Monthly Archives: April 2017

Activation of STAT3 and its impacts on Proteasome Inhibitor-Induced Apoptosis in Laryngeal Sqaumous Carcinoma

DOI: 10.31038/CST.2017215

Abstract

Laryngeal squamous carcinoma (LSC) represents one of the refractory malignant tumors of head and neck squamous cell carcinoma (HNSCC), leading to high patient mortality and unsatisfactory overall 5-year survival. New therapeutic targets and modalities for effective chemoprevention and treatment are required to improve the survival of patients with LSC. Evidence from previous studies has suggested that proteasome inhibitors (PIs) possess effective antitumor activity and have been approved by the United States Food and Drug Administration (FDA) to treat myeloma. However, resistance of myeloma to PIs has been reported, which prevents PIs from their clinical uses in cancer treatment. So far, the underlying mechanisms of tumor cells anti-PIs effects remain unclear. In the present study, we found that constitutive activation of signal transducer and activator of transcription 3 (STAT3) was associated with clinicopathlologic factors in LSC patients, and persistent activation of STAT3 was found in Hep-2 laryngeal carcinoma cells. Moreover, we demonstrated that proteasome inhibitor MG-132 induces apoptosis and cell cycle arrest in Hep-2 cells, accompanied by increased activation of STAT3; knockdown of STAT3 expression by shRNA or application of STAT3 inhibitor AG490 potentiates the antitumor effects of MG-132 in vitro, which is attributed to the down-regulation of cyclin D1 and Bcl-2. Taken together, we identified that STAT3 activation plays a great role in therapeutic resistance to PIs in LSC; blocking STAT3 activation could potentiate the tumor killing effects of PIs in LSC or other malignancies involving STAT3 activation. Combined application of PIs and STAT3 inhibitor may implicate a promising strategy for managing LSC or even other HNSCC.

Keywords

Signal transducer and activator of transcription 3, STAT3; proteasome inhibitor; STAT3 inhibitor; laryngeal carcinoma; apoptosis

Introduction

Laryngeal squamous carcinoma (LSC) is a kind of head and neck squamous cell carcinoma (HNSCC), the latter of which ranks the sixth most common malignant cancer, and accounts for 35,00000 newly diagnosed cases annually worldwide [1]. Despite of successive advancement in diagnosis and treatment, the overall survival of HNSCC has just been slightly improved in recent years. In addition to distant metastasis and relapse, resistance to traditional therapeutic modalities has been the dominant cause of treatment failure, especially in advanced cases. For combating the therapeutic resistance of cancers, some novel concepts and strategies for targeted therapies have been introduced, most of which mainly focused on some key modulators of cell proliferation and apoptosis pathways. Although some preliminary results have been achieved, they are not as effective as expected.

Signal transducer and activator of transcription 3 (STAT3), a member of STAT family, is a focus of many oncogenic receptor tyrosine kinase pathways, such as EGFR, IL-6/JAK and Src [2]. It has been demonstrated that STAT3 can induce up-regulation of prosurvival proteins including Bcl-2, Bcl-xL, survivin and Mcl-1, and promote expression of angiogenesis factors, such as VEGF [3]. In fact, STAT3 is constitutively activated in many tumors [4-7] and plays a great role in proliferation, cell cycle progression and apoptosis of tumor cells [8]. In addition, activation of STAT3 is also associated with chemoresistance in many malignancies [9, 10]. Results from our previous study demonstrate that knockdown of STAT3 with specific shRNA can enhance radiosensitivity in LSC Hep-2 cells both in vitro and in vivo [11, 12]. Therefore, targeting STAT3 is a potential therapeutic strategy for LSC.

As is known, due to rapid proliferation and insufficient blood supplies, the microenvironments of most solid cancers are accompanied by hypoxia, glucose starvation and low PH, leading to endoplasmic reticulum (ER) stress due to causal accumulations of incorrect proteins [13]. Sequently, cancer cells must adapt to these unfavorable conditions for their survival via inhibiting synthesis of proteins and degrading unfolded proteins accumulated in ER by proteasome [14]. Therefore, targeting proteasome becomes a promising strategy for cancer therapy.

Early studies demonstrated that tumor-inhibiting effects of proteasome inhibitors (PIs) are attributed to repression of nuclear factor-κB (NF-κB) signaling and regulation of proteins associated with both cell cycle and apoptosis, such as p27Kip1 [15], p21cip/WAF1 [16], p53 [17], Bcl-2 [18, 19], Bax [17], Bim and Bik [20]. It is now believed that PIs induce ER stress-related cell death through unfolded protein response (UPR) [21] and confer preferential cytotoxicity towards hypoxic tumor cells [22]. However, cases of resistance to PIs are emerging, whereas the related mechanisms are not systematically elucidated [23]. Interestingly, a recent study has demonstrated that a proteasome inhibitor Bortezomib promotes STAT3 activation in HNSCC, suggestive of potential roles of STAT3 in the resistance of HNSCC to the proteasome inhibitor [24]. Therefore, it would be of great interest to elucidate whether the STAT3 plays a role in resistance LSC to PIs.

In the present study, we first assessed the associations between constitutive activation of STAT3 and clinicopathlological factors in LSC. We next investigated the role of STAT3 activation by protesome inhibitor and its impacts on proteasome-induced cell apoptosis in LSC.

Materials and Methods

Patients’ data

50 LSC patients (36 males and 14 females) who received surgical resection at Bethune International Peace Hospital were recruited, and their complete detailed clinicopathologic data were collected. Of the 50 cases, there were 17 cases of supraglottic and 34 cases of glottic cancers. The age of the present series ranged from 36 to 86 years, with a mean age of 58 years. All patients had undergone neither chemotherapy nor radiotherapy prior to surgery. According to UICC cancer staging system (2002), there were 7 cases of stage I, 6 cases of stage II, 17 cases of stage III and 20 cases of stage IV tumors. With regard to the histological grading, 6 cases were well differentiated, 13 cases were moderately differentiated and 31 cases were poorly differentiated squamous cell carcinomas (SCC). Twenty-two cases had neck node metastasis as proven by postoperative pathologic examinations.

Tumor tissue collection

With the informed consent of all patients, tissue specimens were collected immediately after surgical removal. Paired tumor-free paracancerous tissues (PCT) were taken at least 0.5 cm beyond the tumor margins. Each sample was cut in halves. One half was promptly frozen at -70 ℃ for subsequent Western blot, while the other was fixed immediately with 10% neutrally buffered formalin for histopathologic evaluation and immunohistochemistry. The acquisition of paraffin tissues and frozen specimens were approved by the Institutional Review Boards at Bethune International Peace Hospital. Neck dissection specimens were evaluated by two pathologists to confirm the status of neck node metastasis in these patients.

Immunohistochemistry and immunocytochemistry

Immunohistochemical and immunohistocytochemical studies were performed as described previously [25] using SP-9002 kit (including 0.3% hydrogen peroxide, 10% goat serum, biotinated secondary antibodies and horseradish peroxidase marked strepto- antibiotin, Zhong Shan Jin Qiao, Beijing) with some modifications. Briefly, for immunohistochemistry, consecutive sections were deparaffinized and hydrated, and subjected to 0.3% hydrogen peroxide for elimination of endogenous peroxidase at room temperature for 10 min. Slides were then heated for the retrieval of antigens in an autoclave sterilizer for 2 min. Goat serum (10%) was added to block nonspecific protein staining for 10 min. The sections were incubated with anti-STAT3 and anti-p-STAT3 (Santa Cruz Biotechnology Inc.) at 4 ℃overnight. Afterwards, they were rinsed by PBS and incubated at room temperature with biotinated secondary antibodies for 15 min under a humid condition followed by 15-min incubation with horseradish peroxidase-marked strepto- antibiotin at room temperature. Finally, they were detected with DAB kit (Zhong Shan Jin Qiao, Beijing). The immunoreactivity of STAT3 or p-STAT3 was evaluated as described previously [31]. Brown coloration was identified as positive staining. The distribution of positive staining cells was classified in 4 categories: weakly positive (+), positive (++), strongly positive (+++), and negative. For immunocytochemical study, Hep-2 cells were seeded in 6-well plates with a small cover slide in each well. When confluence reached 70% to 80%, cells grown on the cover slide were fixed with cold acetone at room temperature for 15 min. Cover slides were then subjected to 0.3% hydrogen peroxide for elimination of endogenous peroxidase at room temperature for 30 min, washed with PBS and permeabilized with 0.1% TritonX-100 on ice for 2min. The immunostaining of Hep-2 cells was performed as in immunohistocehmistry.

Cell line and culture

Hep-2 human LSC cell line (National Cancer Institute, USA) were cultured in RPMI1640 medium supplemented with 10% fetal bovine serum (FBS) under the condition of 37℃, 20% O2, 5%CO2, 95% humidity in a CO2 incubator. Cells were allowed for attachment overnight and those grown in logarithmic phase were used for subsequent experiments. All experimental groups came from the same parental cells. Cells were passaged every 2 to 3 days to maintain exponential growth.

Transfection of plasmids

The construction of STAT3-targeting shRNA expression vector and the transfection of plasmids were performed as our previous publications [12]. Briefly, the double strands of shRNA targeting gene of STAT3 (sense: 5′-CACCGCAGCAGCTGAACAACATGTTCAAGAGACATGTTGTTCAGCTGCTGCTTTTTTG-3′, the corresponding mRNA coding region is underlined) and the double strands of negative control gene (sense: 5′-CACCGTTCTCCGAACGTGTCACGTCAAGAGATTACGTGACACGTTCGGAGAATTTTTTG-3′, underline shows the target sequence) were inserted into pGPU6/GFP/Neo vector (Gene Pharma Co., Ltd), and named pGPU6/GFP/Neo-shSTAT3 and pGPU6/GFP/Neo-shNC, respectively. The procedure of transient transfection was performed according to LipofectamineTM2000 (Invitrogen, Corporation) manufacturer’s instructions in 96-well plates or/and 6-well plates. The thansfection efficiency was monitored by observation under a fluorescent microscope and by flow cytometry. Sequent assays were performed 24 hours after transfection.

MTT assay

The inhibition of hep-2 cells by chemicals was determined by MTT assay as described previously [26]. Transfected and untransfected hep-2 cells were subjected to MTT assay. After treatments with PIs (MG-132) (MERCK Corporation, Germany) and STAT3 inhibitor (AG940) (Sigma, USA) in different combinations and cisplatin (DDP, Shan Dong Qi Lu Pharmaceutics, China), 20μl 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT, 5mg/ml, Sigma, USA) were added into each well of 96-well plates. After incubation at 37 ℃ for 4h, culture medium was replaced by DMSO to dissolve formazan for coloration. Absorbance was measured at 490 nm using Enzyme-linked Immunosorbent Detector (Model 550, Bio-Rad, USA).

Flow cytometry

The transfection efficiency, apoptosis rate and cell cycle distributions were determined by a FACScan analyzer (Becton Dickinson FAC Sort, USA). Hep-2 cells, with or without treatments, were collected after trypsinization. Fresh cells were used for detection of the efficacy of transfection by flow cytometry. To determine apoptosis rate and cell cycle distribution, cells were washed by PBS for 3 times, and then were fixed with 75% ethanol overnight at 4 ℃. Prior to analysis, ethanol was discarded and 80μl RNase (final concentration: 50mg/L) and 150μl propidium iodide (final concentration: 50mg/L, Sigmar, USA) were used for staining of DNA for 30 min at 4℃. Apoptotic cells appeared as a magnitude of the sub-G1 peak due to nuclear fragmentation and loss of DNA.

