic disease, and referred to Von Hoff stating that HNC are not very likely to grow on agar [88,89]. Surgical HNSCC specimens were disaggregated into cell suspensions and exposed to various drugs for 24 h. The major endpoint was the presence of cell cycle blocks, determined by flow cytometry. Patients were treated with a standardized, undisclosed, regimen of polychemotherapy. Thirteen of the 15 patients showed complete or partial remission. The assay predicted sensitivity to several of the drugs in 11 of these 13 patients (PPV 85%).

3.4.2. Tumor Slices Grown in Test-Tubes

Elprana et al. described a culture system where human HNSCC fragments, from one patient, floated freely in test tubes containing medium with or without drugs [90,91]. In vitro, the tumor was sensitive to cisplatin and 5-fluorouracil. The patient received a combination of these drugs and experienced complete regression in four months, although long-term outcome was not described.

3.4.3. Microdevices

Recently, the group of Greenman cultured HNSCC samples in vitro with a microfluidic device. Medium flowed through the device and was collected after drug or irradiation treatment [92,93,94]. Response to chemotherapy or irradiation is determined by measuring LDH in the effluent. Drug treated samples showed significantly more LDH release than the control groups. No further reports were found that correlated the in vitro response to clinical data. This culture technique has been reviewed by Sivagnanam [95].

3.4.4. Micronucleus Assay

Champion et al. described an assay that involved establishing a monolayer culture of primary HNSCC tumors and immunohistochemical staining of these cultures after irradiation to identify micronuclei [96]. These micronuclei may be visible in dividing cells and are considered as DNA fragments that cannot be incorporated in daughter cells, due to (radiation) damage. The primary endpoint was the correlation between micronuclei formation and the amount of radiation exposure. After optimizing the assay in cell lines, primary HNSCC specimens were tested. Unfortunately no correlation between assay outcome and clinical outcome could be established.

4. Discussion

With this review we aimed to evaluate the most successful in vitro culture technique for HNSCC and to discover which model has the best correlation with clinical response. As the chemotherapeutic repertoire increases, a simple and reliable assay to determine the expected patient response becomes critical in making a correct individualized treatment decision.
Monolayer cell line culture is not a proficient method for the use of a preclinical prediction assay. Reasons for this are the long duration of cell line establishment, low culture success rates [12,15] and senescence, the state in which cells no longer divide [13]. Cell line formation is also accompanied with genetic changes like upregulation of oncogenes, and consequently worse clinical outcome [97,98]. Probably for all these reasons, a good clinical correlation was never shown [26].
Short-term fresh tumor cultures, however, do not experience clonal evolution of tumor cell (sub)populations [99]. Worsham, Ragin and Bjerkvig found genetic and molecular cytogenetic resemblance between HNSCC cultures and the primary tumor in vivo [100,101,102]. The short duration of culture increases the evaluability of these assays, as these are not influenced by senescence [103,104,105].
Although tumor biopsies are fully digested in the short-term CAM assay, the assay is thought to allow for restored cell-cell contact within the anchored monolayer. It was probably thought that this anchorage was required to establish the predictive value of SF2 for clinical control, however SF2 was not significantly related to outcome in these studies [28,30,31,32]. Only the alpha value (initial slope of radiation curve) had a good clinical correlation with local tumor control in two studies [30,31]. Heppner and colleagues argued that tumor sensitivity to therapeutic agents in a clonal monolayer culture differ to that of in vivo-like tissue architectures comprised of heterogeneous cells [106].
Another short-term assay is the soft-agar assay. Von Hoff did a meta-analysis on 54 trials in 1990, using a clonogenic assay, which compared in vitro results to clinical outcome in 2300 cases of solid tumors, including a relatively small number of HNSCCs [107]. Overall, they found a 69% true positive rate and a favorable true negative rate of 91%, with a sensitivity and specificity of 79% and 86% respectively, in predicting outcome. We reviewed several studies using soft-agar showing that plating efficiency of HNSCCs is relatively poor. An explanation may be a rather low subpopulation of stem cells in HNSCC. Moreover, solid HNSCC in these studies were fully digested, likely leading to mechanical trauma to the cell. Some authors propose that enzymatic digestion is preferable to maintain viability and growth potential [108]. In addition, the disruption of intercellular attachments may not only irreversibly damage tumor specimens, but may also lead to higher chemosensitivity of cells, not representing the actual in vivo sensitivity [109,110,111,112,113]. For example, this is seen in experiments on mouse mammary tumor cell lines; Miller found that chemoresistance to melphalan and 5-fluorouracil was up to a 1000-fold higher in 3D collagen gel structures than in monolayer cell lines [109,110,111]. Unfortunately, research concerning clonogenic assays also failed to systematically show predictive value for individual clinical outcome, probably due to disruption of the tissue. Namely, four studies investigating clinical correlations involving soft-agar HNSCC colony forming assay, did not find any correlation between in vitro and in vivo response (Table 2) [39,45,46,47]. In two chemosensitivity studies, plating efficiency was associated with tumor stage, N-class and survival [37] and early mortality [40], however not with therapy response. These studies, nevertheless, describe a low number of tumor cells available. Björk performed a radiosensitivity colony forming assay where SF2 was a significant prognostic factor for local control, but not for overall survival [49].
The use of soft-agar should have the advantage of providing support for solid tumor cells, which frequently have difficulties in attaching to the surface of culture dishes. Tumor cells then grow as spherical colonies in agar, while the growth of benign cells such as fibroblast, that require anchorage to a solid substrate, is thought to be reduced [114]. Several groups investigated the impact of stromal cell contamination on culture and treatment sensitivity and concluded that most colonies consisted of fibroblasts. The SF2 is then mainly determined by fibroblast SF2 instead of overall or tumor SF2, and therefore this may contribute as well in not mimicking the correct response in vivo [45,48,49,50,51,54,55].
Overall, the number of weeks to culture and the low percentage of evaluable results make the soft-agar clonogenic assay less suitable for use in individual clinical decision making in HNSCC.
In 1994, while other research groups were exploring cultures of fully digested tumor specimens (CAM- and soft-agar assays), Robbins et al., adopted the HDRA mode. This short-term, sponge-supported histoculture of HNSCC tissue fragments does not require enzymatic digest, leaving cell-cell adhesions, 3D character, as well as the tumor heterogeneity intact [60,63]. All cells, benign and malignant, are co-cultured together. This method allows for the formation of cell aggregates with identifiable and distinctive tissue patterns simulating the in vivo tumor [57]. This probably explains that, for the first time, very high culture success rates were reached (88%–100%). The hypoxic tumor interior, its low pH and relative inaccessibility to chemotherapeutic agents may be the reason for the high predictive values described for in vivo correlations, compared to clonogenic or monolayer assays. In addition, the HDRA needs short-term culturing and will therefore have few genetic alterations when compared to longer-term cultures. Finally, the tumor microenvironment in the HDRA may be of importance for a proper clinical correlation, as the presence of tumor-infiltrating lymphocytes seems to determine clinical outcome in patients with HNSCC [115]. Indeed the HDRA has been confirmed to be a well feasible culture system for fresh HNSCC tissue, as shown by several other research groups with a good correlation to clinical response [62,63,64,65,66]. Only Singh correlated it with clinical outcome, and found that in vitro chemosensitivity was a significant prognostic variable for survival [64]. However, these studies tested only chemosensitivity in vitro, while patients received chemotherapy often combined with radiotherapy. To improve predictive values and to optimize the clinical relevance for predicting the long-term clinical outcome of HNSCC patients, the HDRA model may be tested not only to determine chemosensitivity, but also radiosensitivity. Furthermore, the clinical follow-up duration or the moment of endpoint determination in the identified studies was not always described or it was short (2 to 4 weeks) [62,66]. This might give an overestimate of the chemosensitivity.
Since 2000, several research groups have focused on growing HNSCC “spheres”, “squamospheres” and “organoids” [79,80,81]. As the term suggests, the investigators aimed to establish a 3D arrangement of tumor cells, forming a sphere or organoid, mimicking solid tumor growth in vivo. Therefore, the in vitro “3D model” might better mimic drug response in vivo. Heimdal and Kross showed the potential importance of immune cells in culture prediction assays using this model. Increased cytokine production in co-culture was significantly higher in direct cell-cell contact between autologous monocytes and tumor [116], and was found to be predictive for a clinical unfavorable prognosis in HNSCC [80]. However, overall, the reviewed studies did not succeed to systematically generate the intended organoid like structures. In addition, growing spheres and organoids as described here is relatively time-consuming. After 2 weeks of spheroid formation, a prediction assay warrants another 2 weeks of incubation. Within the first weeks a decreasing proportion of epithelial cells was seen [80] and a degeneration of histological characteristics [86].

