JCDR - Register at Journal of Clinical and Diagnostic Research
Journal of Clinical and Diagnostic Research, ISSN - 0973 - 709X
Pathology Section DOI : 10.7860/JCDR/2017/25077.10022
Year : 2017 | Month : Jun | Volume : 11 | Issue : 6 Full Version Page : EC09 - EC16

Evaluation of Hypoxia Inducible Factor-1α and Glucose Transporter-1 Expression in Non Melanoma Skin Cancer: An Immunohistochemical Study

Iman Seleit1, Ola Ahmed Bakry2, Dalia Rifaat Al-Sharaky3, Rania Abdel Aziz Ragab4, shimaa Ahmed Al-Shiemy5

1 Professor, Department of Dermatology, Andrology and S.T.Ds, Faculty of Medicine Menoufia University, Egypt.
2 Assistant Professor, Department of Dermatology, Andrology and S.T.Ds, Faculty of Medicine, Menoufia University, Egypt.
3 Assistant Professor, Department of Pathology, Faculty of Medicine, Menoufia University, Egypt.
4 Resident, Department of Dermatology, Andrology and S.T.Ds, Faculty of Medicine, Menoufia University, Egypt.
5 Resident, Department of Dermatology, Andrology and S.T.Ds, Faculty of Medicine, Menoufia University, Egypt.


NAME, ADDRESS, E-MAIL ID OF THE CORRESPONDING AUTHOR: Dr. Dalia Rifaat Al-Sharaky, Assistant Professor, Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia Shebein El Kom, 32817, Egypt.
E-mail: daliah_alsharaky@yahoo.com
Abstract

Introduction

Hypoxia Inducible Factor-1 (HIF-1) is a mediator enabling cell adaptation to hypoxia. It plays its role mainly through transcription of many target genes including Glucose Transporter-1 (GLUT-1) gene.

Aim

The present work aimed at evaluating the pattern and distribution of HIF-1α and GLUT-1 in each case and control.

Materials and Methods

A case-control and retrospective study was conducted on archival blocks diagnosed from pathology department as, Basal Cell Carcinoma (BCC, 20 cases), cutaneous Squamous Cell Carcinoma (SCC, 20 cases) and 20 normal site-matched skin biopsies from age and gender-matched healthy subjects as a control.

Evaluation of both HIF-1α and GLUT1 expression using standard immunohistochemical techniques was performed on cut divtions from selected paraffin embedded blocks.

Results

HIF-1α was expressed in 90%, 35% and 100% of normal skin, BCC and SCC tumour islands respectively. It was up regulated in both BCC and SCC compared with normal skin (p= 0.001, p<0.001 respectively). GLUT-1 was expressed in 100%, 70% and 100% of normal skin, BCC and SCC tumour islands respectively. It was down regulated in Non Melanoma Skin Cancer (NMSC) cases compared with normal skin (p=0.004). HIF-1α and GLUT-1 localization in tumour nests was central, peripheral or central and peripheral. Both HIF-1α and GLUT-1 showed variable expression in stroma, adnexa and inflammatory cells. No significant correlation was found between Histo (H) score or expression percentage values of HIF-1α and those of GLUT-1 in tumour islands or in overlying epidermis either in BCC or SCC.

Conclusion

HIF-1α may have a role in NMSC pathogenesis through adaptation to hypoxia which results from excessive proliferation. GLUT-1 down regulation in NMSC may be explained by its consumption by proliferating tumour cells. The expression of HIF-1α and GLUT-1 in normal epidermis, stromal and adnexal structures needs further research.

Keywords

Introduction

NMSC, consisting of BCC and SCC, is 18-20 times more frequent than cutaneous malignant melanoma [1]. According to The Egyptian Pathology-Based Cancer Registry (2001-2010), NMSC in Egypt represented 4.4% of total malignancies [2].

Hypoxia plays a vital role in carcinogenesis. In tumour microenvironment, metabolic reprogramming and changes in gene expression are necessary for adaptation to decreased oxygen availability. HIFs being oxygen sensitive transcription factors help in adaptation to hypoxic environment. They are important mediators of cellular response to stress. Metabolic changes occurring during tumourigenesis are, in part, under hypoxia and HIFs regulation. Moreover, inflammatory signaling and infiltration secondary to hypoxia are clear drivers of tumour progression [3].

HIF-1 is the intrinsic survival factor of tumour cells to overcome oxygen and nutrient deficits during proliferation and progression [4] through mediation of the transcription of over 200 target genes, including genes of Vascular Endothelial Growth Factor (VEGF) and GLUT-1 [5]. It induces expression of target genes by DNA binding in the area of Hypoxia Response Element (HRE) which gives rise to a series of subsequent processes like inhibition of apoptosis, switch to anaerobic metabolism, angiogenesis, cell proliferation and erythropoiesis, all of which stimulate carcinogenesis and metastasis [6].

HIF-1 is a heterodimeric protein consisting of a constitutively expressed β-subunit (HIF-1β) and α-subunit (HIF-1α). Under hypoxia, HIF-1α is stabilized and dimerizes with HIF-1β interacting with the co-activator CBP/p300 to bind to the HRE on the promoter regions in various target genes [7,8].

GLUT-1 is the most common glucose transporter in humans. It facilitates the transport of glucose across the plasma membranes of mammalian cells [9] and is responsible for the low level of basal glucose uptake required to sustain respiration in all cells [10]. Previous studies indicated that upregulation of GLUT-1 contributed to improve glucose metabolism in rapidly proliferating cancer cells [9], which are energy-dependent [11].

This study aimed at evaluating the pattern and distribution of HIF-1α and GLUT-1 expression in NMSC and their relationship with clinicopathologic parameters of selected cases.

Materials and Methods

This case-control study included 60 subjects (20 cases with nodular BCC, 20 cases with cutaneous SCC and 20 age and sex-matched healthy subjects as a control group). Cases of BCC and SCC were selected from Dermatology Outpatient Clinic, Menoufiya University Hospital, Egypt during the period from March 2014 to March 2015. For the retrospective part of the study, tissue blocks of archived cases in Pathology Department, Menoufiya University were used. Normal skin samples were obtained from subjects attending Plastic Surgery Department. Biopsies from cases and control subjects were site-matched. Clinical data describing patients’ demographics (age and gender) as well as the clinical variables (site, size of lesions and disease duration) were obtained and documented.