Western blot

Western blot was carried out as described previously [25] with minor modifications. For tissue specimens, lysates were obtained from 100mg sample of each frozen tissue by a homogenizer. Subjected Hep-2 cells were harvested following trypsinization and centrifugalization. The cell pellets were resuspended in cell lysis buffer containing 500mM HEPES 25ml (0.1mol/L),100mM NaCl 5ml (1mol/L),2mM EDTA 0.2 ml (0.5mol/ L),sodium deoxycholate 0.25g (0.5%),NP-400 5ml (1%),10%SDS 0.5ml (0.1%),2×10-3 mol/L PMSF and 2×10-6 g/L Lupeptin to collect lysates. Subsequently, lysates originating from tissues or hep-2 cells were acquired in the same way. After determination of protein concentrations in the lysates, equivalent protein (100mg from tissues or 30mg from cells) were separated by electrophoresis on 4% to 10% SDS-PAGE gels. The subjected gels were then transferred onto nitrocellulose filters, and blocked in confining liquid containing 1% nonfat dry milk, 0.02% Tween-20 (Sigmar, USA), 10mmol/LTris-Cl,0.15mmol/L NaCl at 4℃ overnight. The blocked filters were incubated with primary antibodies (Santa Cruz Biotechnology int and Cell Signaling Technology, USA) at 4℃ overnight followed by rinse in TBST. The filters were incubated with secondary antibodies marked by HRE (Zhong Shan Jin Qiao, BeiJing) at room temperature for 1 hour followed by rinse in TBST two times. Finally, the blots were visualized by ECL kit (Pierce, USA).

Statistical analysis

Results were analyzed for statistical significance with the use of SPSS 13.0. Data were presented as mean values ± SD. The method of statistical analysis included student’s t-test for comparision of two groups, one-way ANOVA for comparison of more than two groups combined with or without least significant difference (LSD) followed for comparison of each two groups, and Pearson correlation analysis for correlation evaluation of different factors. P < 0.05 was considered significant.

Results

Constitutive activation of STAT3 is associated with clinicopathlological factors in LSC, including clinical stage, pathological grading and cervical lymph node metastasis. It has been reported that STAT3 is aberrantly activated in many tumors in comparison with its transient activation in normal tissues [27-30]. To determine activation status and expression distribution pattern of p-STAT3 in LSC, we compared levels of p-STAT3 protein of primary LSC tissues with corresponding tumor-free PCT by immunohistochemistry. As can be seen in Fig 1A-D, p-STAT3 was constitutively expressed in LSC compared with PCT. In positive samples, weakly positive staining dominates PCTs, whereas strong positive coloration occupies most of cancer tissues; it was also noted that the staining of p-STAT3 in poorly differentiated LSC is stronger than that in well differentiated LSC.

CST 2017-204_fig_1

Figure 1. The expression of phosphorylated STAT3 (p-STAT3) in human laryngeal squamous cell carcinoma (SP×400). Tissue specimens were collected and p-STAT3 protein expression levels were assessed by immunohistochemistry and western blot. A. Expression of STAT3 in well-differentiated laryngeal squamous carcinoma. B. The expression of p-STAT3 protein in moderately-defferentiated laryngeal squamous cell carcinoma (SP×400). C. The expression of p-STAT3 protein in poorly differentiated laryngeal squamous cell carcinoma (SP×400). D. The expression of p-STAT3 protein in tumor-free paracancerous laryngeal epithelial tissues (SP×200). Yellow arrow indicates representative staining cells. E. Expression of Stat3, p-STAT3, and cyclin Dl in human laryngeal carcinoma by western blot. Elevated levels of p-STAT3 and cyclin Dl in tumor tissues (T) compared to adjacent normal laryngeal mucosa (N) were further confirmed.

These findings suggest that STAT3 is activated constitutively and persistently in LSC rather than in PCT, and its activated form, p-STAT3, is likely to function in nucleus as in other malignancies. Moreover, expression of p-STAT3 is closely related to the expression of cyclin D1. Results from western blot analysis showed the same expression pattern for each protein (Fig 1E) in that there is a correlation between p-STAT3 and the expression of cyclin D1, suggesting that cyclin D1 is regulated by p-STAT3 in LSC tissues.

It has been reported in many studies that activation of STAT3 is related to poor prognosis of various cancers [31-33]. To test whether STAT3 activation is pertinent to clinicopathologic factors of LSC, we analyzed the relationship between p-STAT3 and critical prognostic factors. Results in Table 1 indicates that there is a significant correlation between p-STAT3 expression and clinical stage, pathologic grade and neck node metastasis rather than age, sex, T stage and even the site of primary tumors.

Table 1. The relationship between the expression of Stat3 and phosphorylated Stat3 protein and clinicophalogical features in laryngeal squamous cell carcinoma

n Stat3 p-Stat3
M number (rate%) P value M number (rate%) P value
Age ≥60 28 23(82.14) 0.0715 17(60.71) 0.6609
<60 22 13(59.09) 12(54.54)
Sex M 36 27(75.00) 0.6841 23(63.89) 0.1761
F 14 9 (64.29) 6 (42.86)
Tumor position Supra-glottis 17 10(58.82) 0.2473 10(58.82) 0.9325
glottis 33 26(78.78) 19(57.58)
 

Patho- grade

Well to moderately differentiated 19 10(52.63) 0.0169 5 (26.32) 0.0004
Poorly differentiated 31 26(83.87) 24(77.42)
T stage T1-T2 17 12(70.58) 0.0685 7 (41.18) 0.0836
T3-T4 33 27(81.81) 22(66.67)
Clinical stage Ⅰ-Ⅱ 13 5 (38.46) 0.0056 4 (30.77) 0.0208
Ⅲ-Ⅳ 37 31(83.78) 25(67.57)
Lymph node metastasis Yes 22 20(90.90) 0.0083 17(77.27) 0.0144
No 28 16(57.14) 12(42.86)

Activation of STAT3 is in constant existence in Hep-2 laryngeal carcinoma cells (Hep-2 cells). To test whether STATA3 is also constitutively activated in Hep-2 cells, we examined the expression of STAT3 and its related proteins by immunocytochemistry and western blot. By immunocytochemistry, STAT3 was distributed in cytoplasm of cancer cells, while p-STAT3 was observed mostly within nucleus of Hep-2 cells, which resembles the expression pattern of STAT3 and p-STAT3 in LSC tissues (Fig 2A, B, C). To examine if these proteins were constantly and pervasively expressed, we detected expression of the two proteins using western blot at different time points after the setting of cell culture. The sequent 24 h, 48 h, and 72 h were chosen as time spots of analysis. The results of western blot showed that STAT3 and p-STAT3 (Fig 2D) can be detected at different time points, suggesting that constitutive activation of STAT3 is also present in Hep-2 cells as in primary LSC.

CST 2017-204_fig_2

Figure 2. TExpression of STAT3 and p-STAT3 in human laryngeal carcinoma Hep-2 cells. Immunocytochemistry was used to demonstrate the intracellular distribution of STAT3 and p-STAT. PBS was used as a negative control. Besides, western blot was used to show the expression patterns of STAT3 and p-STAT3 overtimes (0h, 24h, 48h, 72h). β-actin represents the internal protein control. Constitutive expression and activation of STAT3 in human laryngeal carcinoma Hep-2 cells was noted. A. Cytoplasmic staining of STAT3 in Hep-2 cells (SP × 400). B. Nuclear staining of p-STAT3 in Hep-2 cells (SP × 400). C. Negative control (SP × 400). D. Expression of Stat3 and p-STAT3 in Hep-2 cells as demonstrated by western blot.

Proteasome inhibitor MG-132 induces apoptosis in Hep-2 cells, which is accompanied by additional activation of STAT3 and changes in the expression of several proteins related to cell cycle and apoptosis regulation. PIs have been approved by the United States Food and Drug Administration (FDA) for the treatments of multiple myeloma. In addition, PIs have been investigated for the treatments of many cancers. Herein, we aimed to test the therapeutic effects of PIs on Hep-2 cells. First, we tested the inhibition of proliferation in Hep-2 cells by a PI, MG-132, using MTT assay. Medium containing different concentrations of MG-132 was add into each well of treatment groups and maintained for different times. As seen in Fig 3A, the inhibition of Hep-2 cells by MG-132 displayed a concentration-dependent manner (24h:r=0.925,p<0.01;48h:r=0.944,p<0.01). Second, we attempted to confirm if MG-132 inhibits proliferation of Hep-2 cells in a time-dependent manner. For doing this, we selected the 2.5μM (IC50) of MG-132 to stimulate Hep-2 cells for 48 h. The results of Fig 3B show that the effects of MG-132 in proliferation inhibition is enhanced with prolongation of treatment time (r=0.945,p<0.01). Third, using flow cytometry, we determined apoptosis rate and cycle distribution of Hep-2 cells after treatment with MG-132. Similar with the inhibition of proliferation, MG-132 induces apoptosis in time- (r=0.888,p<0.01) and dose-dependent (r=0.842,p<0.01) manners (Fig 3C, 3D ).

CST 2017-204_fig_3

Figure 3. Effects of a proteasome inhibitor, MG-132, on Hep-2 cells. Hep-2 cells were treated with different concentrations of MG-132 for 24 hours. A concentration of 2.5μM was chosen for stimulation of Hep-2 cells for different times. Alterations in cell apoptosis, cell cycle and protein expression of STAT3 and p-STAT3 were assessed simultaneously. A. The dosage and effect curve of MG-132 (n = 3). B. The time and effect curve of MG-132 (n = 3). C. Apoptosis rate of Hep-2 cells treated with MG-132 for different time (n = 3). D. Apoptosis rate of Hep-2 cells treated with MG-132 at different concentrations (*P < 0.05, versus untreated groups (0μM) (n = 3). E. The correlated expression of cell cycle regulation proteins in Hep-2 cells treated with MG-132 for different time. F. Variation of cell cycle of Hep-2 cells treated with MG-132 for different time (n = 3). G. MG-132-induced STAT3 activation and Bcl-2 expression in Hep-2 cells.

To confirm the mechanisms underlying the cell cycle changes induced by MG-132, we used western blot to check the expression levels of some cell cycle regulators. As seen in Fig 3E, the expression of P21 is elevated significantly compared with the decreased levels of the expression of cyclinD1, suggesting that the changes of cell cycle induced by MG-132 were associated with upregulation of P21 and downregulation of cyclinD1. Simultaneously, cell cycle arrest was noted at G0/G1 phase and/or G2/M phase overtimes after treatment with MG-132 (Fig 3F), the former of which is more prominent than the latter.

From the results of western blot in Fig 3G, we can confirm that STAT3 and its activated form, p-STAT3, are expressed at all times in cultured Hep-2 cells, even in the absence of MG-132 treatment, and the level of its expression was relatively constant and stable. In the presence of MG-132, the expression of p-STAT3 was elevated within short period of time and remained at a stable level; however, the increased expression of p-STAT3 was not related to the duration of MG-132 treatment. Bcl-2 was identified as a downstream of STAT3, and facilitates anti-apoptosis activity in cancer cells [34]. Expression of Bcl-2 protein increased overtime after MG-132 treatment, which may contributed to the MG-132-induced STAT3 activation and resistance of Hep-2 cells to MG-132-induced apoptosis.

Knockdown of STAT3 by shRNA, abrogates the expression and thus activation of STAT3, and potentiates the apoptosis-inducing effects of MG-132. To further confirm the role of the STAT3 activation in inducing resistance of Hep-2 cells to MG-132, we used RNAi to knockout the expression of STAT3, and thus the “constitutive” (inherent) and “PI-induced” (acquired) activation of STAT3. The transfection efficiency was 85.76% by flow cytometry (data not shown). The results of Fig 4A showed that combined group has a similar expression level of p-STAT3 with pshSTAT3 group, with a higher inhibition rate than any other groups (all p<0.01). Results from apoptosis detection Fig 4B showed a more striking apoptosis rate 55.80±3.12% in combined group than in any other group (all p<0.01). In pshSTAT3 group, moderate apoptosis and proliferation inhibition rates were induced. Therefore, blockade of STAT3 by shRNA, abrogates the expression and thus both “inherent” and “acquired” activation of STAT3, and potentiates the MG-132-induced apoptosis in Hep-2 cells.