5. Conclusions

Within the treatment of HNSCC, there is a strong need for predicting individual clinical outcome prior to therapy, as the overall patient survival rates are relatively low and reliable biomarkers are not available. Moreover, there is a need to test novel drugs before introduction into clinical practice. A preclinical model that closely resembles the in vivo situation would be highly valuable. In this review, we observed that the most successful cultures rates and best correlations to clinical response were reported with the HDRA technique. The HDRA assay has the benefit of better representing the tumor and its microenvironment as it does not involve tissue disaggregation, thus maintaining cell heterogeneity, cell-cell interactions and tissue architecture. However, the correlation to clinical outcome of the HDRA technique has been reported only on a small group of patients and should be validated in larger patient cohorts. Within the HDRA technique it is important to correct for stroma cell response. Another outstanding and obvious point is that the clinical treatment should be resembled in vitro as closely as possible, including irradiation. This will ultimately determine the success of this culture-based assay for personalized treatment decisions. As it stands, the HDRA technique appears to be the best model to test and identify novel treatment modalities for HNSCC, which is currently specified by a very poor prognosis.

Supplementary Files

Supplementary File 1

Acknowledgements

This work was kindly supported by the Riki Stichting.

Author Contributions

Amy Dohmen: Principal investigator. Acquired the data, analyzed and interpreted data, layout, design, and wrote the manuscript; Justin Swartz: Contributed to the collection of literature and writing of the manuscript; Michiel van den Brekel: Critical input and final approval of the manuscript; Stefan Willems: Critical input and final approval of the manuscript; René Spijker: contributed to the design of the search strategy, collection of the literature and final approval of the manuscript; Jacques Neefjes: Critical input and final approval of the manuscript; Charlotte Zuur: Contributed to writing the manuscript, critical input and final approval of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pulte, D.; Brenner, H. Changes in survival in head and neck cancers in the late 20th and early 21st century: A period analysis. Oncologist 2010, 15, 994–1001. [Google Scholar] [CrossRef] [PubMed]
  2. Surveillance Epidemiology and End Results Program Cancer of the Larynx—SEER Fact Sheets. Available online: http://seer.cancer.gov/statfacts/html/laryn.html (accessed on 21 August 2015).
  3. Surveillance Epidemiology and End Results Program Cancer of the Oral Cavity and Pharynx—SEER Fact Sheets. Available online: http://seer.cancer.gov/statfacts/html/oralcav.html (accessed on 21 August 2015).
  4. Al-Sarraf, M. Treatment of locally advanced head and neck cancer: Historical and critical review. Cancer Control 2002, 9, 387–399. [Google Scholar] [PubMed]
  5. Pignon, J.P.; le Maitre, A.; Maillard, E.; Bourhis, J.; Group, M.-N.C. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): An update on 93 randomised trials and 17,346 patients. Radiother. Oncol. 2009, 92, 4–14. [Google Scholar] [CrossRef] [PubMed]
  6. Fakhry, C.; Westra, W.H.; Li, S.; Cmelak, A.; Ridge, J.A.; Pinto, H.; Forastiere, A.; Gillison, M.L. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J. Natl. Cancer Inst. 2008, 100, 261–269. [Google Scholar] [CrossRef] [PubMed]
  7. Forastiere, A.A.; Zhang, Q.; Weber, R.S.; Maor, M.H.; Goepfert, H.; Pajak, T.F.; Morrison, W.; Glisson, B.; Trotti, A.; Ridge, J.A.; et al. Long-term results of RTOG 91–11: A comparison of three nonsurgical treatment strategies to preserve the larynx in patients with locally advanced larynx cancer. J. Clin. Oncol. 2013, 31, 845–852. [Google Scholar] [CrossRef] [PubMed]
  8. Adelstein, D.J.; Li, Y.; Adams, G.L.; Wagner, H.; Kish, J.A.; Ensley, J.F.; Schuller, D.E.; Forastiere, A.A. An intergroup phase III comparison of standard radiation therapy and two schedules of concurrent chemoradiotherapy in patients with unresectable squamous cell head and neck cancer. J. Clin. Oncol. 2003, 21, 92–98. [Google Scholar] [CrossRef] [PubMed]
  9. Ley, J.; Mehan, P.; Wildes, T.M.; Thorstad, W.; Gay, H.A.; Michel, L.; Nussenbaum, B.; Trinkaus, K.; Adkins, D. Cisplatin versus cetuximab given concurrently with definitive radiation therapy for locally advanced head and neck squamous cell carcinoma. Oncology 2013, 85, 290–296. [Google Scholar] [CrossRef] [PubMed]