Ethics

A written consent form approved by The Local Ethical Research Committee in Menoufia Faculty of Medicine was obtained from every subject before the study initiation. This was also in accordance with the Helsinki Declaration of 1975 (revised in 2000).

Histopathological Examination of H&E Stained Sections was done for

a) BCC: Histopathological types and status of the surgical margins;

b) SCC: Grading according to Broder’s classification [12], Staging according TNM staging system [13], status of the surgical margins and status of the lymph nodes.

Immunohistochemical (IHC) Staining for HIF-1α and GLUT-1

Sections were cut from the paraffin-embedded blocks were stained with both rabbit monoclonal antibody (Cat.# CME349 A,B) raised against HIF-1α which was received as concentrated 0.1mL (BIOCARE MEDICAL, LLC. 4040 Pike Lane Concord, CA 94520 USA) and with rabbit polyclonal antibody (Cat. #RB-9052-R7) raised against GLUT-1. It was received as 7.0 mL ready to use (Lab Vision Corporation 46360 Fermont Blvd. Fermont, CA 94538-6406, USA). Procedure of IHC staining was done according to received datasheet of the used antibodies.

Interpretation of HIF-1αand GLUT-1 Immunostaining

HIF-1α expression was confirmed by cytoplasmic and/or nuclear stain in examined cells [14]. GLUT-1 expression was confirmed by membranous staining, sometimes with cytoplasmic staining [15]. Erythrocyte positivity was considered as internal positive control for GLUT-1 [16].

For every antibody, the following items were assessed:

1- The epidermis of normal skin biopsies, and the overlying epidermis in BCC and SCC were assessed for:

a) Expression: positive or negative

b) Percentage of positive cells: assessed at 200X magnification field [17]:

c) Histoscore (H score): H score was calculated to all studied cases according to equation:

H score = 1x% of mildly stained cells, + 2x% moderately stained cells, + 3x% of strongly stained cells.

d) Distribution: patchy or diffuse;

e) Pattern: cytoplasmic, membranous or membrano-cytoplasmic (for GLUT-1) or cytoplasmic or nuclear (for HIF-1α);

f) Type of the stained epidermal cells: basal or basal and suprabasal;

2- Tumour islands in BCC and SCC were assessed for:

a) Expression: positive or negative;

b) Percentage of positive cells: assessed at 200X magnification field;

c) Histo-score (H score) [18];

d) Distribution: patchy or diffuse;

e) Pattern;

f) Localization: peripheral, central, or central and peripheral immunoreactivity;

3- The dermis of normal skin biopsies, and stroma of BCC and SCC were assessed for the following:

a) Expression: positive or negative;

b) Type of stained dermal cells: inflammatory or adnexa;

Statistical Analysis

Data were collected, tabulated and statistically analyzed using a personal computer with “(SPSS) version 11.0” program (SPSS Inc, Chicago, Illinois, USA). Values were expressed in number, percentage, mean±standard deviation (X±SD) when appropriate. Fisher’s-exact test, Chi-square test (χ2), Mann-Whitney U test, Spearman’s coefficient were used. Differences were considered statistically significant with p< 0.05.

Results

Clinical and histopathological data of the studied cases are summarized in [Table/Fig-1].

Clinical and histopathological characteristics of the studied NMSC cases.

VariableBCC No = 20SCC NO = 20
Age (years)X±SD61.7±9.9662.30±12.36
Duration (months)X±SD10.67±3.427.82±2.13
Size (cm)X±SD1.67±0.683.43±2.09
GenderMales FemalesNo (%)5 (25)15 (75)No (%)8 (40)12 (60)
SiteFace and scalp ExtremitiesVulva20 (100)0 (0)0 (0)8 (40)5 (25)7 (35)
Clinical TypesNodulo-ulcerative20(100)--
Surgical MarginsInvolved Free8 (40)12 (60)10 (50)10 (50)
Histologic gradeI IIIII--7 (35) 10 (50)3 (15)
Histologic typesSolid 18 (90)Adenoid 1 (5)keratotic 1 (5)Conventional 15 (75) Sarcomatoid 5 (25)
StagingEarly stageLate stage--13 (65)7 (35)
Lymph nodesInvolvedNot involved--9 (45)11 (55)

Immunohistochemical expression of HIF-1α and GLUT-1 in studied groups: Expression of HIF-1a and GLUT-1 as shown in [Table/Fig-2,3,4,5,6,7,8 and 9].

Comparison between the studied groups regarding HIF-1α expression.

VariableBCC N = 20SCC N=20Normal skin N=20p-value
No%No%No%
Overlying epidermis
ExpressionPositiveNegative191955164802018 29010p1 = 1.0p2=0.66p3=1.0p4=0.34
DistributionPatchyDiffuse10952.647.4412257514477.822.2p1=0.11p2=0.002p3=0.009p4=0.1
PatternCytoplasmicNuclear Nucleo- cytoplasmic0217010.589.5001600100601233.3066.7p1=0.01p2=0.02p3=0.001p4=0.49
LocalizationBasalAll layers21710.589.501601000180100p1=0.49p2=NAp3=0.54p4=0.49
PercentX±SDRange62.89±25.5110 – 9080.0±13.2950 – 9513.33±6.4210 – 30p1<0.001p2<0.001p3<0.001p4=0.02
Η scoreX±SDRange168.16±93.3710 – 270240.0±39.87150 – 28518.33±16.45 10 – 60p1<0.001p2<0.001p3<0.001p4=0.01
Tumour islands
ExpressionPositiveNegative713356520010001829010p1<0.001p2=0.49p3=0.06p4<0.001
DistributionPatchyDiffuse701000614307014477.822.2p1=0.29p2=0.003p3=0.04p4=0.002
PatternCytoplasmicNuclearNucleo– cytoplasmic00 700100051502575601233.3066.7p1=0.14p2=0.008p3=0.003p4=0.55
LocalizationCentral PeripheralCentral and peripheral61085.714.30130765035-----------p4=0.006
PercentX±SDRange18.57±4.7610 – 2561.5±22.0720 – 9013.33±6.4210 – 30p1=0.02p2<0.001 p3<0.001p4<0.001
Η scoreX±SDRange52.86±15.2430 – 75184.5±66.2160 – 27018.33±16.4510 – 60p1=0.001p2<0.001p3<0.001p4<0.001
Stroma
ExpressionPositiveNegative200100020010000200100p1<0.001p2<0.001p3<0.001p4=NA
TypeInflammatory AdnexaAdnexa and inflammatory2513102565140670030000000p1<0.001p2<0.001p3<0.001p4=0.49

(a) HIF-1α in normal skin in the basal and suprabasal layers of the epidermis (red arrow) (Immunoperoxidase X40); (b) higher power showing nuclear patchy expression of HIF-1α (red circles) in epidermis with negative dermal immunoreactivity (Immunoperoxidase X400).