CST 2017-204_fig_4

Figure 4. Effects of MG-132 combined with STAT3 RNAi on proliferation inhibition and apoptosis in Hep-2 cells, as assessed by MTT assay and flow cytometry. Hep-2 cells were seeded into 96-well plates for MTT assay, or 6-well plates for flow cytometry. Subjected Hep-2 cells were categorized as five groups: normal control group (untreated Hep-2 cells), negative control group (Hep-2 cells transfected with negative plasmid, pshNeg group), MG-132 group (Hep-2 cells treated with 2.5μM MG-132 only), combined group (Hep-2 cells transfected with positive plasmid combined with 2.5μM MG-132 treatment), pshSTAT3 group (Hep-2 cells transfected with positive plasmid without any other treatment). A. The proliferation inhibition rate of Hep-2 cells in different groups (n=3) (versus pshNeg group, all *P<0.01; pshSTAT3 group versus MG-132 group, #P<0.01). B. The apoptosis rate of Hep-2 cells in different groups (n=3) (control group versus pshNeg group, *P<0.01; pshSTAT3 group versus MG-132 group, #P<0.01). C. Expression of p-STAT3, cyclin D1 and Bcl-2 in Hep-2 cells in different groups was analyzed by Western blotting.. Lane 1: control group; lane 2: pshNeg group; lane 3: MG-132 group; lane 4: combined group; lane 5: pshSTAT3 group.

With regard to the activation of STAT3 at different situations, as seen in Fig 4C, p-STAT3 was expressed at the highest level in MG-132 group. By contrast, p-STAT3, cyclin D1, and Bcl-2 were expressed at lower levels in combined group, compared with each of the normal control group, negative control group and MG-132 group. The similar expression and activation patterns of STAT3 noticed in combined group and pshSTAT3 group indicated that p-STAT3 may participate in the formation of resistance of Hep-2 cells to MG-132 at a transcriptional level. Taken together, activation of STAT3 is responsible for the resistance of Hep-2 cells to MG-132 at least in part by regulating the anti-apoptosis factor Bcl-2 expression.

Combined treatment with STAT3 inhibitor AG490 and proteasome inhibitor MG-132 induces apoptosis more effectively in Hep-2 cells. In view of the fact that activation of STAT3 played a role in the therapeutic resistance to MG-132 in Hep-2 cells, which can be sensitized by knockdown of STAT3, we studied the effects of combined using STAT3 inhibitor AG490 and proteasome inhibitor MG-132 on proliferation and apoptosis. As seen in Fig 5 A, B, combination treatment by AG490 and MG-132 can induce significant increase in cell proliferation inhibition and apoptosis, indicative of effective synergism.

CST 2017-204_fig_5

Figure 5. Effects of AG-490 combined with MG-132 on Hep-2 cells. Hep-2 cells were seeded into 96-well or 6-well plates followed by treatment with 1μM of MG-132 for 48h with or without presence of different concentrations (10, 20, 40, 80μM) of AG490. A concentration of 10μmol/L was for analysis of AG490-induced apoptosis. A. The proliferation inhibition rate of Hep-2 cells treated with the different concentrations AG490 alone and in combination with MG-132 for 48h (n=3). B. The apoptosis rate of Hep-2 cells treated with AG490 alone and in combination with MG-132(1μM) for 48h (n=3) (compared with control group,*P<0.01; compared with AG490 group and MG-132 group, all #P<0.01).

Discussion

Like other malignancies in HNSCC, LSC often exhibits resistance to various therapeutic strategies including chemoradiation and targeted therapies, especially in advanced cases. This difficult situation is often attributed to aberrant expression and activation of some specific genes, such as STAT3, which are utilized by cancer cells to maintain their biological behaviors. In this regard, cancer cells may become vulnerable to the assigned treatment modalities secondary to abrogating the functions of major responsible oncogenes, the so-called “Achilles heel”, which constitutes the fundamental rationale for targeted therapy [35].

Previous studies have demonstrated that activation of STAT3 is associated with chemo- and/or radioresistance in Hep-2 cells, and knockdown of STAT3 by siRNA or blockade of STAT3 activation by its inhibitors sensitizes chemo- and/or radiotherapy both in vitro or in vivo [11, 12, 36, 37]. Importantly, we also demonstrated in the present investigation that constitutive activation of STAT3 was associated with clinicopathlologic factors in LSC, which include clinical stage, pathological grading and cervical lymph node metastasis. This is consistent with the findings in the study of Rosen et al, in which they demonstrated that p-STAT3 was associated with poor prognosis of ovarian cancer [38]. Moreover, results of Kusaba’s study [33] also demonstrated that p-STAT3 is correlated with invasion of vein, nodal metastasis and Dukes grades instead of pathological grading in ovarian cancer. In the present study, we found expression of cyclin D1, one of the important factors downstream of STAT3, was also elevated. Taken together, correlations between activation of STAT3 and clinicopathlological factors of different cancers may depend on cell types from which they originate.

In addition to intrinsic elements, cancer cells in solid tumors also depends on extrinsic factors that form surrounding microenvironment (also called “niche”) for their proliferation and survival. The feature of the niche is characterized by low PH, hypoxia and glucose-deprived starvation as a result of rapid proliferation by tumor cells and insufficient delivery by intra-tumor capillary [39]. Under the stressful conditions, a mass of misfolded or unfolded proteins accumulate in endocytoplasmic reticulum (ER). Tumor cells must activate UPR to inhibit synthesis of protein and mRNA and degrade excessive protein by proteasome to accommodate ER-stress, which is rarely experienced by normal tissue cells [40]. It has been demonstrated that severe ER-stress caused by accumulated misfolded proteins can activate programmed death in tumor cells [39]. Therefore, proteasome can serve as a potential target for cancer therapy.

Recently, PIs was introduced into clinical treatment of cancers, either as a single agent or a combined therapy to sensitize traditional chemo- and/or radiotherapy. However, the resistance to PIs remains a problem to solve, of which underlying mechanisms have not been explained pertinently [23]. The prerequisite for us to select PIs as an anti-LSC agent resides on the fact that PIs is effective against cyclin D1 highly-expressed cancers [41]. However, it remains to be elucidated whether other HNSCC cells were dependent or independent on cyclin D1 expression for their proliferation, survival and progression.

Given the fact that activation of STAT3 is associated with clinicopathologic behaviors and chemoradiation resistance in LSC, STAT3 is definitely a potential target for therapeutic purposes. The pro-survival and –proliferation effects of p-STAT3 let us to postulate that the constitutive (inherent) along with PI-induced (acquired) activation of STAT3 may constitute a major cause of resistance to PIs in LSC. Abrogation of both existed and acquired STAT3 activation may sensitize the LSC cells to PI treatment. In present investigation, we found for the first time that activation of STAT3 contributed to resistance of LSC to MG-132 at least in part by upregulating expression of antiapoptotic protein Bcl-2, which is partly consistent with results of a recent study [24]. Our results showed that PIs plus AG490 displayed a more powerful killing effect to LSC cells in a synergetic manner. However, such a combination may not be applied to all cancers, because overexpression of Bcl-2 is associated with a good prognosis in some other malignancies [42].

In the present study, we also demonstrated that MG-132 can induce G0/G1 cell cycle arrest, via downregulation of cyclin D1 in LSC, which is also observed in some other cancers [43-45]. It can be inferred that an ideal local control is to be achieved by PIs through inducing quiescence in LSC cells. However, a fact that cannot be ignored is that G0/G1 cycle arrest also constitutes a cause of therapeutic resistance in some tumors.

Although inhibiting STAT3 is capable of inducing quiescence and potentiating PI-induced apoptosis in LSC cells, there has been no effective STAT3 inhibitor that can be safely used for humans in clinic for treatment of HNSCC. We recently defined dihydroartemisinin as putative STAT3 inhibitor, which might be safely used in clinic for cancer treatment [46]. Much work has to be done for developing new STAT3-inhibiting agents and exploring new treatment strategies and modalities based on the synergized apoptosis-inducing effects of PIs with STAT3 inhibitors.

Acknowledgements: The research work in the present investigation is supported by Key Basic Research Project Fund of Hebei Province, China (Fund No. 14967721D) and in part by Scientific Research Fund of Bethune International Peace Hospital, China.