  10. Caudell, J.J.; Sawrie, S.M.; Spencer, S.A.; Desmond, R.A.; Carroll, W.R.; Peters, G.E.; Nabell, L.M.; Meredith, R.F.; Bonner, J.A. Locoregionally Advanced Head and Neck Cancer Treated With Primary Radiotherapy: A Comparison of the Addition of Cetuximab or Chemotherapy and the Impact of Protocol Treatment. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 676–681. [Google Scholar] [CrossRef] [PubMed]
  11. Wright, J.C.; Cobb, J.P.; Gumport, S.L.; Golomb, F.M.; Safadi, D. Investigation of the relation between clinical and tissue-culture response to chemotherapeutic agents on human cancer. N. Engl. J. Med. 1957, 257, 1207–1211. [Google Scholar] [CrossRef] [PubMed]
  12. Carey, T.E. Head and Neck Tumor Cell Lines. In Atlas of Human Tumor Cell Lines; Hay, R.J., Park, J., Gazdar, A., Eds.; Academic Press, Inc.: San Diego, CA, USA, 1994; pp. 79–120. [Google Scholar]
  13. Rheinwald, J.G.; Beckett, M.A. Tumorigenic keratinocyte lines requiring anchorage and fibroblast support cultured from human squamous cell carcinomas. Cancer Res. 1981, 41, 1657–1663. [Google Scholar] [PubMed]
  14. Easty, D.M.; Easty, G.C.; Carter, R.L.; Monaghan, P.; Butler, L.J. Ten human carcinoma cell lines derived from squamous carcinomas of the head and neck. Br. J. Cancer 1981, 43, 772–785. [Google Scholar] [CrossRef] [PubMed]
  15. Sacks, P.G. Cell, tissue and organ culture as in vitro models to study the biology of squamous cell carcinomas of the head and neck. Cancer Metastasis Rev. 1996, 15, 27–51. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, C.J.; Grandis, J.R.; Carey, T.E.; Gollin, S.M.; Whiteside, T.L.; Koch, W.M.; Ferris, R.L.; Lai, S.Y. Head and neck squamous cell carcinoma cell lines: Established models and rationale for selection. Head Neck 2007, 29, 163–188. [Google Scholar] [CrossRef] [PubMed]
  17. Owen, J.H.; Hauff, S.J.; Tang, A.L.; Graham, M.P.; Czerwinski, M.J.; Kaddoura, M.; Papagerakis, S.; Bradford, C.R.; Carey, T.E.; Prince, M.E. UM-SCC-103: A Unique Tongue Cancer Cell Line That Recapitulates the Tumorigenic Stem Cell Population of the Primary Tumor. Ann. Otol. Rhinol. Laryngol. 2014, 123, 662–672. [Google Scholar] [CrossRef] [PubMed]
  18. Van Zeeburg, H.J.T. Generation and Molecular Characterization of Head and Neck Squamous Cell Lines of Fanconi Anemia Patients. Cancer Res. 2005, 65, 1271–1276. [Google Scholar] [CrossRef] [PubMed]
  19. Bijman, J.T.; Wagener, D.J.; van Rennes, H.; Wessels, J.M.; van den Broek, P. Flow cytometric evaluation of cell dispersion from human head and neck tumors. Cytometry 1985, 6, 334–341. [Google Scholar] [CrossRef] [PubMed]
  20. Kudo, Y.; Kitajjma, S.; Sato, S.; Miyauchi, M.; Ogawa, I.; Takata, T. Establishment of an oral squamous cell carcinoma cell line with high invasive and p27 degradation activities from a lymph node metastasis. Oral Oncol. 2003, 39, 515–520. [Google Scholar] [CrossRef]
  21. Taitz, A.; Petruzzelli, G.; Pak, A.S.; Wright, M.A.; Matthews, J.P.; Raslan, W.F.; Lozano, Y.; Young, M.R. Immune parameters of mice bearing human head and neck cancer. Cancer Immunol. Immunother. 1995, 40, 283–291. [Google Scholar] [CrossRef] [PubMed]
  22. Ji, Z.W.; Oku, N.; Umeda, M.; Komori, T. Establishment of an oral squamous cell carcinoma cell line (NOS-1) exhibiting amplification of the erbB-1 oncogene and point mutation of p53 tumor suppressor gene: Its biological characteristics and animal model of local invasion by orthotopic transplanta. Oral Oncol. 2001, 37, 386–392. [Google Scholar] [CrossRef]
  23. Kim, S.Y.; Chu, K.C.; Lee, H.R.; Lee, K.S.; Carey, T.E. Establishment and characterization of nine new head and neck cancer cell lines. Acta Otolaryngol. 1997, 117, 775–784. [Google Scholar] [CrossRef] [PubMed]
  24. Charoenrat, P.; Rhys-Evans, P.; Eccles, S. Characterization of ten newly-derived human head and neck squamous carcinoma cell lines with special reference to c-erbB proto-oncogene expression. Anticancer Res. 2001, 21, 1953–1963. [Google Scholar]
  25. Owen, J.H.; Graham, M.P.; Chinn, S.B.; Darr, O.F.; Chepeha, D.B.; Wolf, G.T.; Bradford, C.R.; Carey, T.E.; Prince, M.E.P. Novel method of cell line establishment utilizing fluorescence-activated cell sorting resulting in 6 new head and neck squamous cell carcinoma lines. Head Neck 2015, 55, 691–696. [Google Scholar] [CrossRef] [PubMed]
  26. Grénman, R.; Carey, T.E.; McClatchey, K.D.; Wagner, J.G.; Pekkola-Heino, K.; Schwartz, D.R.; Wolf, G.T.; Lacivita, L.P.; Ho, L.; Baker, S.R.; et al. In vitro radiation resistance among cell lines established from patients with squamous cell carcinoma of the head and neck. Cancer 1991, 67, 2741–2747. [Google Scholar] [CrossRef]