Comparison between the studied groups regarding GLUT-1 expression.

VariableBCC n = 20SCC N=20Normal skin N=20p-value
No%No%No%
Overlying epidermis
ExpressionPositiveNegative200100020010002001000.0p1=NAp2=NAp3=NAp4=0.41
DistributionPatchyDiffuse12666.733.39660404162080p1=0.004p2=0.01p3=0.02p4=0.69
PatternCytoplasmicMembranousMembrano- cytoplasmic0162088,911.1012308020401620080p1<0.001p2<0.001p3<0.001p4=0.49
LocalizationBasalAll layers81044.455.68753.346.70200100p1=0.001p2<0.001p3<0.001p4=0.61
PercentX±SDRange35.56±9.8310 - 5042.0±16.5620 - 9058.75±19.5920 - 85p1<0.001p2=0.01p3<0.001p4=0.3
H scoreX±SDRange90.55±31.3410 - 150109.33±48.9160 - 240133.25±78.9120 - 215p1=0.09p=20.41p3=0.13p4=0.3
Tumour islands
ExpressionPositiveNegative14670302001000.02001000.0p1=0.02p2=NAp3=0.17p4=0.02
DistributionPatchyDiffuse1401000.021810904162080p1<0.001p2=0.66p3=0.04p4<0.001
Pattern CytoplasmicMembranousMembrano- cytoplasmic068042.957.1017308515401620080p1=0.003p2<0.001p3<0.001p4=0.01
LocalizationCentral PeripheralCentral and peripheral121185.77.17.1017308515----------p4<0.001
PercentX±SDRange29.28±9.9710 - 4053.0±12.1830 - 7058.75±19.5920 - 85p1<0.001p2=0.15p3=0.004p4<0.001
H scoreX±SDRange82.14±34.4610 - 120144.0±45.060 - 210133.25±78.9120 - 215p1=0.09p2=0.98p3=0.39p4<0.001
Stroma
ExpressionPositiveNegative200100020010001829010p1=0.49p2=0.49p3=0.04p4=NA
TypeInflammatoryAdnexaAdnexa and inflammatory1901950517128551018009000p1=1.0p2=0.23p3=0.38p4=0.49

GLUT-1 expression in normal skin showing a) Membranocytoplasmic patchy immunoreactivity distributed in the basal and suprabasal epidermis (Immunoperoxidase 20X). b) Membranocytoplasmic GLUT-1 is diffusely distributed in all epidermal layers (Immunoperoxidase 20X).

a) Adenoid BCC negatively expressed HIF-1α in tumour nests with positive expression in inflammatory infiltrates and the intervening stroma (red arrows) (Immunoperoxidase 4X); b) Cytoplasmic and nucleocytoplasmic expression of HIF-1α in the overlying epidermis (black arrow) of BCC (red arrow). Tumour nests show positive patchy nucleocytoplasmic expression (Immunoperoxidase 10X); c) HIF-1α expression in a keratotic BCC showing cytoplasmic immunoreactivity in the basaloid cells (red circle) and in the foci of keratotic differentiation (red arrows), while sparing the vast majority of the tumour nests (Immunoperoxidase 10X); d) Higher power showing the cytoplasmic localization of HIF-1α in the area of keratotic differentiation (Immunoperoxidase 40X).

a) GLUT-1 expression in nodular BCC mainly occupies the central part of each nodule (red circles) (immunoperoxidase 4X);b) Membranous GLUT-1 expression confined to the basal layer of epidermis (black arrow) overlying nodular BCC (red arrow). |iInflammatory cells show positive immunoreactivity (immunoperoxidase 10X);c) GLUT-1 expression in keratotic BCC, mainly at the central part around areas of keratotic differentiation of each tumour island (Immunoperoxidase 20X);d) Higher power showing membranous GLUT-1 around keratotic differentiation while sparing the basaloid cells (Immunoperoxidase 40X).

a) HIF-1α expression in well differentiated SCC showing diffuse, central and peripheral immunoreactivity (red arrows). Inflammatory and stromal cells show positive expression (Immunoperoxidase 4X); b) Higher power showing nuclear (red circles) and cytoplasmic (red arrows) expression of HIF-1α in well differentiated nests of SCC (Immunoperoxidase 20X); c) Nuclear (red circles) and nucleocytoplasmic (red arrow) expression of HIF-1α in moderately differentiated SCC (Immunoperoxidase 4X); d) Nuclear (red circle) and cytoplasmic (red arrow) expression of HIF-1α in the nest of poorly differentiated SCC (Immunoperoxidase 20X).

a) Confinement of GLUT-1 to the periphery of tumour islands in well differentiated SCC (Immunoperoxidase 4X); b) GLUT-1 in moderately differentiated SCC; shows membranocytoplasmic (black arrows) immunoreactivity confined to the periphery of the tumour islands sparing the centre (Immunoperoxidase 40X); c) Membranous GLUT-1 in poorly differentiated SCC (Immunoperoxidase 4X); d) Poorly differentiated SCC exhibiting membranous GLUT-1 occupying the periphery (blue arrow) or centre and periphery of the tumour islands (Immunoperoxidase 4X).

(a,b) significant association between higher H score and percent values of HIF-1α and grade I SCC; (c,d) Negative correlation between H score and percent values of HIF-1α and tumour grade.

Comparison between studied groups with regard to HIF-1α and GLUT-1 Expression

Basal Cell Carcinoma cases versus Normal skin: Comparing the epidermis overlying BCC and normal epidermis demonstrated that higher expression percentage, higher H score value (p<0.001 for both), nuclear and nucleocytoplasmic immunoreactivity (p=0.01) of HIF-1α were significantly associated with BCC. Comparing tumour islands and normal epidermis showed that higher H score (p=0.001) and higher expression percentage (p=0.02) of HIF-1α were significantly associated with BCC [Table/Fig-2].