Conflict of Interest: None

References

  • Parkin DM, Bray F, Ferlay J, Pisani P (2005) Global cancer statistics, 2002. CA Cancer J Clin 55: 74-108. [crossref]
  • Leeman RJ, Lui VW, Grandis JR (2006) STAT3 as a therapeutic target in head and neck cancer. Expert Opin Biol Ther 6: 231-241. [crossref]
  • Chen SH, Murphy DA, Lassoued W, Thurston G, Feldman MD, et al. (2008) Activated STAT3 is a mediator and biomarker of VEGF endothelial activation. Cancer Biol Ther 7: 1994-2003. [crossref]
  • Grandis JR, Drenning SD, Chakraborty A, Zhou MY, Zeng Q, et al. (1998) Requirement of Stat3 but not Stat1 activation for epidermal growth factor receptor- mediated cell growth In vitro. J Clin Invest 102: 1385-1392. [crossref]
  • Guo C, Yang G, Khun K, Kong X, Levy D, et al. (2009) Activation of Stat3 in renal tumors. Am J Transl Res 1: 283-290. [crossref]
  • Kim SM, Kwon OJ, Hong YK, Kim JH, Solca F, et al. (2012) Activation of IL-6R/JAK1/STAT3 signaling induces de novo resistance to irreversible EGFR inhibitors in non-small cell lung cancer with T790M resistance mutation. Mol Cancer Ther 11: 2254-2264. [crossref]
  • Sánchez A, Nagy P, Thorgeirsson SS (2003) STAT-3 activity in chemically-induced hepatocellular carcinoma. Eur J Cancer 39: 2093-2098. [crossref]
  • Turkson J, Jove R (2000) STAT proteins: novel molecular targets for cancer drug discovery. Oncogene 19: 6613-6626. [crossref]
  • Bonner JA, Trummell HQ, Willey CD, Plants BA, Raisch KP (2009) Inhibition of STAT-3 results in radiosensitization of human squamous cell carcinoma. Radiother Oncol 92: 339-344. [crossref]
  • Ara T, Nakata R, Sheard MA, Shimada H, Buettner R, et al. (2013) Critical role of STAT3 in IL-6-mediated drug resistance in human neuroblastoma. Cancer Res 73: 3852-3864. [crossref]
  • Li X, Wang H, Lu X and Di B (2010) Silencing STAT3 with short hairpin RNA enhances radiosensitivity of human laryngeal squamous cell carcinoma xenografts in vivo. Exp Ther Med 1: 947-953.
  • Li X, Wang H, Lu X and Di B (2010) STAT3 blockade with shRNA enhances radiosensitivity in Hep-2 human laryngeal squamous carcinoma cells. Oncol Rep 23: 345-353.
  • Määttänen P, Gehring K, Bergeron JJ, Thomas DY (2010) Protein quality control in the ER: the recognition of misfolded proteins. Semin Cell Dev Biol 21: 500-511. [crossref]
  • Orlowski RZ (2005) The ubiquitin proteasome pathway from bench to bedside. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program: 220-225
  • Uddin S, Ahmed M, Bavi P, et al. (2008) Bortezomib (Velcade) induces p27Kip1 expression through S-phase kinase protein 2 degradation in colorectal cancer. Cancer Res 68: 3379-3388.
  • Mitsiades CS, McMillin D, Kotoula V, et al (2006) Antitumor effects of the proteasome inhibitor bortezomib in medullary and anaplastic thyroid carcinoma cells in vitro. The Journal of clinical endocrinology and metabolism 91: 4013-402.
  • Jones RJ, Chen Q, Voorhees PM, Young KH, Bruey-Sedano N, et al. (2008) Inhibition of the p53 E3 ligase HDM-2 induces apoptosis and DNA damage–independent p53 phosphorylation in mantle cell lymphoma. Clin Cancer Res 14: 5416-5425. [crossref]
  • Li B and Dou QP (2000) Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression. Proc Natl Acad Sci U S A 97: 3850-3855.
  • Lara PN, Bold RJ, Mack PC, Davies AM, Gumerlock PH and Gandara DR (2005) Proteasome inhibition in small-cell lung cancer: preclinical rationale and clinical applications. Clinical lung cancer 7 Suppl 2: S67-7
  • Li C, Li R, Grandis JR and Johnson DE (2008) Bortezomib induces apoptosis via Bim and Bik up-regulation and synergizes with cisplatin in the killing of head and neck squamous cell carcinoma cells. Mol Cancer Ther 7: 1647-1655
  • McConkey DJ, Zhu K (2008) Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist Updat 11: 164-179. [crossref]
  • Fels DR, Ye J, Segan AT, et al. (2008) Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of endoplasmic reticulum stress pathways. Cancer Res 68: 9323-9330.
  • Orlowski RZ, Kuhn DJ (2008) Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res 14: 1649-1657. [crossref]
  • Li C, Zang Y, Sen M, Leeman-Neill RJ, Man DS, et al. (2009) Bortezomib up-regulates activated signal transducer and activator of transcription-3 and synergizes with inhibitors of signal transducer and activator of transcription-3 to promote head and neck squamous cell carcinoma cell death. Mol Cancer Ther 8: 2211-2220. [crossref]
  • Wang Q, He Z, Zhang J, et al. (2005) Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance. Cancer detection and prevention 29: 544-55.
  • Song X, Liu X, Chi W, Liu Y, Wei L, et al. (2006) Hypoxia-induced resistance to cisplatin and doxorubicin in non-small cell lung cancer is inhibited by silencing of HIF-1alpha gene. Cancer Chemother Pharmacol 58: 776-784. [crossref]
  • Zhou Y, Li M, Wei Y, Feng D, Peng C, et al. (2009) Down-regulation of GRIM-19 expression is associated with hyperactivation of STAT3-induced gene expression and tumor growth in human cervical cancers. J Interferon Cytokine Res 29: 695-703. [crossref]
  • Corvinus FM, Orth C, Moriggl R, Tsareva SA, Wagner S, et al. (2005) Persistent STAT3 activation in colon cancer is associated with enhanced cell proliferation and tumor growth. Neoplasia 7: 545-555. [crossref]
  • Yu CL, Jove R and Burakoff SJ (1997): Constitutive activation of the Janus kinase-STAT pathway in T lymphoma overexpressing the Lck protein tyrosine kinase. J Immunol 159: 5206-5210.
  • Rahaman SO, Harbor PC, Chernova O, Barnett GH, Vogelbaum MA, et al. (2002) Inhibition of constitutively active Stat3 s.uppresses proliferation and induces apoptosis in glioblastoma multiforme cells. Oncogene 21: 8404-8413
  • Masuda M, Suzui M, Yasumatu R, et al.(2002) Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res 62: 3351-3355.
  • Horinaga M, Okita H, Nakashima J, Kanao K, Sakamoto M, et al. (2005) Clinical and pathologic significance of activation of signal transducer and activator of transcription 3 in prostate cancer. Urology 66: 671-675.
  • Kusaba T, Nakayama T, Yamazumi K, et al. (2005) Expression of p-STAT3 in human colorectal adenocarcinoma and adenoma; correlation with clinicopathological factors. J Clin Pathol 58: 833-838
  • Groner B, Lucks P, Borghouts C (2008) The function of Stat3 in tumor cells and their microenvironment. Semin Cell Dev Biol 19: 341-350. [crossref]
  • Christoffersen T, Guren TK, Spindler KL, Dahl O, Lonning PE, et al. (2009) Cancer therapy targeted at cellular signal transduction mechanisms: strategies, clinical results, and unresolved issues. Eur J Pharmacol 625: 6-22.
  • Hsu HW, Gridley DS, Kim PD, Hu S, de Necochea-Campion R, et al. (2013) Linifanib (ABT-869) enhances radiosensitivity of head and neck squamous cell carcinoma cells. Oral Oncol 49: 591-597. [crossref]
  • Sen M, Joyce S, Panahandeh M, Li C, Thomas SM, et al. (2012) Targeting Stat3 abrogates EGFR inhibitor resistance in cancer. Clin Cancer Res 18: 4986-4996. [crossref]
  • Rosen DG, Mercado-Uribe I, Yang G, Bast RC Jr, Amin HM, et al. (2006) The role of constitutively active signal transducer and activator of transcription 3 in ovarian tumorigenesis and prognosis. Cancer 107: 2730-2740. [crossref]
  • Wang G, Yang ZQ and Zhang K (2010) Endoplasmic reticulum stress response in cancer: molecular mechanism and therapeutic potential. Am J Transl Res 2: 65-74.
  • Healy SJ, Gorman AM, Mousavi-Shafaei P, Gupta S and Samali A (2009) Targeting the endoplasmic reticulum-stress response as an anticancer strategy. Eur J Pharmacol 625: 234-246.
  • Ishii Y, Pirkmaier A, Alvarez JV, Frank DA, Keselman I, et al. (2006) Cyclin D1 overexpression and response to bortezomib treatment in a breast cancer model. J Natl Cancer Inst 98: 1238-1247. [crossref]
  • Zhuang L, Lee CS, Scolyer RA, et al. (2007) Mcl-, Bcl-XL and Stat3 expression are associated with progression of melanoma whereas Bcl-2, AP-2 and MITF levels decrease during progression of melanoma. Mod Pathol 20: 416-426.
  • Wang Y, Rishi AK, Puliyappadamba VT, et al. (2010) Targeted proteasome inhibition by Velcade induces apoptosis in human mesothelioma and breast cancer cell lines. Cancer Chemother Pharmacol 66: 455-466.
  • Bazzaro M, Lee MK, Zoso A, et al. (2006) Ubiquitin-proteasome system stress sensitizes ovarian cancer to proteasome inhibitor-induced apoptosis. Cancer Res 66: 3754-3763
  • Liu T, Peng H, Zhang M, Deng Y and Wu Z (2010) Cucurbitacin B, a small molecule inhibitor of the Stat3 signaling pathway, enhances the chemosensitivity of laryngeal squamous cell carcinoma cells to cisplatin. Eur J Pharmacol 641: 15-22.
  • Jia L, Song Q, Zhou C, et al. (2016) Dihydroartemisinin as a Putative STAT3 Inhibitor, Suppresses the Growth of Head and Neck Squamous Cell Carcinoma by Targeting Jak2/STAT3 Signaling. PLoS One 11: e0147157. [crossref]

Overexpression of IFN-γR Increases the Radiotherapy Resistance of Nasopharyngeal Carcinoma

DOI: 10.31038/CST.2017214

Abstract

Purpose: To assess the relationship between IFN-γR expression and clinical radiotherapy outcome in nasopharyngeal carcinoma.

Methods: IFN-γR expression was examined by immunohistochemistry from 70 nasopharyngeal carcinoma patients. Then, statistical analysis was conducted to explore the correlation between IFN-γR expression and tumor clinicopathological charateristics. Finally, CNE-1 cells were employed to detect the effects of IFN-γR expression on radiotherapy outcomes by in vitro assays.

Results: IFN-γR was upregulated dramatically in nasopharyngeal carcinoma tissues. Elevated expression of IFN-γR correlated significantly with tumor stage, positive lymph node metastasis and poor prognosis in patients with nasopharyngeal carcinoma. In vitro assays demonstrated that overexpression of IFN-γR promoted the radio-resistance ability in CNE-1 cells. Knockdown of IFN-γR facilitated CNE-1 cells more sensitive to radiation, and a wobble mutant of IFN-γR could restore this effect.

Conclusions: Overexpression of IFN-γR may correlate positively with radiotherapy resistance of nasopharyngeal carcinoma.

Keywords

IFN-γR; overexpression; prognosis; radiation resistance; NPC

Introduction

Nasopharyngeal carcinoma is one of the most common malignancies and occupies the seventh leading cause of cancer-related deaths in China [1]. Particularly in South China, nasopharyngeal carcinoma is the most prevalent. Locally, advanced nasopharyngeal carcinoma patients are generally treated with radiotherapy, and the prognosis is related with radiotherapy [2]. Both local-regional failure and early systemic dissemination of the disease always contribute to treatment failure, which makes radio-sensitization promising in improving the outcome of therapeutic irradiation [3].

IFN-γ plays a pleiotropic role in modulating immunity and antiviral activity. It stimulates macrophages and NK cells to produce MHC class I and II molecules, while IFN-γR is not required for the development of the immune system [4]. Researches showed that IFN-γR was overexpressed in breast cancer and its amplification was closely related with lymph node metastasis and poor prognosis [5-7]. Until now, there are still no reports about whether IFN-γR has an effect on the radiotherapy for NPC. In the present study, we found for the first time that IFN-γR expression correlated with the prognosis of NPC patients who received radiotherapy. High IFN-γR levels improved the radiation resistance ability of NPC cells.

Patients and methods

Tissue samples

Seventy tissues of nasopharyngeal carcinoma were freshly collected by nasopharyngoscope and procured by the Department of Pathology, Cancer Center of Wuhan Union Hospital, China. The collection of tumor samples was approved by the institutional ethics committee. Informed consent forms had been signed for sample collection before. Tumor regions were technically separated by experienced pathologists and promptly stored at liquid nitrogen until needed. All patients included underwent concurrent chemotherapy (Cisplatin, 40mg/m2, weekly) with radiation therapy for the first time. They received irradiation with daily fraction doses of 2.12Gy for a total dose 70 Gy, which was administered with linear accelerators. The clinicopathologic features of patients were showed in Table 1.

CST 2017-203 Tab1

Immunohistochemical staining

Immunohistochemical staining was conducted as previously described. After rehydration, the paraffin-embedded nasopharyngeal carcinoma tissues were immersed in 3% hydrogen peroxide solution for 10 min, and then heated in EDTA buffer (pH 8.0) for 25 min. The slides were blocked with 10% normal rabbit serum at 37°C for 30 min, and then incubated with rabbit polyclonal antibody against primary antibody (IFN-γR, 1:200, ABCAM) overnight at 37°C. The slides were washed with PBS, and then incubated with biotinylated second antibody (diluted 1:200) for another 30 min at 37°C. Finally, streptavidin-peroxidase reactivity was detected with DAB solution. Cell nucleus was counterstained with hematoxylin. The immunohistochemical slides were graded according to the ratio of tumor cells with nuclear labeling. The level of IFN-γR- positive staining was scored into three grades: 0 for < 10%, positive for 10–30%, strong positive for > 30%.

Plasmid constructs and small interfering RNA synthesis

Double-stranded oligonucleotide with the following sequence was synthesized into the pSilencer 3.1- H1 neo small interfering RNA (siRNA) expression vector, and the scrambled siRNA was applied as control. Mutant DNA-IFN-γR (BOSTER, Wuhan, China) was used to resist the effect of IFN-γR-RNAi.