  27. Baker, F.L.; Spitzer, G.; Ajani, J.A.; Brock, W.A.; Lukeman, J.; Pathak, S.; Tomasovic, B.; Thielvoldt, D.; Williams, M.; Vines, C.; et al. Drug and radiation sensitivity measurements of successful primary monolayer culturing of human tumor cells using cell-adhesive matrix and supplemented medium. Cancer Res. 1986, 46, 1263–1274. [Google Scholar] [PubMed]
  28. Brock, W.A.; Baker, F.L.; Wike, J.L.; Sivon, S.L.; Peters, L.J. Cellular radiosensitivity of primary head and neck squamous cell carcinomas and local tumor control. Int. J. Radiat. Oncol. Biol. Phys. 1990, 18, 1283–1286. [Google Scholar] [CrossRef]
  29. Brock, W.A.; Baker, F.L.; Peters, L.J. Radiosensitivity of human head and neck squamous cell carcinomas in primary culture and its potential as a predictive assay of tumor radiocurability. Int. J. Radiat. Biol. 1989, 56, 751–760. [Google Scholar] [CrossRef] [PubMed]
  30. Girinsky, T.; Lubin, R.; Pignon, J.P.; Chavaudra, N.; Gazeau, J.; Dubray, B.; Cosset, J.M.; Socie, G.; Fertil, B. Predictive value of in vitro radiosensitivity parameters in head and neck cancers and cervical carcinomas: Preliminary correlations with local control and overall survival. Int. J. Radiat. Oncol. Biol. Phys. 1993, 25, 3–7. [Google Scholar] [CrossRef]
  31. Girinsky, T.; Bernheim, A.; Lubin, R.; Tavakoli-Razavi, T.; Baker, F.; Janot, F.; Wibault, P.; Cosset, J.M.; Duvillard, P.; Duverger, A. In vitro parameters and treatment outcome in head and neck cancers treated with surgery and/or radiation: Cell characterization and correlations with local control and overall survival. Int. J. Radiat. Oncol. Biol. Phys. 1994, 30, 789–794. [Google Scholar] [CrossRef]
  32. Eschwege, F.; Bourhis, J.; Girinski, T.; Lartigau, E.; Guichard, M.; Deblé, D.; Kepta, L.; Wilson, G.D.; Luboinski, B. Predictive assays of radiation response in patients with head and neck squamous cell carcinoma: A review of the Institute Gustave Roussy experience. Int. J. Radiat. Oncol. Biol. Phys. 1997, 39, 849–853. [Google Scholar] [CrossRef]
  33. Puck, T.T.; Marcus, P.I. A rapid method for viable cell titration and clone production with hela cells in tissue culture: The use of x-irradiated cells to supply conditioning factors. Proc. Natl. Acad. Sci. USA 1955, 41, 432–437. [Google Scholar] [CrossRef] [PubMed]
  34. Puck, T.T.; Marcus, P.I.; Cieciura, S.J. Clonal growth of mammalian cells in vitro; growth characteristics of colonies from single HeLa cells with and without a feeder layer. J. Exp. Med. 1956, 103, 273–283. [Google Scholar] [CrossRef] [PubMed]
  35. Hamburger, A.W.; Salmon, S.E. Primary bioassay of human tumor stem cells. Science 1977, 197, 461–463. [Google Scholar] [CrossRef] [PubMed]
  36. Courtenay, V.D.; Mills, J. An in vitro colony assay for human tumours grown in immune-suppressed mice and treated in vivo with cytotoxic agents. Br. J. Cancer 1978, 37, 261–268. [Google Scholar] [CrossRef] [PubMed]
  37. Johns, M.E. The clonal assay of head and neck tumor cells: Results and clinical correlations. Laryngoscope 1982, 92, 1–26. [Google Scholar] [CrossRef] [PubMed]
  38. Macpherson, I.; Montagnier, L. Agar suspension culture for the selective assay of cells transformed by polyoma virus. Virology 1964, 23, 291–294. [Google Scholar] [CrossRef]
  39. Mattox, D.E.; von Hoff, D.D.; Clark, G.M.; Aufdemorte, T.B. Factors that influence growth of head and neck squamous carcinoma in the soft agar cloning assay. Cancer 1984, 53, 1736–1740. [Google Scholar] [CrossRef]
  40. Mattox, D.E.; von Hoff, D.D. In vitro stem cell assay in head and neck squamous carcinoma. Am. J. Surg. 1980, 140, 527–530. [Google Scholar] [CrossRef]
  41. Mattox, D.E.; von Hoff, D.D. Culture of human head and neck cancer stem cells using soft agar. Arch. Otolaryngol. 1980, 106, 672–674. [Google Scholar] [CrossRef] [PubMed]
  42. Schiff, L.J.; Shugar, M.A. Growth of human head and neck squamous cell carcinoma stem cells in agarose. Cancer 1984, 53, 286–290. [Google Scholar] [CrossRef]
  43. Cobleigh, M.A.; Gallagher, P.A.; Hill, J.H.; Applebaum, E.L.; McGuire, W.P. Growth of human squamous head and neck cancer in vitro. Am. J. Pathol. 1984, 115, 397–402. [Google Scholar] [PubMed]
  44. Rofstad, E.K.; Wahl, A.; Brustad, T. Radiation sensitivity in vitro of cells isolated from human tumor surgical specimens. Cancer Res. 1987, 47, 106–110. [Google Scholar] [PubMed]
  45. Stausbol-Gron, B.; Nielsen, O.S.; Moller Bentzen, S.; Overgaard, J. Selective assessment of in vitro radiosensitivity of tumour cells and fibroblasts from single tumour biopsies using immunocytochemical identification of colonies in the soft agar clonogenic assay. Radiother. Oncol. 1995, 37, 87–99. [Google Scholar] [CrossRef]
  46. Stausbol-Gron, B.; Bentzen, S.M.; Jorgensen, K.E.; Nielsen, O.S.; Bundgaard, T.; Overgaard, J. In vitro radiosensitivity of tumour cells and fibroblasts derived from head and neck carcinomas: Mutual relationship and correlation with clinical data. Br. J. Cancer 1999, 79, 1074–1084. [Google Scholar] [CrossRef] [PubMed]