Regarding GLUT-1 immunoreactivity, the overlying epidermis of normal skin showed more diffuse GLUT-1 expression (p=0.004) with basal and suprabasal localization (p=0.001). Membrano-cytoplasmic pattern was significantly associated with normal skin (p<0.001). Comparing tumour islands and normal epidermis showed that GLUT-1 expression (p=0.02) and expression percentage (p<0.001) were significantly higher in normal epidermis. Membrano-cytoplasmic pattern was significantly associated with normal skin (p<0.001) [Table/Fig-4].

Squamous cell carcinoma cases versus normal skin: Higher expression percentage of HIF-1α, higher H score value (p<0.001 for both), nucleocytoplasmic immunoreactivity (p=0.02), and diffuse distribution (p=0.002) were significantly associated with the overlying epidermis in SCC cases. Comparing HIF-1α immunoreactivity in tumour islands versus normal epidermis showed that higher expression percentage, higher H score value (p<0.001 for both), diffuse distribution (p=0.003) and nucleocytoplasmic immunoreactivity (p=0.008) were significantly associated with SCC [Table/Fig-2].

In overlying epidermis, GLUT-1 expression percentage (p=0.01), membrano-cytoplasmic pattern, basal and suprabasal immunoreactivity (p<0.001 for both) were significantly associated with normal skin. Comparing tumour islands and normal epidermis showed that membranous pattern was significantly associated with SCC cases (p<0.001) [Table/Fig-4].

Basal cell carcinoma versus squamous cell carcinoma: In overlying epidermis, higher expression percentage of HIF-1α (p=0.02) and higher H score value (p=0.01) were significantly associated with SCC. In tumour islands, higher expression, higher expression percentage, higher H score value (p<0.001 for all), central and peripheral immunoreactivity (p=0.006) and diffuse staining (p=0.002) were significantly associated with SCC [Table/Fig-2].

Regarding GLUT-1 immunostaining in overlying epidermis, there was no significant difference in any of the studied parameters. In tumour islands, GLUT-1 expression (p= 0.02), higher expression percentage, higher H score, peripheral immunoreactivity and diffuse staining (p<0.001 for all) were significantly associated with SCC compared with normal skin [Table/Fig-4].

NMSC cases (BCC+SCC) versus normal skin: In overlying epidermis, HIF-1α immunostaining showed that higher expression percentage (p=0.02), higher H score (p=0.01), nucleocytoplasmic pattern (p=0.001) and diffuse staining (p=0.009) were significantly associated with NMSC cases. Comparing HIF-1α immunoreactivity in tumour islands versus normal epidermis showed that higher expression percentage, higher H score (p<0.001 for both), nucleocytoplasmic pattern (p=0.003) and diffuse staining (p=0.04) were significantly associated with NMSC cases [Table/Fig-2].

GLUT-1 expression percentage (p<0.001) in the overlying epidermis was down regulated in NMSC cases compared with normal skin. Membranous pattern and basal and suprabasal immunoreactivity (p<0.001 for both) were significantly associated with normal skin. Comparing tumour islands and normal epidermis showed that, expression percentage (p=0.004) was down regulated in NMSC cases. Membranous pattern was significantly associated with NMSC cases (p<0.001) [Table/Fig-4].

Relationship between expression percentage and H score values of HIF-1α and GLUT-1 and clinicopathologic parameters of studied cases:

Basal cell carcinoma: No significant association was detected between H score and expression percentage values of HIF-1α and GLUT-1 and clinical (age of cases, gender, disease duration, lesion size, lesion site) or pathological (histopathological type, involvement of surgical margin) parameters of studied cases. No significant correlation was detected between HIF-1α and GLUT-1 expression percentage or H score values and age of cases or size of tumour in studied cases whether in overlying epidermis or in tumour islands.

Squamous cell carcinoma: Higher H score values and expression percentage were significantly associated with Grade I tumours (p=0.02 for both) [Table/Fig-10a,b]. Significant negative correlation was detected between H score values (r=-0.6, p=0.003) and expression percentage (r=-0.5, p=0.01) and tumour grade [Table/Fig-10c,d].

Regarding GLUT-1 immunostaing, higher expression percentage and H score values were significantly associated with lesions on extremities (p=0.03 for both). No significant correlation was found between expression percentage or H score values and both age of patients, size and grade of the tumour whether in overlying epidermis or in tumour islands (data not shown in tables or figures).

Relationship between pattern of HIF-1α and GLUT-1 and clinicopathologic parameters of studied cases: No significant association was demonstrated between their patterns of expression and clinical or histopathological characters of studied BCC and SCC cases.

Correlation between H score and percent values of GLUT-1 and HIF-1α in studied cases: No significant correlation was found between H score or expression percentage values of both of them in tumour islands or in overlying epidermis in BCC and SCC.

Discussion

HIF-1α expression: In the current study, HIF-1α was expressed in basal and suprabasal layers in 90% of normal skin biopsies. To the best of our knowledge, HIF-1α expression has not been studied before in normal skin. Its expression in normal tissues showed controversial findings. Some previous studies reported absent expression of HIF-1α in normal tissues including prostate [19-21] breast [22,23] and kidney [24,25]. Giatromanolaki A et al., reported weak or negative reactivity of normal lung tissue and areas distal to malignant lung neoplasm [26] and Yoshimura H et al., reported weak normal colorectal mucosal cell staining of HIF-1α [27].

The current finding can be explained by the need of epidermal HIF-1 to help in cellular adaptation to hypoxia. This is because the epidermis is avascular while dermal oxygen demand is satisfied by dermal vasculature [28].

Going with our finding, Bedogni et al., reported that human and mouse skin includes areas with low oxygen content, demonstrated in their study by positive carbonic anhydrase IX and 2-nitroimidazole EF5 immunoreactivity which correlate with GLUT-1 [29]. These markers correlate with GLUT-1 staining, a well known HIF-1α direct target gene.

In addition, HIF-1α has a key biological role in defense against invasive microorganisms, since Peyssonnaux1 C et al., demonstrated its role in supporting inducible cathelidin production and its consequent antibacterial activity through in vitro studies in cultured human keratinocyte cell lines [28].