Cell culture and transfection

The CNE-1 cells (one of the esophageal squamous carcinoma cell lines) were cultured in RPMI 1640 (Invitrogen) with 10% fetal bovine serum. Cell transfection was carried out using Lipofectamine 2000 (Invitrogen) following the manufacturer’s instruction. Cells were harvested after transient transfection for 48 h. For stable transfection, cells were selected with 200 μg/ml G418, and passaged by serial dilution. During the next two weeks, colonies were screened by selective medium. The positive colonies with IFN-γR overexpression and IFN-γR knockdown were chosen for experiments.

Western blot analysis

Total proteins were isolated from patient tissues and cultured cells with lysis buffer [10 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP40] containing protease inhibitors. After centrifugation (15,000 RCF, 30 min, 4°C), supernatants were recovered for immunoblot analysis. The proteins were subjected to SDS-PAGE and then transferred onto polyvinylidenedifluoride (PVDF) membranes (Millipore). Next, the membranes were blocked and then incubated with primary antibody against IFN-γR (1:500, ABCAM) at 4°C overnight, then incubated with horseradish peroxidase-conjugated accordingly secondary antibody for 1 h at room temperature and developed using ECL detection reagent (BOSTER).

Ionizing radiation

CNE-1 cell line was exposed to IR in a JL Shepherd Model 143 137Cesium gamma and irradiated at a rate of 2.4 Gy/min.

Colony formation assay

A total of 400 cells were aliquoted into 6- well plate and exposed to γ rays in a dose of 5 Gy in triplicate. After incubation for 10 days, visible colonies were fixed in methanol and stained with Giemsa. Colonies were counted in each well. Each experiment was performed three times independently.

Results

Overexpression of IFN-γR in esophageal squamous cell carcinoma

IFN-γR expression was examined in 70 NPC samples and adjacent normal tissue by immunohistochemistry. Results showed that IFN-γR was hardly expressed or revealed weak expression in normal epithelial cells. While, in 36 cases of 70 NPC samples, IFN-γR expression exhibited significantly enhanced trend compared to the normal epithelium [Table 1]. Statistical analysis indicated that elevated expression of IFN-γR was significantly related to tumor stage, clinical stage and lymph node metastasis (p < 0.05) [Table 2].

CST 2017-203 Tab2

Next, the effect of IFN-γR expression on patient’s survival was analyzed. IFN-γR expression is positive in some patients and negative positive in others [Figure 1A]. After the included patients underwent chemotherapy combined with radiation therapy (55 out of 70), compared to NPC patients with low IFN-γR expression in cancerous tissue was significantly higher, patients with cancerous tissue of high IFN-γR expression showed significantly lower survival rate (p = 0.001) [Figure 1B].

Figure 1. Representative IFN-γR immunostaining in NPC and effects of IFN-γR expression on patient’s survival. A: IFN-γR expression is positive in some patients and negative positive in others. B: In the patients that received concurrent chemotherapy with radiation therapy (55 out of 70), the survival rate of patients with low IFN-γR expression was significantly higher than that of patients with high IFN-γR expression (p = 0.001).

Figure 1. Representative IFN-γR immunostaining in NPC and effects of IFN-γR expression on patient’s survival. A: IFN-γR expression is positive in some patients and negative positive in others. B: In the patients that received concurrent chemotherapy with radiation therapy (55 out of 70), the survival rate of patients with low IFN-γR expression was significantly higher than that of patients with high IFN-γR expression (p = 0.001).

Effects of IFN-γR on the radio-sensitivity of CNE-1 cells

To confirm the clinical data, CNE-1 cells were radiated with a dose of 5 Gy for 0, 2, 4, 8 and 12 h to know IFN-γR expression change. IFN-γR expression was increased after radiation in a time-dependent manner, which implied that IFN-γR may be associated with the radiation response of CNE-1 cells [Figure 2].

Figure 2. Expression of IFN-γR induced by radiation in CNE-1 cells. Elevated IFN-γR was observed after γ ray radiation with a dose of 5 Gy, and its level was highest at 10 hours.

Figure 2. Expression of IFN-γR induced by radiation in CNE-1 cells. Elevated IFN-γR was observed after γ ray radiation with a dose of 5 Gy, and its level was highest at 10 hours.

Colony formation assay was conducted to detect the effects of IFN-γR on the radio-sensitivity of CNE-1 cells [8]. IFN-γR knock-down cells showed a significantly lower survival fraction than control groups [Figures 3A and B]. Conversely, the survival fraction of cells with forced IFN-γR expression was higher [Figures 3C and D]. Therefore, results indicated a positive relationship between IFN-γR expression and radio-sensitivity of CNE-1 cells.

Figure 3. Effects of IFN-γR expression on the radio-sensitivity of CNE-1 cells. A: Expression of IFN-γR decreased in IFN-γR RNAi cells. B: Knock-down of IFN-γR promoted the radio-sensitivity of CNE-1 cells. C: Expression of IFN-γR increased after positive transfection. D: Forced IFN-γR expression inhibited the radio-sensitivity of CNE-1 cells.

Figure 3. Effects of IFN-γR expression on the radio-sensitivity of CNE-1 cells. A: Expression of IFN-γR decreased in IFN-γR RNAi cells. B: Knock-down of IFN-γR promoted the radio-sensitivity of CNE-1 cells. C: Expression of IFN-γR increased after positive transfection. D: Forced IFN-γR expression inhibited the radio-sensitivity of CNE-1 cells.

Discussion

Overexpression of IFN-γR has been observed in various types of human tumors, and elevated expression of IFN-γR indicated poor prognoses for patients, such as gastric carcinoma, prostate cancer, oral squamous cell carcinoma and non-small cell lung cancer. Particularly, IFN-γR hyperexpression was found to be associated with poor prognosis and reduced p27 expression in gastric carcinoma. Some studies have revealed that IFN-γR was amplified and upregulated in breast cancer. Moreover, its overexpression indicated worse tumor stage and lymphatic metastasis. However, there have been no reports on IFN-γR expression in NPC until now. Our results showed for the first time that IFN-γR expression increased in NPC, which significantly related with tumor stage and positive lymphatic metastasis. Coincidently, other studies have demonstrated that elevated IFN-γR level was correlated with poor prognosis in colorectal carcinoma [9,10]. However, no significance was shown between IFN-γR level and overall survival of NPC patients in the present study. We could not help putting forward the speculation whether IFN-γR expression affects radiotherapy outcome. Survival analysis unveiled that the overall survival rate of patients received radiation therapy was indeed affected by IFN-γR expression. Histological detection also showed a strong association betweeen IFN-γR hyperexpression and T-stage and lymphatic metastasis. Besides, in vitro assay further confirmed this point. This was the first report about the correlation between IFN-γR expression and radiation resistant of carcinoma.

Some studies reported that downregulation of IFN-γR suppressed prostate cancer cells proliferation, anchorage-independent growth and migration 11,12], while knockdown of Cks2 induced apoptosis [13,14]. Decreased IFN-γR promoted G2/M phase arrest and programmed cell death in human lung cancer cells [15]. Some study also found that overexpressin of IFN-γR in breast cancer cells repressed cell apoptosis through the MEK-Erk pathway [16]. However, no reports have described the apoptotic effects of IFN-γR expression in radiation-treated cells. In our next study, we may find out that overexpression of IFN-γR could induce apoptosis in CNE-1 cells after radiation.

To conclude, this study identified that overexpression of IFN-γR correlated with the poor prognosis of NPC patients receiving radiotherapy and elevated IFN-γR expression promoted the radiation resistance ability of NPC cells.

Conflict of interest: None declared.

Source of funding: None.

Consent: Written informed consent was obtained from the patients.

Acknowledgments: The Authors thank all the people involved in the project.

References

  • Chua ML, Wee JT, Hui EP, Chan AT (2016) Nasopharyngeal carcinoma. Lancet 387: 1012-1024. [Crossref]
  • Qu S, Liang ZG, Zhu XD (2015) Advances and challenges in intensity-modulated radiotherapy for nasopharyngeal carcinoma. Asian Pacific journal of cancer prevention: APJCP16:1687-1692.
  • Tang LL, Chen L, Mao YP, Li WF, et al. (2015) Comparison of the treatment outcomes of intensity-modulated radiotherapy and two-dimensional conventional radiotherapy in nasopharyngeal carcinoma patients with parapharyngeal space extension. Radiotherapy and oncology: journal of the European Society for Therapeutic Radiology and Oncology116:167-173.
  • Swisher JF, Feldman GM (2015) The many faces of Fcgamma RI: implications for therapeutic antibody function. Immunological reviews 268:160-174.
  • Nagai Y, Tsuchiya H, Runkle EA, Young PD et al. (2015) Disabling of the erbB Pathway Followed by IFN-gamma Modifies Phenotype and Enhances Genotoxic Eradication of Breast Tumors. Cell reports12:2049-2059.
  • Saad S, Dunn LB, Koetters T, Dhruva A, et al. (2014) Cytokine gene variations associated with subsyndromal depressive symptoms in patients with breast cancer. European journal of oncology nursing: The official journal of European Oncology Nursing Society18:397-404.
  • Chen C, Guo L, Shi M, Hu M et al. (2012) Modulation of IFN-gamma receptor 1 expression by AP-2alpha influences IFN-gamma sensitivity of cancer cells. The American journal of pathology180:661-671.
  • Zhao X, Feng X, Peng D, Liu W et al. (2016) Anticancer activities of alkaloids extracted from the Ba lotus seed in human nasopharyngeal carcinoma CNE-1 cells. Experimental and therapeutic medicine12:3113-3120.
  • Wang L, Wang Y, Song Z, Chu J et al. (2015) Deficiency of interferon-gamma or its receptor promotes colorectal cancer development. Journal of interferon & cytokine research: the official journal of the International Society for Interferon and Cytokine Research35:273-280.
  • Ni C, Wu P, Zhu X, Ye J et al. (2013) IFN-gamma selectively exerts pro-apoptotic effects on tumor-initiating label-retaining colon cancer cells. Cancer letters 336:174-184.
  • Wang X, Breeze A, Kulka M. (2015) N-3 polyunsaturated fatty acids inhibit IFN-gamma-induced IL-18 binding protein production by prostate cancer cells. Cancer Immunol Immunother 64:249-258.
  • Wee ZN, Li Z, Lee PL, Lee ST et al. (2014) EZH2-mediated inactivation of IFN-gamma-JAK-STAT1 signaling is an effective therapeutic target in MYC-driven prostate cancer. Cell reports 8:204-216.
  • Lan Y, Zhang Y, Wang J, Lin C et al. (2008) Aberrant expression of Cks1 and Cks2 contributes to prostate tumorigenesis by promoting proliferation and inhibiting programmed cell death. International journal of cancer 123:543-551.
  • You H, Lin H, Zhang Z (2015) CKS2 in human cancers: Clinical roles and current perspectives (Review). Molecular and clinical oncology 3:459-463.
  • Yang D, Stewart TJ, Smith KK, Georgi D et al. (2008) Downregulation of IFN-gammaR in association with loss of Fas function is linked to tumor progression. International journal of cancer122:350-362.
  • Chatterji U, Riby JE, Taniguchi T, Bjeldanes EL et al. (2004) Indole-3-carbinol stimulates transcription of the interferon gamma receptor 1 gene and augments interferon responsiveness in human breast cancer cells. Carcinogenesis 25:1119-1128.

Long Term Results of High Dose Rate Brachytherapy and External Beam Radiotherapy for Local and Locally Advanced Prostate Cancer

DOI: 10.31038/CST.2017213

Abstract

Purpose: Several studies provided evidence for the efficacy of dose-escalation on biochemical control (BC) of prostate cancer and High-dose-rate brachytherapy (HDR) is one method for it.