  47. Stausbol-Gron, B.; Overgaard, J. Relationship between tumour cell in vitro radiosensitivity and clinical outcome after curative radiotherapy for squamous cell carcinoma of the head and neck. Radiother. Oncol. 1999, 50, 47–55. [Google Scholar] [CrossRef]
  48. Björk-Eriksson, T.; West, C.M.; Karlsson, E.; Slevin, N.J.; Davidson, S.E.; James, R.D.; Mercke, C. The in vitro radiosensitivity of human head and neck cancers. Br. J. Cancer 1998, 77, 2371–2375. [Google Scholar] [CrossRef] [PubMed]
  49. Björk-Eriksson, T.; West, C.; Karlsson, E.; Mercke, C. Tumor radiosensitivity (SF2) is a prognostic factor for local control in head and neck cancers. Int. J. Radiat. Oncol. Biol. Phys. 2000, 46, 13–19. [Google Scholar] [CrossRef]
  50. Dollner, R.; Dietz, A.; Kopun, M.; Helbig, M.; Wallner, F.; Granzow, C. Ex vivo responsiveness of head and neck squamous cell carcinoma to glufosfamide, a novel alkylating agent. Anticancer Res. 2004, 24, 2947–2951. [Google Scholar] [PubMed]
  51. Dollner, R.; Granzow, C.; Tschop, K.; Dietz, A. Ex vivo responsiveness of head and neck squamous cell carcinoma to vinorelbine. Anticancer Res. 2006, 26, 2361–2365. [Google Scholar] [PubMed]
  52. Dollner, R.; Granzow, C.; Helmke, B.M.; Ruess, A.; Schad, A.; Dietz, A. The Impact of Stromal Cell Contamination on Chemosensitivity Testing of Head and Neck Carcinoma. Anticancer Res. 2004, 24, 325–331. [Google Scholar] [PubMed]
  53. Dollner, R.; Granzow, C.; Neudert, M.; Dietz, A. Ex vivo chemosensitivity of head and neck carcinoma to cytostatic drug combinations. Anticancer Res. 2006, 26, 1651–1655. [Google Scholar] [PubMed]
  54. Horn, I.-S.; Wichmann, G.; Mozet, C.; Dietz, A.; Dollner, R.; Tschöp, K.; Boehm, A. Heterogeneity of epithelial and stromal cells of head and neck squamous cell carcinomas in ex vivo chemoresponse. Cancer Chemother. Pharmacol. 2010, 65, 1153–1163. [Google Scholar] [CrossRef] [PubMed]
  55. Dietz, A.; Boehm, A.; Reiche, A.; Mueller, G.; Kruber, P.; Keilholz, U.; Niederwieser, D.; Wichmann, G. Prediction of outcome of TPF with or without cetuximab induction chemotherapy in head and neck squamous cell carcinoma ( HNSCC) using the FLAVINO assay. J. Clin. Oncol. 2010, 28, 15s. [Google Scholar]
  56. Wichmann, G.; Körner, C.; Boehm, A.; Mozet, C.; Dietz, A. Stimulation by Monocyte Chemoattractant Protein-1 Modulates the Ex-vivo Colony Formation by Head and Neck Squamous Cell Carcinoma Cells. Anticancer Res. 2015, 35, 3917–3924. [Google Scholar] [PubMed]
  57. Leighton, J. A sponge matrix method for tissue culture; formation of organized aggregates of cells in vitro. J. Natl. Cancer Inst. 1951, 12, 545–561. [Google Scholar] [PubMed]
  58. Sherwin, R.P.; Richters, A.; Yellin, A.E.; Donovan, A.J. Histoculture of human breast cancers. J. Surg. Oncol. 1980, 13, 9–20. [Google Scholar] [CrossRef] [PubMed]
  59. Furukawa, T.; Kubota, T.; Hoffman, R.M. Clinical applications of the histoculture drug response assay. Clin. Cancer Res. 1995, 1, 305–311. [Google Scholar] [PubMed]
  60. Hoffman, R.M. In vitro sensitivity assays in cancer: A review, analysis, and prognosis. J. Clin. Lab. Anal. 1991, 5, 133–143. [Google Scholar] [CrossRef] [PubMed]
  61. Burford-Mason, A.P.; Dardick, I.; MacKay, A. Collagen gel cultures of normal salivary gland: Conditions for continued proliferation and maintenance of major cell phenotypes in vitro. Laryngoscope 1994, 104, 335–340. [Google Scholar] [CrossRef] [PubMed]
  62. Hasegawa, Y.; Goto, M.; Hanai, N.; Ijichi, K.; Adachi, M.; Terada, A.; Hyodo, I.; Ogawa, T.; Furukawa, T. Evaluation of optimal drug concentration in histoculture drug response assay in association with clinical efficacy for head and neck cancer. Oral Oncol. 2007, 43, 749–756. [Google Scholar] [CrossRef] [PubMed]
  63. Robbins, K.T.; Connors, K.M.; Storniolo, A.M.; Hanchett, C.; Hoffman, R.M. Sponge-gel-supported histoculture drug-response assay for head and neck cancer. Correlations with clinical response to cisplatin. Arch. Otolaryngol. Head Neck Surg. 1994, 120, 288–292. [Google Scholar] [CrossRef] [PubMed]
  64. Singh, B.; Li, R.; Xu, L.; Poluri, A.; Patel, S.; Shaha, A.R.; Pfister, D.; Sherman, E.; Goberdhan, A.; Hoffman, R.M.; et al. Prediction of survival in patients with head and neck cancer using the histoculture drug response assay. Head Neck 2002, 24, 437–442. [Google Scholar] [CrossRef] [PubMed]
  65. Ariyoshi, Y.; Shimahara, M.; Tanigawa, N. Study on chemosensitivity of oral squamous cell carcinomas by histoculture drug response assay. Oral Oncol. 2003, 39, 701–707. [Google Scholar] [CrossRef]