GLUT-1 expression: GLUT-1 was expressed in all normal skin biopsies with basal and suprabasal localization in all examined sections. Our results are concordant with Parente E et al., [30]. Gherzi R et al., [31] detected GLUT-1 expression in the basal layer and to a lower extent in the immediately suprabasal layer of the epidermis with decreased immunoreactivity in upper epidermal layers. Authors postulated that GLUT-1 immunoreactivity gradually declines with cell cornification while Voldstedlund and Dabelsteen came to a conclusion that epidermal GLUT-1 immunoreactivity is mainly due to glucose consumption by keratinocytes which depends on glycolysis [32]. On the other hand, Yuan SM et al., reported complete absence of GLUT-1immunoreactivity in normal skin [33]. Therefore, the expression of GLUT-1 in proliferating and differentiating cells is a matter of debate for further investigation.

The prevalence of membranocytoplasmic expression of GLUT-1 in the normal epidermis, in the current study, could be attributed to the series of changes in GLUT-1 upon exposure to hypoxic conditions as has been reviewed by Zhang J et al., [34]. The sequence of events begin with unmasking of GLUT-1 with increase in glucose binding followed by additional stimulation with hypoxia, which causes protein translocation from cytoplasm to cell membrane with increase in GLUT-1 mRNA synthesis [35]. Therefore, membranous immunoreactivity was considered as an indicator of hypoxia according to earlier reports [36-38].

In the current study, HIF-1α was up regulated in NMSC cases compared with normal skin. This may be due to excessive tumour proliferation that is adapted to hypoxia by activation of anaerobic metabolism and induction of angiogenesis [39]. Furthermore, with low oxygen content, HIF-1α interacts with p53 causing an increase in p53 half-life and a decrease in HIF-1α half-life [40]. The HIF-1α mediated stabilization of p53 may contribute to hypoxia mediated apoptosis and p53 loss of function. HIF-1α also increases the expression of VEGF and thus promotes angiogenisis [41].

HIF-1α was not investigated before in NMSC. However, going with our findings, Kurokawa T et al., [42] detected HIF-1α up regulation in oral SCC. HIF-1α was also found to be overexpressed in carcinomas of different organs including nasopharyngeal carcinoma [43] ovarian carcinoma [44] bladder adenocarcinoma [45], gastric carcinoma [46], pancreatic carcinoma [47] renal cell carcinoma [25] and hepatocellular carcinoma [48].

GLUT-1 was down regulated in NMSC cases compared with normal skin. Our results are on the contrary to previous reports that stated overexpression of GLUT-1 in various human tumours [49,50]. However, going with our findings, Parente et al., reported that GLUT-1 is down regulated in 55% of malignant melanomas compared to melanocytic and Spitz nevi [30]. Hauptmann S et al., showed that GLUT-1 in colorectal adenocarcinoma cell lines is inversely correlated with tumour cell proliferation [51]. Therefore, we can postulate that, although GLUT-1 is a proproliferative, it could be down regulated in hyperproliferative disorders in an attempt to compensate for the tumour growth and/or there might be factors other than hypoxia and ischemia regulating GLUT-1 expression.

The current work showed significant difference between SCCs and BCCs regarding HIF-1α and GLUT-1 expression, since higher expression percentage and H score values of both GLUT-1 and HIF-1α, were in favor of SCC. Therefore, the high proliferative index, the infiltrative and rapidly destructive nature of SCC may contribute to an evident hypoxic environment, in comparison to BCC, which is an important regulator of both HIF-1α and GLUT-1.

The demonstrated nucleocytoplasmic immunoreactivity of HIF-1α in 100% and 85% of our BCC and SCC cases respectively and the nuclear pattern in 15% of SCC cases may be due to the cytoplasmic-nuclear trafficking of the HIF-1α and its protein fusion between normal and tumour cells. Under normal oxygen tension, von Hippel Lindau protein (pVHL) is engaged in a constitutive shuttling between nucleus and cytoplasm. The subcellular localization of HIF-1α depends on pVHL. However, it has not been fully established whether proteasomal degradation of HIF-1α occurs in the nucleus, in the cytoplasm, or in both compartments. Prior reports are somewhat contradictory [52].

Di Cristofano C et al., detected strong nuclear localization of HIF-1α in clear renal cell carcinoma with reduced pVHL trafficking [53]. The same was detected by Bos R et al., in ductal carcinoma in situ of the breast [22] and by Kurokawa T et al., in oral SCC [42]. In these studies, nuclear staining was coupled with increased degree of malignancy and short survival. In the current work, no significant association was demonstrated between nuclear immunoreactivity and tumour grade, tumour stage or lymph node involvement in SCC cases.

GLUT-1 showed membranous and membrano-cytoplasmic patterns of immunoreactivity in BCC and SCC tumour islands, included in the current work. Ariely R et al., suggested that co-localization of GLUT-1 with the Golgi leads to combined membrane and cytoplasmic expression [54]. Baer SC et al., and Ayala FR et al., demonstrated membranous expression in cutaneous and oral SCCs respectively [49,50]. Takanaka H and Frommer WB concluded that GLUTs mediate endoplasmic reticulum glucose transport en route to the plasma membrane [55].

The positive HIF-1α expression in inflammatory cells in BCC and SCC cases, demonstrated in the current work, can be explained by the effect of HIF-1α on T cell differentiation that was demonstrated by Dang EV et al., [56]. In addition, HIF-1α has role in host immune response through recognition of Pathogen-Associated Molecular Patterns (PAMPs) such as Lipopolysaccharides (LPS) by Toll Like Receptors (TLRs) in human monocytes [57].

The positive stromal immunoreactivity for GLUT-1 and HIF-1α in BCC and SCC cases was not reported previously. However, Pinkus H mentioned that, as the connective tissue stroma proliferates with the tumour and is arranged in parallel bundles around the tumour masses, a mutual relationship seems to exist between the parenchyma of the tumour and its stroma [58]. In addition, the expansion and growth of tumour masses may occur on the expense of stromal cells which may suffer from hypoxia as well.

In the present work, adnexal structures in BCC and SCC showed positive HIF-1α and GLUT-1 immunoreactivity. The significance of this finding is not clear. However, Parente P et al., reported GLUT-1 expression in the eccrine ducts and the perineurium of the small nerve trunks of normal skin, nevi and malignant melanoma [30]. Voldstedlund M and Dabelsteen E showed GLUT-1 expression in ductal and myoepithelial cells of minor salivary glands and perineural sheath located in the lamina propria of oral mucosa [32]. Further studies are needed to clarify its significance.