Materials and Methods: Patients with histological diagnosis Gleason scored (GS), clinical stage T1 to T3a, no evidence of metastatic disease, prostate volume

Results: From 1997 to 2005 there were 273 patients treated with this treatment combination at AC Camargo Cancer Center, Sao Paulo, Brazil. The median age and FU time were 64.7 and 10.3 years, respectively. Two hundred thirteen (78.0%) patients had FU longer than 5 years. Actuarial 10-year overall survival (OS), Clinical Specific Survival (CSS) and BC were 89.8%, 63.6% and 71.8%, respectively. On univariate analysis GS<7, clinical stage<T2b, low risk group (LR), absence adjuvant androgen deprivation (ADT), age>65, PSAi<10, localized EBRT and 3D-HDR plan were associated with improved CSS and BC, excluding PSAi, age for the last one. Multivariate Cox regression analysis confirmed LR, GS<7, PSAi<10, absence of ADT, age

Conclusion: The present data represents a unique uni-institutional study at long FU for the given technique. A comparison with the current literature confirms the excellent results achieved with this treatment modality. HDR has also the advantage of treatment time reduction and increasing in the capability of work load of the linear accelerators, especially in developing countries, where waiting lists and lack of radiation oncology facilities are a reality.

Keywords

prostate cancer, radiotherapy, high-dose rate brachythrerapy, biochemical control, PSA

Abbreviations and Acronyms

ADT: Adjuvant Androgen Deprivation
AJCC: American Joint Committee on Cancer
BC: Biochemical Control
CSS: Clinical Specific Survival
EBRT: External Beam Radiotherapy
FU: Fallow Up
GS: Gleason Score
HDR: High-Dose-Rate Brachytherapy
LR: Low Risk Group
NAAD: Neoadjuvant Hormonal Therapy
OS: Overall Survival
PSAi: Initial Prostate-Specific Antigen
TRUS: Trans Rectal Ultrasound

Introduction

More than 62% of Prostate Cancers (PCa) are diagnosed in men over 65 years. It has become a public health and socioeconomic problem with increasing incidence, in special due to a rapidly aging population worldwide [1]. In Brazil it was expected the diagnosis of 61,200 new cases of PCa in 2016, and the crude mortality for 2013 was around 14,000 deaths [2].

Management options for localized and locally advanced PCa are controversial and include active surveillance, radical prostatectomy, external beam radiotherapy (EBRT) and brachytherapy with low or high dose rate sources. [3]

Several studies provided evidence for the efficacy of dose-escalation on biochemical control (BC) of PCa. Mature results from randomized trials show a direct relation between increasing the radiation dose given to the prostate and/or seminal vesicles and BC [4-7].

High-dose-rate after loading brachytherapy (HDR) is one method that can deliver a high localized radiation dose to the tumor with excellent BC when combined to EBRT [8]. One prospective randomized trial with up to 10 years follow up has proved that HDR plus EBRT is more efficient than EBRT alone in terms of BC with less acute rectal toxicity and improved quality of life [9].

The aim of this retrospective study is to evaluate the mature results of patients with local and locally advanced PCa treated with combination of HDR and EBRT.

Materials and Methods

Patients with confirmed histological diagnosis Gleason scored (GS) of PCa, AJCC clinical stage T1 to T3a, with no evidence of metastatic disease and initial PSA <60mg/ml, prostate volume

This single-centre institutional protocol of treatment was performed in compliance with the Declaration of Helsinki and approved by the local research Ethics Committee. Written informed consent was mandatory.

External Beam Radiotherapy

The EBRT target volume was defined using diagnostics CT images on conventional two dimensional or 3D planning. The targets were the prostate gland and the proximal seminal vesicles with a 1 to 1.5 cm margin except to the posterior region, which margins were reduced to 0.5 to 1.0cm. The EBRT dose ranged from 45 to 54 Gy prescribed to the intersection point. Further details of the radiotherapy schedules have been published previously [10].

High Dose Rate Brachytherapy

HDR was done under spinal anesthesia. Using TRUS with a perineal template affixed to perineum the exact needles positions were determined intraoperatively. In a first moment treatments were planned based on semi-orthogonal X-rays – two dimensional planning (2D) – and after that we moved to three dimensional (3D) planning, based on CT images. The prostate gland, the rectum, and the urethral trajectory and length were countered and identified in both situations. Implant dosimetry geometric optimization was initially utilized, followed later by use of inverse planning. Treatment parameters and dose constraints changed minimally throughout the years. The patients considered low risk had 16 Gy given in 4 fractions BID, one single implant. Intermediate and high risk patients had 20 Gy given in the same treatment schedule. The dose-volume histogram constraints were as follows: the TRUS or CT-based prostate´s volume receiving 100% of the dose (V100) should be >95%, the uniformity index should be more than 50%, and the V150 less than 30%. The urethra maximum punctual and the maximal dose to 1cc of anterior rectal wall should not exceed 135% and 75% of prescribed doses, respectively.

Definition of end points and statistical analysis

BC was measured using PSA tests and assessed according to the Phoenix definitions [11]. Clinical Specific Survival (CSS) was calculated from the start of treatment to the lost of BC, diagnose of metastatic disease or death from PCa. The BC was evaluated from the date of start the treatment until date of first biochemical failure. The follow up (FU) program also included clinical investigation, digital rectal examination and image studies.

The statistical program SPSS (statistical package for the social sciences) Inc., released 2008, Statistics for Windows, version 20.0 (SPSS Inc., Chicago, IL) was used for all statistical analysis. The analysis of OS, CSS and BC was made using the Kaplan–Meier method. The log-rank test was used to test the significance when comparing different subgroups. Univariate and multivariate Cox regression analysis were also performed. The alpha level considered for statistically significant differences was 0.05.

Results

Between March, 1997 and March, 2005 there were 305 patients treated with combination of HDR and EBRT at the Department of Radiation Oncology, AC Camargo Cancer Center, Sao Paulo, Brazil. Thirty two patients were lost of FU and the data of 273 patients was available for analysis. Sixty four (27.1%) patients had pelvic EBRT and the remaining 209 (76.5%) localized EBRT. Clinical and treatments characteristics are depicted in Tables 1 and 2.

Table 1. Patients Characteristics

Median Range Variable n %
Age (years) 64.7 42-82
Prostate Vol (cc) 36.3 19-72 <35 121 44.3
>35 152 55.7
PSAi (ng/ml) 10.3 1-52 <10 173 16.8
10-20 54 19.8
>20 46 63.4
Gleason Score <7 190 69.6
=7 58 21.2
>7 25 21.2
Yes 47 17.2
No 226 82.8
Clinical Stage <T2b 192 70.3
T2b-c 47 17.2
>T2c 34 12.5
Risk Group Low 133 48.7
Interm 76 27.8
High 64 23.4
ADT NAAD Yes 93 34.1
No 180 65.9
ADJ 91 33.3
Salvage 37 13.6
WO 145 53.2
EBRT Pelvic 64 23.4
Localized 209 76.6
Comorbidities No 146 53.5
SAH 42 15.4
Diabetes 19 7.0
Other 39 14.3
TOTAL 273 100.0

Legend: ADJ (adjuvant hormonal therapy), ADT (Androgen deprivation therapy), BF (Bichemical failure, EBRT (External beam radiotherapy), NAAD (neoadjuvant hormonal therapy), SAH (Systemic arterial hypertension), Salvage (Salvage hormonal therapy)

Table 2. Treatment  Characteristics

Median Range Variable n %
Dose EBRT 50 40-54 < 50 149 54.6
>50 124 45.4
HDR 16 133 48.7
18.3 16-20 20 140 51.3
HDR plan 2D 167 61.2
3D 106 38.8
Interval 18.5 9-61 <18 173 63.4
>18 100 36.6

Legend: EBRT (External beam radiotherapy), HDR (High-dose-rate brachytherapy), HDR plan 2D/3D (two or three dimensional planning)

The median age and FU time were 64.7 (range, 42-82) and 10.3 (range, 1-15) years, respectively. Two hundred thirteen (78.0%) patients had FU longer than 5 years, and of these 153 (56.1%) longer than 10 years.

Androgen deprivation therapy

Androgen deprivation therapy (ADT) in a short course neo-adjuvant ADT, was prescribed for less than 6 months. Neoadjuvant hormonal therapy (NAAD) was administered to 34.1% of the patients. Adjuvant hormonal therapy (ADJ) was observed, mostly, for intermediate and high risk patients (33.4%), generally for no more than 6 months for intermediate risk and up to 3-years in high risk patients. Salvage ADT was observed in 37 (90.2%) of 41 patients dead due PCa. The profile of hormonal therapy is shown in Tables 3 and 4.

Table 3. Neoadjuvant Hormonal therapy according to Risk Group – Risk NAAD Cross tabulation

NAAD
 Risk Group WO % YES  % Total %
Low 74 27.1 8 2.9 82 30.0
Interm 65 23.8 35 12.8 100 36.6
High 41 15.0 50 18.3 91 33.3
             Total 180 65.9 93 34.1 273 100

Legend: ADJ (adjuvant hormonal therapy), Interm (intermediate), NAAD (neoadjuvant hormonal therapy), WO (without hormonal therapy)

Table 4. Adjuvant and Salvage Hormonal therapy according to Risk Group

WO   % ADJ   % Salv    % Total    %
Low 72 26.4 7 2.6 3 1.1 82 30.0
Interm 57 20.9 34 12.5 9 3.3 100 36.6
High 16 5.9 50 18.3 25 9.2 91 33.3
Total 145 53.2 91 33.4 37 13.6 273 100

Legend: ADJ (adjuvant hormonal therapy), Interm (intermediate), NAAD (neoadjuvant hormonal therapy), Salv (Salvage hormonal therapy), WO (without hormonal therapy)

The crude 10-year overall survival (OS) rate at was 52.7%. Actuarial 5- and 10-year OS, CSS and BC were 80.1%, 89.8%, 83.7%, 63.6%, 85.5% and 71.8%, respectively. (Figures 1-3)

Figure 1- Overall Survival

    Figure 1. Overall Survival

Figure 2 - Clinical Specific Survival

Figure 2. Clinical Specific Survival

Figure 3 - Clinical Specific Survival by Risk Group

Figure 3. Clinical Specific Survival by Risk Group

Univariate and multivariate analysis

On univariate analysis GS <7, clinical stage <T2b, low risk group, absence adjuvant ADT, older age (>65-years), PSAi Univariate analysis failed to identify neoadjuvant androgen deprivation therapy (NAAD) as a predictor for BC in all group risks (p=ns). When we pooled the intermediate and high risk group into a unique denominated unfavorable risk group, NAAD also failed to predict improved CSS and BC.

Multivariate Cox regression analysis confirmed low risk group (HR 0.03, 95% CI 0.006-0.116, p<0.001) and intermediate risk (HR 0.09, 95% CI 0.042-0.216, p<0.001) compared to high risk, presence of ADT (HR 0.39, 95% CI 0.179-0.868, p=0.021) as favorable predictors for CSS. GS >7 (HR 3.24, 95% CI 1.279-8.220, p<0.001), PSA >10 (HR 6.19, 95% CI 2.015-19.041, p=0.001), age >65 years (HR 2.87, 95% CI 1.264-6.504, p=0.012) and EBRT dose >50 Gy (HR 14.50, 95% CI 1.874-112.164, p=0.010) were confirmed as adverse predictors for CSS. Tables 6-8, Figures 4-9.