  66. Pathak, K.A.; Juvekar, A.S.; Radhakrishnan, D.K.; Deshpande, M.S.; Pai, V.R.; Chaturvedi, P.; Pai, P.S.; Chaukar, D.A.; D’Cruz, A.K.; Parikh, P.M. In vitro chemosensitivity profile of oral squamous cell cancer and its correlation with clinical response to chemotherapy. Indian J. Cancer 2007, 44, 142–146. [Google Scholar] [CrossRef] [PubMed]
  67. Kubota, T.; Sasano, N.; Abe, O.; Nakao, I.; Kawamura, E.; Saito, T.; Endo, M.; Kimura, K.; Demura, H.; Sasano, H.; et al. Potential of the histoculture drug-response assay to contribute to cancer patient survival. Clin. Cancer Res. 1995, 1, 1537–1543. [Google Scholar] [PubMed]
  68. Suda, S.; Akiyama, S.; Sekiguchi, H.; Kasai, Y.; Ito, K.; Nakao, A. Evaluation of the histoculture drug response assay as a sensitivity test for anticancer agents. Surg. Today 2002, 32, 477–481. [Google Scholar] [CrossRef] [PubMed]
  69. Ohie, S.; Udagawa, Y.; Kozu, A.; Komuro, Y.; Aoki, D.; Nozawa, S.; Moossa, A.R.; Hoffman, R.M. Cisplatin sensitivity of ovarian cancer in the histoculture drug response assay correlates to clinical response to combination chemotherapy with cisplatin, doxorubicin and cyclophosphamide. Anticancer Res. 2000, 20, 2049–2054. [Google Scholar] [PubMed]
  70. Nakada, S.; Aoki, D.; Ohie, S.; Horiuchi, M.; Suzuki, N.; Kanasugi, M.; Susumu, N.; Udagawa, Y.; Nozawa, S. Chemosensitivity testing of ovarian cancer using the histoculture drug response assay: Sensitivity to cisplatin and clinical response. Int. J. Gynecol. Cancer 2005, 15, 445–452. [Google Scholar] [CrossRef] [PubMed]
  71. Jung, P.-S.; Kim, D.-Y.; Kim, M.-B.; Lee, S.-W.; Kim, J.-H.; Kim, Y.-M.; Kim, Y.-T.; Hoffman, R.M.; Nam, J.-H. Progression-free survival is accurately predicted in patients treated with chemotherapy for epithelial ovarian cancer by the histoculture drug response assay in a prospective correlative clinical trial at a single institution. Anticancer Res. 2013, 33, 1029–1034. [Google Scholar] [PubMed]
  72. Gerlach, M.M.; Merz, F.; Wichmann, G.; Kubick, C.; Wittekind, C.; Lordick, F.; Dietz, A.; Bechmann, I. Slice cultures from head and neck squamous cell carcinoma: A novel test system for drug susceptibility and mechanisms of resistance. Br. J. Cancer 2014, 110, 479–488. [Google Scholar] [CrossRef] [PubMed]
  73. Peria, M.; Donnadieu, J.; Racz, C.; Ikoli, J.-F.; Galmiche, A.; Chauffert, B.; Page, C. Evaluation of individual sensitivity of Head and Neck Squamous Cell Carcinoma to cetuximab by short-term culture of tumor slices. Head Neck 2015. [Google Scholar] [CrossRef] [PubMed]
  74. Freudlsperger, C.; Horn, D.; Weißfuß, S.; Weichert, W.; Weber, K.-J.; Saure, D.; Sharma, S.; Dyckhoff, G.; Grabe, N.; Plinkert, P.; et al. Phosphorylation of AKT(Ser473) serves as an independent prognostic marker for radiosensitivity in advanced head and neck squamous cell carcinoma. Int. J. Cancer 2015, 136, 2775–2785. [Google Scholar] [CrossRef] [PubMed]
  75. Freshney, R.I. Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications, 6th ed.; Wiley-Liss: Wilmington, DE, USA, 2010. [Google Scholar]
  76. Freyer, J.P. Role of necrosis in regulating the growth saturation of multicellular spheroids. Cancer Res. 1988, 48, 2432–2439. [Google Scholar] [PubMed]
  77. Schwachöfer, J.H.; Acker, H.; Crooijmans, R.P.; van Gasteren, J.J.; Holtermann, G.; Hoogenhout, J.; Jerusalem, C.R.; Kal, H.B. Oxygen tensions in two human tumor cell lines grown and irradiated as multicellular spheroids. Anticancer Res. 1991, 11, 273–279. [Google Scholar] [PubMed]
  78. Heimdal, J.H.; Aarstad, H.J.; Olofsson, J. Monocytes secrete interleukin-6 when co-cultured in vitro with benign or malignant autologous fragment spheroids from squamous cell carcinoma patients. Scand. J. Immunol. 2000, 51, 271–278. [Google Scholar] [CrossRef] [PubMed]
  79. Kross, K.W.; Heimdal, J.-H.; Olsnes, C.; Olofsson, J.; Aarstad, H.J. Head and neck squamous cell carcinoma spheroid- and monocyte spheroid-stimulated IL-6 and monocyte chemotactic protein-1 secretion are related to TNM stage, inflammatory state and tumor macrophage density. Acta Otolaryngol. 2005, 125, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  80. Kross, K.W.; Heimdal, J.H.; Olsnes, C.; Olofsson, J.; Aarstad, H.J. Co-culture of head and neck squamous cell carcinoma spheroids with autologous monocytes predicts prognosis. Scand. J. Immunol. 2008, 67, 392–399. [Google Scholar] [CrossRef] [PubMed]
  81. Lim, Y.C.; Oh, S.-Y.; Cha, Y.Y.; Kim, S.-H.; Jin, X.; Kim, H. Cancer stem cell traits in squamospheres derived from primary head and neck squamous cell carcinomas. Oral Oncol. 2011, 47, 83–91. [Google Scholar] [CrossRef] [PubMed]