A significant association between higher H score values of HIF-1α expression and Grade I in SCC cases was detected in our results. Significant negative correlation was also noted between H scores and tumour grade. Similarly, an inverse correlation was shown with tumour differentiation in hepatocellular carcinoma [59] and ovarian carcinoma cases [60]. Nevertheless our finding may be considered as a good prognostic sign as postulated by Fillies J et al., [61] On the contrary, Korkolopoulou P et al., found that HIF-1α protein expression is increased significantly with increasing grade and proliferative potential in astrocytomas [62].

The current study demonstrated the presence of GLUT-1 mainly in the central portion of malignant basaloid nests. A similar finding was reported by Youssef KK et al., who suggested that this may be related to focal keratinization in BCC as an indicator of follicular differentiation [63]. In addition, Oliver RJ et al., suggested that GLUT-1 staining in central tumour nests, distal to blood vessels, may signify the existence of hypoxia-induced GLUT-1 [64].

The presence of GLUT-1 mainly in the peripheral part of tumour nests in SCC is similar to Oliver RJ et al., who demonstrated that, GLUT-1 staining occurs in outer epithelial layers as a function of the invasion process [64]. Ayala FR et al., demonstrated that, GLUT-1 expression was observed in the majority of poorly differentiated cases of oral SCC in central and perinecrotic zones, while well-differentiated SCC showed absent GLUT-1 staining in the central zones of the tumour, with its expression localized in the peripheral neoplastic area [50].

The positive HIF-1α mainly in central parts of BCC and SCC tumour islands went with the report of Fillis T et al., who showed that HIF-1α expression is usually diffuse and tended to be more prominent towards the centre of tumour fields [61].

The significantly higher H score values of GLUT-1 in SCC extremities’ lesions was not reported previously but can be explained by the earlier observation that sun exposure induces an up regulation of GLUT-1 gene expression [65].

Absence of a significant correlation between HIF-1α and GLUT-1 in both BCC and SCC has been demonstrated by Koukourakis MI et al., in colorectal carcinoma [66] and Schrijvers ML et al., in laryngeal carcinoma [67].

limitation

Results of this study may not be completely generalizable because the small sample size.

Conclusion

In conclusion, HIF-1α may have a role in NMSC pathogenesis through adaptation to hypoxia which results from excessive proliferation. GLUT-1 down regulation in NMSC may be explained by its consumption by proliferating tumour cells. The expression of HIF-1α and GLUT-1 in normal skin requires further investigation. The positive stromal and adnexal immunoreactivity to both HIF-1α and GLUT-1 and the up regulation of GLUT-1 in peripheral SCC lesions also needs further research.

References

[1]Diepgen TL, Mahler V, The epidemiology of skin cancer Br J Dermatol 2002 146:01-06.  [Google Scholar]

[2]Helal T, Salman M, Ezz Elarab S, Egyptian Pathology –Based Cancer Registry 2001-2010 Ain Shams Faculty of Medicine, Cairo 2015 Chapter 11:95-96.  [Google Scholar]

[3]Shay JE, Celeste simon M, Hypoxia-inducible factors: crosstalk between inflammation and metabolism Semin Cell Dev Biol 2012 23:389-94.  [Google Scholar]

[4]Vaupel P, Tumour microenvironmental physiology and its implications for radiation oncology Semin Radiat Oncol 2004 14:198-206.  [Google Scholar]

[5]Loor G, Schumacker PT, Role of hypoxia-inducible factor in cell survival during myocardial ischemia-reperfusion Cell Death Differ 2008 15:686-90.  [Google Scholar]

[6]Dondajewska E, Suchorska W, Hypoxia-inducible factor as a transcriptional factor regulating gene expression in cancer cells Wspolczesna Onkol 2011 15:234-39.  [Google Scholar]

[7]Hallis AL, Hypoxia, a key regulatory factor in tumour growth Nat Rev Cancer 2002 2:38-47.  [Google Scholar]

[8]Semenza GL, HIF-1 and tumour progression: Pathophysiology and therapeutics Trends Mol Med 2002 8:S62-S67.  [Google Scholar]

[9]Gatenby RA, Smallbone K, Maini PK, Rose F, Averill J, Nagle RB, Cellular adaptations to hypoxia and acidosis during somatic evolution of breast cancer Br J Cancer 2007 64:653-56.  [Google Scholar]

[10]Montel-hagen A, Kinet S, Manel N, Mongellaz C, Prohaska R, Battini JL, Erythrocyte glut1 triggers dehydroascorbic acid uptake in mammals unable to synthesize vitamin C Cell 2008 132:1039-48.  [Google Scholar]

[11]Isselbacher KJ, Sugar and amino acid transport by cells in culture-differences between normal and malignant cells N Engl J Med 2010 286:929-33.  [Google Scholar]

[12]Broders AC, The grading of carcinoma Minn Med 1925 8:726-30.  [Google Scholar]

[13]Sobin LH, Wittekind C, TNM classification of malignant tumours 2002 New YorkWiley-Liss  [Google Scholar]

[14]Zhong H, De Marzo AM, Laughner E, Overexpression of hypoxia inducible factor l alpha in common human cancers and their metastases Cancer Res 1999 59:5830-35.  [Google Scholar]

[15]Monaco SE, Dabbs DJ, Immunocytology Diagnostic Immunohistochemistry: The ranostic and Genomic Applications 2014 4th ed:829-53.chapter 21  [Google Scholar]

[16]Tsukioka M, Matsumoto Y, Noriyuki M, Yoshida C, Nobeyama H, Yoshida H, Expression of glucose transporters in epithelial ovarian carcinoma: correlation with clinical characteristics and tumour angiogenesis Oncol Rep 2007 18:361  [Google Scholar]

[17]Bahnassy AA, Zekri AR, El-Houssini S, El-Shehaby AM, Mahmoud MR, Abdallah S, Cyclin A and cyclin D1 as significant prognostic markers in colorectal cancer patients BMC Gastroenterol 2004 23:4-22.  [Google Scholar]

[18]Bilalovic N, Sandstad B, Golouh R, Nesland JM, Selak I, Torlakovic EE, CD10 protein expression in tumour and stromal cells of malignant melanoma is associated with tumour progression Mod Pathol 2004 17:1251-58.  [Google Scholar]