Low risk group compared to intermediate, (HR 0.70, 95% CI 0.023-0.213, p<0.001) and high risk (HR 0.99, 95% CI 0.052-0.190, p<0.001) groups, was a favorable predictive factor for BC. GS >7 (HR 3.09, 95% CI 1.473-6.473, p=0.003) and PSAi >10 (HR 6.18, 95% CI 2.331-16.393, p<0.001) were negative predictive factor for BC.

Low risk group was confirmed as the only predictive factor for OS when compared to intermediate (HR 0.28, 95% CI 0.133-0.608, p=0.001) and high (HR 0.38, 95% CI 0.223-0.665, p=0.001) risk groups.

Figure 4. Clinical Specific Survival by Clinical Stage

Figure 4. Clinical Specific Survival by Clinical Stage

Figure 5. Clinical Specific Survival by Gleason Score

Figure 5. Clinical Specific Survival by Gleason Score

Figure 6. Clinical Specific Survival by Initial PSA

Figure 6. Clinical Specific Survival by Initial PSA

Figure 7. Hazard Plots - Clinical Specific Survival by Risk Group

Figure 7. Hazard Plots – Clinical Specific Survival by Risk Group

Figure 8. Biochemical Control

Figure 8. Biochemical Control

Figure 9. Hazard Plots - Biochemical Control by Risk Group

Figure 9. Hazard Plots – Biochemical Control by Risk Group

Table 6. Cox regression for CSS

B SE Wald df Sig. Exp(B) 95.0% CI for Exp(B)
Lower Upper
LR 44.294 2 .000
IR -3.641 .761 22.910 1 .000 .026 .006 .116
HR -2.351 .418 31.579 1 .000 .095 .042 .216
EBRT

Pelvic x Local

-.110 .312 .124 1 .724 .896 .486 1.651
2D x 3D -1.861 1.134 2.693 1 .101 .156 .017 1.436
ADT -.931 .403 5.336 1 .021 .394 .179 .868
PSAi <10 10.858 2 .004
PSAi (>10<20) 1.925 .661 8.481 1 .004 6.856 1.877 25.048
PSAi (>20) 1.824 .573 10.133 1 .001 6.195 2.015 19.041
GS <7 7.328 2 .026
GS = 7 .335 .709 .223 1 .636 1.398 .349 5.608
GS >7 1.176 .475 6.145 1 .013 3.243 1.279 8.220
CS <T2b 3.350 2 .187
CS T2b/T2c -.662 .551 1.443 1 .230 .516 .175 1.519
CS >T2c .464 .818 .321 1 .571 1.590 .320 7.895
Age <65y 1.053 .418 6.352 1 .012 2.867 1.264 6.504
EBRT <50Gy 2.674 1.044 6.562 1 .010 14.498 1.874 112.164
HDR dose <20Gy 1.763 1.023 2.973 1 .085 5.830 .786 43.261
Interval EBRT to HDR -.187 .303 .381 1 .537 .829 .458 1.502

Legend: 2D (two dimensional plan), 3D (tridimensional plan), ADT (Androgen deprivation therapy), BC (biochemical control), Comorb (Comorbidites), CS (Clinical stage), CSS (Clinical Specific Survival), EBRT (External beam radiotherapy), GS (Gleason score), IR (Intermediate risk group), HDR (High-dose-rate brachytherapy), HR (High risk group), LR (Low risk group), NAAD (neoadjuvant hormonal therapy), OS (overall survival), P vol. (Prostate volume cc)

Table 7. Cox regression for BC

B SE Wald df Sig. Exp(B) 95.0% CI for Exp(B)
Lower Upper
LR 62.053 2 .000
IR -2.660 .568 21.948 1 .000 .070 .023 .213
HR -2.309 .330 48.920 1 .000 .099 .052 .190
2D x 3D -.789 .656 1.448 1 .229 .454 .126 1.642
ADT -.455 .307 2.192 1 .139 .634 .347 1.159
PSAi <10 13.634 2 .001
PSAi (>10<20) 1.822 .498 13.403 1 .000 6.182 2.331 16.393
PSAi >20 1.346 .462 8.494 1 .004 3.843 1.554 9.502
GS <7 8.944 2 .011
GS =7 .957 .465 4.240 1 .039 2.603 1.047 6.469
GS >7 1.127 .378 8.915 1 .003 3.088 1.473 6.473
CS <T2b 2.825 2 .244
CS T2b/T2c -.647 .410 2.493 1 .114 .523 .234 1.169
CS >T2c -.306 .580 .278 1 .598 .737 .236 2.294
EBRT 50Gy 1.411 .550 6.590 1 .010 4.101 1.396 12.044
HDR dose <20Gy -.470 .680 .477 1 .490 .625 .165 2.370
Interval EBRT to HDR -.064 .230 .078 1 .780 .938 .597 1.473

Legend: 2D (two dimensional plan), 3D (tridimensional plan), ADT (Androgen deprivation therapy), BC (biochemical control, Comorb (Comorbidites), CS (Clinical stage), CSS (Clinical Specific Survival), EBRT (External beam radiotherapy), GS (Gleason score), IR (Intermediate risk group), HDR (High-dose-rate brachytherapy), HR (High risk group), LR (Low risk group), NAAD (neoadjuvant hormonal therapy), OS (overall survival), P vol. (Prostate volume)

Table 8. Cox regression for OS

B SE Wald df Sig. Exp(B) 95.0% CI for Exp(B)
  Lower Upper
ADJ .149 0.3 0.3 1 .580 1.161 .684 1.970
Age <65y .223 0.2 1.0 1 .322 1.250 .804 1.942
Comorbidity -.310 0.2 2.1 1 .149 .734 .482 1.117
EBRT

Pelvic x Local

-.089 0.2 0.1 1 .722 .915 .560 1.494
LR 14.3 2 .001
IR -1.259 0.4 10.5 1 .001 .284 .133 .608
HR -.954 0.3 11.7 1 .001 .385 .223 .665

Legend: ADJ (adjuvant hormonal therapy, EBRT (External beam radiotherapy), IR (Intermediate risk group), HDR, HR (High risk group), LR (Low risk group), OS (overall survival)

Discussion

Conventional EBRT to treat PCa is securely limited to doses of 64–70 Gy in 1.8–2.0 Gy fractions. These levels of doses are determined by the risk of long-term toxic effects to the bladder and rectum. The clinical and biochemical relapse rates associated with these dose levels are around 33% within 5 years. Peeters et al 12 published the results of a randomized trial comparing total doses of 68 Gy and 78 Gy using EBRT alone. The 5-year OS in the higher dose arm of that study was 83% using ASTRO definition and the BC was significantly better in the 78-Gy arm compared to the 68-Gy arm, with an adjusted hazard ratio of 0.74 (p=0.02). Other published reports using dose-escalated photon beam EBRT alone also point in the same direction with respect to their long-term BC results [13-14].

HDR can escalate the dose given to the prostate by the combination with EBRT, and further more, in locally advanced disease has also the possibility of including the seminal vesicles when they needed to be encompassed. HDR has also a potential biological advantage through the delivery of high doses per fraction [10]. It is important to note that the comparisons between series published are difficult due differences in the techniques and planning for both, EBRT and HDR.

The combination of values of PSAi, GS and CS to identify a more or less aggressive disease has being extensively discussed in the literature, and was confirmed by this study. The most frequent challenge is to identify, for example the indication of prostate versus pelvic EBRT, varying risk categories, absence or use of NAAD, ADT and their length. Despite this, the combination of HDR and EBRT provides an optimal modulation of dose delivery. Results of this combination, in terms of BC and with more than 5 years of FU, range from 57% to 100% according to the risk group for biochemical failure (Table 9).

Table 9. Results of biochemical control by risk groups in series of HDR plus EBRT with more than 5-year follow up

Reference n Median FU  (months) Biochemical control by risk groups (%)

Low       Intermediate       High

Dose in Gy

(HDR(n.fx)/EBRT)

13 344 61 84 74 19.5(3) / 46
14 121 63 91 10(1)/50
15 313 68 100 88 79 23(2)/46
16 229 61 95 90 57 21(3)/50.4
17 64 105 84 80 18(3)/45
18 90 95 80 16.5(3)/45
19 264 75 97 18(3)/45
20 100 62 84 82 10(1)/60
21 64 61 100 91 21(3)/50
22 196 66 86 18(3)/46
23 131 63 87 71 30(4)/45

The search for factors predicting BC and CSS is important on defining what patients should be treated more aggressively. We, as other authors [26-28] observed that PSAi <10 ng/ml was confirmed as a favorable predictive factor related to BC and CSS.

Age<65 years was found to be an adverse prognostic factor in our analysis for CSS, with a marginally statically significance impact on OS, not confirmed on multivariate analysis. Smolska-Ciszewska et al, conversely to our results, noted that younger age at time of treatment impacted only on OS, not explaining if there was any association between treatment and side effects or worsening of associated comorbidities. As in our analysis, they found that low risk group and association of HDR to EBRT correlated with improved BC [29].

As in our results, Kamrava et al found that T stage, GS, and use of ADT were significantly associated with CSS on univariate analysis, but on multivariate analysis only GS and use of ADT were significantly associated CSS [30]

It is expected that the grouping of the patients in risk groups for biochemical failure based on PSAi, GS and CS, to aggregate patients with adverse features in the intermediate and high risk, leading to worse CSS and BC for the two last one, what was confirmed in our analysis. This was also observed by Morris et al [31].

In our series, as in others, patients who received adjuvant ADT had significantly higher risk features suggesting patient selection bias for CSS in this group of patients, instead of a negative interaction between HDR and EBRT [31-32].

Mature data in the literature evaluated the 10-year outcomes of intermediate- and high-risk patients noting a clear dose response by increasing the dose escalation through HDR doses [27]. More recently the use of Intensity modulated radiation therapy combined to HDR has being investigated. Chen et al. published the results of 148 patients treated with HDR – 22 Gy in 4 fractions followed IMRT up to 50.4Gy. All patients with Gleason score of 8 or higher had ADT for 1 year. They noted a 4-year actuarial CSS of 96.8% and of 100%, 100% and 94% for low, intermediate and high risk, respectively [32].

The results of the first randomized prospective trial, which has addressed dose escalation using an HDR and EBRT is a trial with a relatively slow accrual rate. There are some critics that must be addressed as the changes in EBRT technique during the time of the study, and, by current standards, the control arm is a relatively low-dose treatment. Despite that, the study reported the results of 218 patients treated between 1997 and 2005. There were 108 patients assigned to EBRT alone and 110 patients treated by EBRT followed by HDR. They noted that CSS was significantly higher in patients treated with combined modality (p = 0.04). In multivariate analysis the category treatment modality and ADT were significant covariates for BC, but with no differences in OS, as observed in our study. After a median follow-up time of 10.5 year follow-up, an 18% increase in CSS was obtained relative to EBRT alone, reflecting a 31% reduction in the risk of recurrence (p = 0.01) and no evidence of an increase in long-term severe morbidity [9].

Surgically induced gland deformation is inevitable during brachytherapy procedures. We observed that the use more of advanced methods of images (3D plan based on CT, MR or TRUS) images to identify the target, organs at risk and needles is important and have already been reported to impact on BC and CSS [33]. In our analysis Multivariate Cox regression analysis confirmed EBRT dose >50 Gy as predictor for CSS and BC, but with no impact on OS. This may be explained by the relative low dose per fraction schedule used in both groups. Of importance is to note that the dose given by HDR was relative constant thorough the risk groups, leading to higher biological effective dose to intermediate and high risk patients, and even though, these patients had a worse outcome, showing that there is space for further studies of dose escalation or treatment combination. After 2005 with the introduction of real time TRUS image acquisition and planning we have changed our protocol and moved forward for a more intense dose escalation, increasing dose and reducing the number of fractions.