  82. Pozzi, V.; Sartini, D.; Rocchetti, R.; Santarelli, A.; Rubini, C.; Morganti, S.; Giuliante, R.; Calabrese, S.; di Ruscio, G.; Orlando, F.; et al. Identification and Characterization of Cancer Stem Cells from Head and Neck Squamous Cell Carcinoma Cell Lines. Cell. Physiol. Biochem. 2015, 36, 784–798. [Google Scholar] [CrossRef] [PubMed]
  83. Lim, Y.C.; Oh, S.-Y.; Kim, H. Cellular characteristics of head and neck cancer stem cells in type IV collagen-coated adherent cultures. Exp. Cell Res. 2012, 318, 1104–1111. [Google Scholar] [CrossRef] [PubMed]
  84. Leong, H.S.; Chong, F.T.; Sew, P.H.; Lau, D.P.; Wong, B.H.; Teh, B.-T.; Tan, D.S.W.; Iyer, N.G. Targeting Cancer Stem Cell Plasticity Through Modulation of Epidermal Growth Factor and Insulin-Like Growth Factor Receptor Signaling in Head and Neck Squamous Cell Cancer. Stem Cells Transl. Med. 2014, 3, 1055–1065. [Google Scholar] [CrossRef] [PubMed]
  85. Sun, S.; Liu, S.; Duan, S.Z.; Zhang, L.; Zhou, H.; Hu, Y.; Zhou, X.; Shi, C.; Zhou, R.; Zhang, Z. Targeting the c-Met/FZD8 Signaling Axis Eliminates Patient-Derived Cancer Stem-like Cells in Head and Neck Squamous Carcinomas. Cancer Res. 2014, 74, 7546–7559. [Google Scholar] [CrossRef] [PubMed]
  86. Kopf-Maier, P.; Kolon, B. An organoid culture assay (OCA) for determining the drug sensitivity of human tumors. Int. J. Cancer 1992, 51, 99–107. [Google Scholar] [CrossRef] [PubMed]
  87. Kopf-Maier, P. A new approach for realizing the “antioncogram”. Life Sci. 1992, 50, 1711–1718. [Google Scholar] [CrossRef]
  88. Von Hoff, D.D. “Send this patient’s tumor for culture and sensitivity”. N. Engl. J. Med. 1983, 308, 154–155. [Google Scholar] [CrossRef] [PubMed]
  89. Garozzo, A.; Rossi, M.; Denaro, A.; Allegra, E.; Santangelo, G.; Amato, M.; Tomasello, C. In vitro short-term chemosensitivity test in head and neck tumors. J. Chemother. 1989, 1, 59–63. [Google Scholar] [PubMed]
  90. Elprana, D.; Schwachöfer, J.; Kuijpers, W.; van den Broek, P.; Wagener, D.J. In vitro chemosensitivity testing of squamous cell carcinoma of the head and neck. A preliminary report. Acta Otolaryngol. 1987, 103, 529–536. [Google Scholar] [PubMed]
  91. Elprana, D.; Schwachöfer, J.; Kuijpers, W.; van den Broek, P.; Wagener, D.J. Cytotoxic drug sensitivity of squamous cell carcinoma as predicted by an in vitro testing model. Anticancer Res. 1989, 9, 1089–1094. [Google Scholar] [PubMed]
  92. Hattersley, S.M.; Sylvester, D.C.; Dyer, C.E.; Stafford, N.D.; Haswell, S.J.; Greenman, J. A microfluidic system for testing the responses of head and neck squamous cell carcinoma tissue biopsies to treatment with chemotherapy drugs. Ann. Biomed. Eng. 2012, 40, 1277–1288. [Google Scholar] [CrossRef] [PubMed]
  93. Sylvester, D.; Hattersley, S.M.; Stafford, N.D.; Haswell, S.J.; Greenman, J. Development of microfluidic-based analytical methodology for studying the effects of chemotherapy agents on cancer tissue. Curr. Anal. Chem. 2013, 9, 2–8. [Google Scholar] [CrossRef]
  94. Carr, S.D.; Green, V.L.; Stafford, N.D.; Greenman, J. Analysis of radiation-induced cell death in head and neck squamous cell carcinoma and rat liver maintained in microfluidic devices. Otolaryngol. Head Neck Surg. 2014, 150, 73–80. [Google Scholar] [CrossRef] [PubMed]
  95. Sivagnanam, V.; Gijs, M.A.M. Exploring living multicellular organisms, organs, and tissues using microfluidic systems. Chem. Rev. 2013, 113, 3214–3247. [Google Scholar] [CrossRef] [PubMed]
  96. Champion, A.R.; Hanson, J.A.; Venables, S.E.; McGregor, A.D.; Gaffney, C.C. Determination of radiosensitivity in established and primary squamous cell carcinoma cultures using the micronucleus assay. Eur. J. Cancer 1997, 33, 453–462. [Google Scholar] [CrossRef]
  97. Liu, C.; Tsao, M.S. Proto-oncogene and growth factor/receptor expression in the establishment of primary human non-small cell lung carcinoma cell lines. Am. J. Pathol. 1993, 142, 413–23. [Google Scholar] [PubMed]
  98. Köhler, G.; Veelken, H.; Rosenthal, F.; Mackensen, A.; Lindemann, A.; Schaefer, H.E.; Lahn, M. Oncogene and HSP-70 expression in primary tumor cell cultures of renal cell carcinoma compared to their corresponding cell line. Anticancer Res. 1997, 17, 3225–3231. [Google Scholar] [PubMed]
  99. Shapiro, J.R.; Shapiro, W.R. The subpopulations and isolated cell types of freshly resected high grade human gliomas: Their influence on the tumor’s evolution in vivo and behavior and therapy in vitro. Cancer Metastasis Rev. 1985, 4, 107–124. [Google Scholar] [CrossRef] [PubMed]
  100. Worsham, M.J.; Wolman, S.R.; Carey, T.E.; Zarbo, R.J.; Benninger, M.S.; van Dyke, D.L. Chromosomal aberrations identified in culture of squamous carcinomas are confirmed by fluorescence in situ hybridisation. Mol. Pathol. 1999, 52, 42–46. [Google Scholar] [CrossRef] [PubMed]