[19]Hao P, Chen X, Geng H, Gu L, Chen J, Lu G, Expression and implication of hypoxia inducible factor-1alpha in prostate neoplasm J Huazhong Univ Sci Technolog Med Sci 2004 24:593-95.  [Google Scholar]

[20]Zhong H, Semenza GL, Simons JW, De Marzo AM, Upregulation of hypoxia-inducible factor 1alpha is an early event in prostate carcinogenesis Cancer Detect Prev 2004 28:88-93.  [Google Scholar]

[21]Wang L, Chen ZJ, Wang QT, Cao WF, Jian Y, Wang SX, Expression of hypoxia-inducible factor lalpha and vascular endothelial growth factor in prostate cancer and its significance Zhonghua Nan Ke Xue 2006 12:57-59.  [Google Scholar]

[22]Bos R, Zhong H, Hanrahan CF, Mommers EC, Semenza GL, Pinedo HM, Levels of hypoxia-inducible factor-1 alpha during breast carcinogenesis J Natl Cancer Inst 2001 93:309-14.  [Google Scholar]

[23]Okada K, Osaki M, Araki K, Ishiguro K, Ito H, Ohgi S, Expression of hypoxia-inducible factor (HIF-1alpha), VEGF-C and VEGF-D in non-invasive and invasive breast ductal carcinomas Anticancer Res 2005 25:3003-09.  [Google Scholar]

[24]Shao ZQ, Zheng SB, Xiao YJ, Tan WL, Chen T, Qi H, Expressions of hypoxia inducible factor-1alpha and vascular endothelial growth factor in human renal cell carcinoma Di Yi Jun Yi Da Xue Xue Bao 2005 25:1034-36.  [Google Scholar]

[25]Zhang N, Gong K, Yang XY, Xin DQ, Na YQ, Expression of hypoxia-inducible factor-1-alpha, hypoxia-inducible factor-2 alpha and vascular endothelial growth factor in sporadic clear cell renal carcinoma and their significance in the pathogenesis thereof Zhonghua Yi Xue Za Zhi 2006 86:1526-29.  [Google Scholar]

[26]Giatromanolaki A, Koukourakis M, Sivridis E, Turley H, Talks K, Pezzella F, Relation of hypoxia inducible factor 1a and 2a in operable non-small cell lung cancer to angiogenic molecular profile of tumours and survival Br J Cancer 2001 85:881-90.  [Google Scholar]

[27]Yoshimura H, Dhar DK, Kohno H, Kubota H, Fujii T, Ueda S, Prognostic impact of hypoxia-inducible factors 1alpha and 2alpha in colorectal cancer patients: correlation with tumour angiogenesis and cyclooxygenase-2 expression Clin Cancer Res 2004 10:8554-60.  [Google Scholar]

[28]Peyssonnaux1 C, Boutin1 AT, Zinkernagel AS, Datta V, Nizet V, Johnson RS, Critical role of HIF-1a in keratinocyte defense against bacterial infection J Invest Dermatol 2008 128:1964-68.  [Google Scholar]

[29]Bedogni B, Welford SM, Cassarino DS, Nickoloff BJ, Giaccia AJ, Powell MB, The hypoxic microenvironment of the skin contributes to Akt-mediated melanocyte transformation Cancer cell 2005 8:443-54.  [Google Scholar]

[30]Parente P, Coli A, Massi G, Mangoni A, Fabrizi MM, Bigotti G, Immunohistochemical expression of the glucose transporters Glut-1 and Glut-3 in human malignant melanomas and benign melanocytic lesions J Exp Clin Cancer Res 2008 2:27-34.  [Google Scholar]

[31]Gherzi R, Melioli G, de Luca M, D’Agostino A, Distefano G, Guastella M, HepG2/erythroid/ brain” type glucose transporter (GLUT1) is highly expressed in human epidermis: keratinocyte differentiation affects GLUT1 levels in reconstituted epidermis J Cell Physiol 1992 150:463-74.  [Google Scholar]

[32]Voldstedlund M, Dabelsteen E, Expression of GLUT1 in stratified squamous epithelia and oral carcinoma from humans and rats APMIS 1997 105:537-45.  [Google Scholar]

[33]Yuan SM, Jiang HQ, Hong ZJ, Wang J, Hu XB, Ouyang TX, The expression and role of glucose transporter-1 in infantile hemangioma Chinese 2007 23:90-93.  [Google Scholar]

[34]Zhang J, Behrooz A, Ismail-Begi F, Regulation of glucose transport by hypoxia Am J Kidney Dis 1999 34:189-202.  [Google Scholar]

[35]Wertheimer E, Sasson S, Cerasi E, Ben-Neriah Y, The ubiquitous glucose transporter Glut-1 belongs to the glucose-regulated protein family of stress-inducible proteins Proc Natl Acad Sci USA 1991 88:2525-29.  [Google Scholar]

[36]Wincewicz A, Sulkowska M, Koda M, Sulkowski S, Clinicopathological significance and linkage of the distribution of HIF-1alpha and GLUT-1 in human primary colorectal cancer Pathol Oncol Res 2007 13:15-20.  [Google Scholar]

[37]Airley RE, Loncaster J, Raleigh JA, Harris AL, Davidson SE, Hunter RD, GLUT-1 and CAIX as intrinsic markers of hypoxia in carcinoma of the cervix: relationship to pimonidazole binding Int J Cancer 2003 104:85-91.  [Google Scholar]

[38]Elson DA, Ryan HE, Snow JW, Johnson R, Arbeit JM, Coordinate up-regulation of hypoxia inducible factor (HIF)-1αand HIF-1 target genes during multi-stage epidermal carcinogenesis and wound healing Cancer Res 2000 60:6189-95.  [Google Scholar]

[39]Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, Hypoxia-inducible factor-1αis a positive factor in solid tumour growth Cancer Res 2000 60:4010-15.  [Google Scholar]

[40]An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM, Stabilization of wild type p53 by hypoxia-inducible factor 1alpha Nature 1998 392:405-08.  [Google Scholar]

[41]Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Regulation of tumour angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha Genes Dev 2000 14:34-44.  [Google Scholar]

[42]Kurokawa T, Miyamoto M, Kato K, Cho Y, Kawarada Y, Hida Y, Overexpression of hypoxia-inducible-factor 1 alpha (HIF-1alpha) in oesophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage Br J Cancer 2003 8:1042-47.  [Google Scholar]

[43]Hui EP, Chan AT, Pezzella F, Turley H, To KF, Poon TC, Coexpression of hypoxia-inducible factors 1alpha and2alpha, carbonic anhydrase IX, and vascular endothelial growth factor in nasopharyngeal carcinoma and relationship to survival Clin Cancer Res 2002 8:2595-604.  [Google Scholar]

[44]Wong C, Wellman TL, Lounsbury KM, VEGF and HIF- 1alpha expression are increased in advanced stages of epithelial ovarian cancer Gynecol Oncol 2003 91:513-17.  [Google Scholar]

[45]Deng JH, Bai JL, Ma PC, Zhang SS, Wan JH, Expression and significance of GST-Pi and HIF-1alpha in bladder carcinoma Ai Zheng 2006 25:190-93.  [Google Scholar]

[46]Urano N, Fujiwara Y, Doki Y, Tsujie M, Yamamoto H, Miyata H, Overexpression of hypoxia-inducible factor-1 alpha in gastric adenocarcinoma Gastric Cancer 2006 9:44-49.  [Google Scholar]

[47]Couvelard A, O’Toole D, Leek R, Turley H, Sauvanet A, Degott C, Expression of hypoxia-inducible factors is correlated with the presence of a fibrotic focus and angiogenesis in pancreatic ductal adenocarcinomas Histopathol 2005 46:668-76.  [Google Scholar]

[48]Huang GW, Yang LY, Lu WQ, Expression of hypoxia inducible factor 1alpha and vascular endothelial growth factor in hepatocellular carcinoma: Impact on neovascularization and survival World J Gastroenterol 2005 11:1705-08.  [Google Scholar]

[49]Baer SC, Casaubon L, Younes M, Expression of the human erythrocyte glucose transporter Glut1 in cutaneous neoplasia J Am Acad Dermatol 1997 37:575-77.  [Google Scholar]

[50]Ayala FR, Rocha RM, Carvalho KC, Carvalho AL, da Cunha IW, Lourenço SV, GLUT1 and GLUT3 as potential prognostic markers for oral squamous cell carcinoma Molecule 2010 15:2374-87.  [Google Scholar]

[51]Hauptmann S, Grünewald V, Molls D, Schmitt WD, Köbel M, Kriese K, Glucose transporter GLUT1 in colorectal adenocarcinoma cell lines is inversely correlated with tumour cell proliferation Anticancer Res 2005 25:3431-36.  [Google Scholar]

[52]Moroz E, Carlin S, Dyomina1 K, Burke S, Thaler HT, Blasberg R, Real-Time Imaging of HIF-1a Stabilization and Degradation PLoS ONE 2009 4:5077-85.  [Google Scholar]

[53]Di Cristofano C, Minervini A, Menicagli M, Salinitri G, Bertacca G, Pefanis G, Nuclear expression of hypoxia-inducible factor-1alpha in clear cell renal cell carcinoma is involved in tumour progression Am J Surg Pathol 2007 31:1875-81.  [Google Scholar]

[54]Airley R, Loncaster J, Davidson S, Bromley M, Roberts S, Patterson A, Glucose transporter Glut-1 expression correlates with tumour hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix Clin Cancer Res 2001 7:928-34.  [Google Scholar]

[55]Takanaga H, Frommer WB, Facilitative plasma membrane transporters function during ER transit FASEB J 2010 24:2849-58.  [Google Scholar]

[56]Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Control of T(H)17/ T(reg) balance by hypoxia-inducible factor 1 Cell 2011 146:772-84.  [Google Scholar]

[57]Frede S, Stockmann C, Freitag P, Fandrey J, Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-kappa B Biochem J 2006 396:517-27.  [Google Scholar]

[58]Pinkus H, Epithelial and fibroepithelial tumours Arch Dermatol 1965 91:24-37.  [Google Scholar]

[59]Ding L, Chen XP, Wang HP, Expression and clinical significance of HIF-1a protein in hepatocellular carcinoma tissues Zhonghua Gan Zang Bing Za Zhi 2004 12:656-59.  [Google Scholar]

[60]Nakayama K, Kanzaki A, Hata K, Katabuchi H, Okamura H, Miyazaki K, Hypoxia-inducible factor 1 alpha (HIF-1 alpha) gene expression in human ovarian carcinoma Cancer Lett 2002 176:215-23.  [Google Scholar]

[61]Fillies T, Werkmeister R, van Diest PJ, Brandt B, Joos U, Buerger H, HIF1-alpha overexpression indicates a good prognosis in early stage squamous cell carcinomas of the oral floor BMC Cancer 2005 5:84-87.  [Google Scholar]

[62]Korkolopoulou P, Patsouris E, Konstantinidou AE, Pavlopoulos PM, Kavantzas N, Boviatsis E, Hypoxia-inducible factor 1alpha/vascular endothelial growth factor axis in astrocytomas. Associations with microvessel morphometry, proliferation and prognosis Neuropathol Appl Neurobiol 2004 30:267-78.  [Google Scholar]

[63]Youssef KK, Van Keymeulen A, Lapouge G, Beck B, Michaux C, Achouri Y, Identification of the cell lineage at the origin of basal cell carcinoma Nat Cell Biol 2010 12:299-305.  [Google Scholar]

[64]Oliver RJ, Woodwards RTM, Sloan P, Thakker NS, Stratford IJ, Airley RE, Prognostic value of facilitative glucose transporter Glut-1 in oral squamous cell carcinomas treated by surgical resection: results of EORTC Translation Research Eur J Cancer 2004 40:503-07.  [Google Scholar]

[65]Steinberg ML, Hubbard K, Utti C, Clas B, Hwang BJ, Hill HZ, Patterns of persistent DNA damage associated with sun exposure and the glutathione S-transferase M1 genotype in melanoma patients Photochem Photobiol 2009 85:397-86.  [Google Scholar]

[66]Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E, Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumour-associated stroma Cancer Res 2006 66:632-37.  [Google Scholar]

[67]Schrijvers ML, van der Laan BF, de bock GH, Pattje WJ, Mastik MF, Menkema L, Overexpression of intrinsic hypoxia markers HIF1αand CA-IX predict for local recurrence in stage t1-t2 glottic laryngeal carcinoma treated with radiotherapy Int J radiat oncolo Biol Phys 2008 72:161-69.  [Google Scholar]