  101. Bjerkvig, R. Spheroid Culture in Cancer Research, 1st ed.; CRC Press: Boca Raton, FL, USA, 1992. [Google Scholar]
  102. Ragin, C.C.; Reshmi, S.C.; Gollin, S.M. Mapping and analysis of HPV16 integration sites in a head and neck cancer cell line. Int. J. Cancer 2004, 110, 701–709. [Google Scholar] [CrossRef] [PubMed]
  103. Gisselsson, D.; Jonson, T.; Yu, C.; Martins, C.; Mandahl, N.; Wiegant, J.; Jin, Y.; Mertens, F.; Jin, C. Centrosomal abnormalities, multipolar mitoses, and chromosomal instability in head and neck tumours with dysfunctional telomeres. Br. J. Cancer 2002, 87, 202–207. [Google Scholar] [CrossRef] [PubMed]
  104. Leess, F.R.; Bredenkamp, J.K.; Lichtenstein, A.; Mickel, R.A. Lymphokine-activated killing of autologous and allogeneic short-term cultured head and neck squamous carcinomas. Laryngoscope 1989, 99, 1255–1261. [Google Scholar] [CrossRef] [PubMed]
  105. Jin, Y.; Mertens, F.; Mandahl, N.; Heim, S.; Olegård, C.; Wennerberg, J.; Biörklund, A.; Mitelman, F. Chromosome abnormalities in eighty-three head and neck squamous cell carcinomas: Influence of culture conditions on karyotypic pattern. Cancer Res. 1993, 53, 2140–2146. [Google Scholar] [PubMed]
  106. Heppner, G.H.; Miller, B.E. Therapeutic implications of tumor heterogeneity. Semin. Oncol. 1989, 16, 91–105. [Google Scholar] [PubMed]
  107. Von Hoff, D.D. He’s not going to talk about in vitro predictive assays again, is he? J. Natl. Cancer Inst. 1990, 82, 96–101. [Google Scholar] [CrossRef] [PubMed]
  108. Moscona, A. Cell suspensions from organ rudiments of chick embryos. Exp. Cell. Res. 1952, 3, 535–539. [Google Scholar] [CrossRef]
  109. Miller, B.E.; Miller, F.R.; Heppner, G.H. Assessing tumor drug sensitivity by a new in vitro assay which preserves tumor heterogeneity and subpopulation interactions. J. Cell. Physiol. Suppl. 1984, 3, 105–116. [Google Scholar] [CrossRef] [PubMed]
  110. Lawler, E.M.; Miller, F.R.; Heppner, G.H. Significance of three-dimensional growth patterns of mammary tissues in collagen gels. In Vitro 1983, 19, 600–610. [Google Scholar] [CrossRef] [PubMed]
  111. Miller, B.E.; Miller, F.R.; Heppner, G.H. Factors affecting growth and drug sensitivity of mouse mammary tumor lines in collagen gel cultures. Cancer Res. 1985, 45, 4200–4205. [Google Scholar] [PubMed]
  112. Kobayashi, H.; Man, S.; Graham, C.H.; Kapitain, S.J.; Teicher, B.A.; Kerbel, R.S. Acquired multicellular-mediated resistance to alkylating agents in cancer. Proc. Natl. Acad. Sci. USA 1993, 90, 3294–3298. [Google Scholar] [CrossRef] [PubMed]
  113. Miller, B.E.; Miller, F.R.; Heppner, G.H. Interactions between tumor subpopulations affecting their sensitivity to the antineoplastic agents cyclophosphamide and methotrexate. Cancer Res. 1981, 41, 4378–4381. [Google Scholar] [PubMed]
  114. Lawton, P.A.; Hodgkiss, R.J.; Eyden, B.P.; Joiner, M.C. Growth of fibroblasts as a potential confounding factor in soft agar clonogenic assays for tumour cell radiosensitivity. Radiother. Oncol. 1994, 32, 218–225. [Google Scholar] [CrossRef]
  115. Balermpas, P.; Rödel, F.; Rödel, C.; Krause, M.; Linge, A.; Lohaus, F.; Baumann, M.; Tinhofer, I.; Budach, V.; Gkika, E.; et al. CD8+ tumour-infiltrating lymphocytes in relation to HPV status and clinical outcome in patients with head and neck cancer after postoperative chemoradiotherapy: A multicentre study of the German cancer consortium radiation oncology group (DKTK-ROG). Int. J. Cancer 2015. [Google Scholar] [CrossRef] [PubMed]
  116. Heimdal, J.H.; Aarstad, H.J.; Olsnes, C.; Olofsson, J. Human autologous monocytes and monocyte-derived macrophages in co-culture with carcinoma F-spheroids secrete IL-6 by a non-CD 14-dependent pathway. Scand. J. Immunol. 2001, 53, 162–170. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Dohmen, A.J.C.; Swartz, J.E.; Van Den Brekel, M.W.M.; Willems, S.M.; Spijker, R.; Neefjes, J.; Zuur, C.L. Feasibility of Primary Tumor Culture Models and Preclinical Prediction Assays for Head and Neck Cancer: A Narrative Review. Cancers 2015, 7, 1716-1742. https://doi.org/10.3390/cancers7030858

AMA Style

Dohmen AJC, Swartz JE, Van Den Brekel MWM, Willems SM, Spijker R, Neefjes J, Zuur CL. Feasibility of Primary Tumor Culture Models and Preclinical Prediction Assays for Head and Neck Cancer: A Narrative Review. Cancers. 2015; 7(3):1716-1742. https://doi.org/10.3390/cancers7030858

Chicago/Turabian Style

Dohmen, Amy J. C., Justin E. Swartz, Michiel W. M. Van Den Brekel, Stefan M. Willems, René Spijker, Jacques Neefjes, and Charlotte L. Zuur. 2015. "Feasibility of Primary Tumor Culture Models and Preclinical Prediction Assays for Head and Neck Cancer: A Narrative Review" Cancers 7, no. 3: 1716-1742. https://doi.org/10.3390/cancers7030858

Article Metrics

Back to TopTop