Other information of this study is that presence of NAAD had no impact on DSS, BC or OS in any risk group, showing that this treatment strategy should be reserved for downsizing the prostate prior to treatment, in special for low risk patients.

In conclusion, this report demonstrates that HDR combined with EBRT is an important and effective method in achieving dose escalation in the radical radiotherapy of PCa. This combination has also the advantage of treatment time reduction and in increasing in the capability of work load of the linear accelerators, especially in developing countries, where waiting lists and lack of radiation oncology facilities are a reality. The present data represents a unique uni-institutional study at long FU for the given technique, and a comparison with the current literature confirms the excellent results achieved with this treatment modality. The satisfactory BC, CSS and OS are probably result of improved LC achieved with dose escalation, showing that HDR is an optimal alternative method of local dose escalation when combined to EBRT.

References

  • Quinn MJ, d’Onofrio A, Møller B, Black R, Martinez-Garcia C, et al. (2003) Cancer mortality trends in the EU and acceding countries up to 2015. Ann Oncol 14: 1148-1152. [crossref]
  • http://www.inca.gov.br/estimativa/2014/sintese-de-resultados- comentarios.asp
  • Novara G, Ficarra V, Mocellin S, et al. (2012) Systematic review and meta-analysis of studies reporting oncologic outcome after robot-assisted radical prostatectomy. Eur Urol 62: 382.
  • Al-Mamgani A, van Putten WL, Heemsbergen WD, van Leenders GJ, Slot A, et al. (2008) Update of Dutch multicenter dose-escalation trial of radiotherapy for localized prostate cancer. Int J Radiat Oncol Biol Phys 72: 980-988. [crossref]
  • Dearnaley DP, Sydes MR, Graham JD, et al. (2007) Escalated-dose versus standard-dose conformal radiotherapy in prostate cancer: first results from the MRC RT01 randomized controlled trial. Lancet Oncol 8: 475.
  • Kuban DA, Tucker SL, Dong L, Starkschall G, Huang EH, et al. (2008) Long-term results of the M. D. Anderson randomized dose-escalation trial for prostate cancer. Int J Radiat Oncol Biol Phys 70: 67-74. [crossref]
  • Zietman AL, DeSilvio ML, Slater JD, et al. (2005) Comparison of conventional-dose vs high-dose conformal radiation therapy in clinically localized adenocarcinoma of the prostate: a randomized controlled trial. JAMA 294: 1233.
  • Pellizzon AC, Salvajoli JV, Maia MA, et al. (2004) Late urinary morbidity with high dose prostate brachytherapy as a boost to conventional external beam radiation therapy for local and locally advanced prostate cancer. J Urol 171: 1105.
  • Hoskin PJ, Rojas AM, Bownes PJ, et al. (2012) Randomized trial of external beam radiotherapy alone or combined with high-dose-rate brachytherapy boost for localized prostate cancer. Radiother Oncol 103: 217.
  • Pellizzon AC, Nadalin W, Salvajoli JV, Fogaroli RC, et al. (2003) Results of high dose rate afterloading brachytherapy boost to conventional external beam radiation therapy for initial and locally advanced prostate cancer. Radiother Oncol 66: 167.
  • Roach M 3rd, Hanks G, Thames H Jr, et al. (2006) Defining biochemical failure following radiotherapy with or without hormonal therapy in men with clinically localized prostate cancer: recommendations of the RTOG-ASTRO Phoenix Consensus Conference. Int J Radiat Oncol Biol Phys 65: 965.
  • Peeters ST, Heemsbergen WD, Koper PC, et al. (2006) Dose-response in radiotherapy for localized prostate cancer: results of the Dutch multicenter randomized phase III trial comparing 68 Gy of radiotherapy with 78 Gy. J Clin Oncol
  • Pollack A, Zagars GK, Starkschall G, Antolak JA, Lee JJ, et al. (2002) Prostate cancer radiation dose response: results of the M. D. Anderson phase III randomized trial. Int J Radiat Oncol Biol Phys 53: 1097-1105. [crossref]
  • Dearnaley DP, Sydes MR, Graham JD, et al. (2007) Escalated dose versus standard-dose conformal radiotherapy in prostate cancer: first results from the MRC RT01 randomized controlled trial. Lancet Oncol 8: 475.
  • Khor R, Duchesne G, Tai KH, et al. (2013) Direct 2-arm comparison shows benefit of high-dose-rate brachytherapy boost vs external beam radiation therapy alone for prostate cancer. Int J Radiat Oncol Biol Phys 85: 679.
  • Cury FL, Duclos M, Aprikian A, et al. (2012) Single-fraction high-dose-rate brachytherapy and hypofractionated external beam radiation therapy in the treatment of intermediate-risk prostate cancer – long term results. Int J Radiat Oncol Biol Phys 82: 1417.
  • Prada PJ, González H, Fernández J, Jiménez I, Iglesias A, et al. (2012) Biochemical outcome after high-dose-rate intensity modulated brachytherapy with external beam radiotherapy: 12 years of experience. BJU Int 109: 1787-1793. [crossref]
  • Kotecha R, Yamada Y, Pei X, Kollmeier MA, Cox B, et al. (2013) Clinical outcomes of high-dose-rate brachytherapy and external beam radiotherapy in the management of clinically localized prostate cancer. Brachytherapy 12: 44-49. [crossref]
  • Kaprealian T, Weinberg V, Speight JL, et al. (2012) High-dose-rate brachytherapy boost for prostate cancer: comparison of two different fractionation schemes. Int J Radiat Oncol Biol Phys 82: 222.
  • Savdie R, Symons J, Spernat D, Yuen C, Pe Benito RA, et al. (2012) High-dose rate brachytherapy compared with open radical prostatectomy for the treatment of high-risk prostate cancer: 10 year biochemical freedom from relapse. BJU Int 110 Suppl 4: 71-76. [crossref]
  • Aluwini S, van Rooij PH, Kirkels WJ, Jansen PP, Praag JO, et al. (2012) High-dose-rate brachytherapy and external-beam radiotherapy for hormone-naïve low- and intermediate-risk prostate cancer: a 7-year experience. Int J Radiat Oncol Biol Phys 83: 1480-1485. [crossref]
  • Agoston P, Major T, Fröhlich G, et al. (2011) Moderate dose escalation with single-fraction high-dose-rate brachytherapy boost for clinically localized intermediate- and high-risk prostate cancer: 5-year outcome of the first 100 consecutively treated patients. Brachytherapy 10: 376.
  • Ghadjar P, Keller T, Rentsch CA, Isaak B, Behrensmeier F, et al. (2009) Toxicity and early treatment outcomes in low- and intermediate-risk prostate cancer managed by high-dose-rate brachytherapy as a monotherapy. Brachytherapy 8: 45-51. [crossref]
  • Zwahlen DR, Andrianopoulos N, Matheson B, Duchesne GM, Millar JL (2010) High-dose-rate brachytherapy in combination with conformal external beam radiotherapy in the treatment of prostate cancer. Brachytherapy 9: 27-35. [crossref]
  • Viani GA, Pellizzon AC, Guimarães FS, Jacinto AA, dos Santos Novaes PE, et al. (2009) High dose rate and external beam radiotherapy in locally advanced prostate cancer. Am J Clin Oncol 32: 187-190. [crossref]
  • Aoki M, Miki K, Kido M, et al. (2014) Analysis of prognostic factors in localized high-risk prostate cancer patients treated with HDR brachytherapy, hypofractionated 3D-CRT and neoadjuvant/adjuvant androgen deprivation therapy (trimodality therapy). J Radiat Res 55: 527.
  • Martinez AA, Gonzalez J, Ye H, et al. (2011) Dose escalation improves cancer-related events at 10 years for intermediate- and high-risk prostate cancer patients treated with hypofractionated high-dose-rate boost and external beam radiotherapy. Int J Radiat Oncol Biol Phys 79: 363.
  • Shigehara K, Mizokami A, Komatsu K, Koshida K, Namiki M (2006) Four year clinical statistics of iridium-192 high dose rate brachytherapy. Int J Urol 13: 116-121. [crossref]
  • Smolska-Ciszewska B, Miszczyk L, Bialas B, et al. (2015) The effectiveness and side effects of conformal external beam radiotherapy combined with high-dose-rate brachytherapy boost compared to conformal external beam radiotherapy alone in patients with prostate cancer. Radiat Oncol 10: 366.
  • Kamrava M, Rwigema JC, Chung M, Banerjee R, Wang J, et al. (2013) Predictors of distant metastasis after combined HDR brachytherapy and external beam radiation for prostate cancer. J Contemp Brachytherapy 5: 127-133. [crossref]
  • Morris LM, Izard MA, Wan WY (2015) Does prostate-specific antigen nadir predict longer-term outcome es of prostate cancer after neoadjuvant and adjuvant androgen deprivation therapy in conjunction with brachytherapy? Brachytherapy 14: 322.outcome
  • Chen WC, Tokita KM, Ravera J, Fu P, Jiang Y, et al. (2013) Four-year outcomes of hypofractionated high-dose-rate prostate brachytherapy and external beam radiotherapy. Brachytherapy 12: 422-427. [crossref]
  • Pellizzon AC, Salvajoli J, Novaes P, et al. (2008) The relationship between the biochemical control outcomes and the quality of planning of high-dose rate brachytherapy as a boost to external beam radiotherapy for locally and locally advanced prostate cancer using the RTOG-ASTRO Phoenix definition. Int J Med Sci 5: 113.

Is Intravenous Iron Treatment a Risk for Infection?

DOI: 10.31038/IMROJ.2017214

Abstract

Background: Iron is a pro-oxidant and an essential nutrient for different bacteria. Experimental animal studies have demonstrated exacerbating sepsis episodes. Intravenous iron treatment has been implicated, at least theoretically in increasing infection episodes. However, these studies did not obtain any conclusion in this respect. Therefore, the aim of our study was to evaluate whether there is a risk between intravenous iron treatment and infection episodes following iron treatment.

Methods: In our study, 186 patients who received intravenous iron treatment were included. The biochemical parameters included “pre and post iron treatment” (e.g., ferritin, serum iron, transferring saturation, and hemoglobin) and the later emergence of infections. Moreover, we evaluated all possible risk factors that could interfere with the subsequence appearance of infection.

Results: Infectious complications were reported in 24 patients (12.8%) after the iron treatments began, and no infection was reported in 87.15% (n=162) of the patients. The most common infections were urinary (33%) and respiratory infections (25%). The most common infectious agents were enterobacteriae (e.g., E coli, E faecalis, and E faecium). Notably, we did not observe any intracellular pathogens. In our statistical survey, we did not find a relationship between infection onset and intravenous iron treatment. To analyze the baseline disease, comorbidities, such as diabetes and other treatment (e.g., corticosteroids or anti-TNF) were evaluated, and, no association was found.

Conclusion: In our study, intravenous iron was an effective treatment to correct anemia, and it did not appear to correlate with the development of secondary infections.

Key words

iron therapy; intravenous iron; iron infection

Abbreviations

IV – intravenous; HD – hemodialysis infection

Introduction

Iron is an important nutrient for many bacteria, and in laboratory animals, it was shown to exacerbate sepsis. [1] It has been postulated that increased plasma iron is responsible for the association of hemochromatosis with infections by Vibrio vulnificus, Yersinia enterocolitica, and Escherichia coli. [2] Accordingly, there are host defense mechanisms that tend to restrict the available iron from invading pathogens. [3] For example, the transferrin receptor is the major pathway for delivery of iron to peripheral tissues through endocytosis of its ligand, transferrin, which binds to iron and circulates in the plasma. Down regulation of the transferrin receptor limits the cell’s ability to acquire transferrin-bound iron and concomitantly reduces the endosomal pool of iron that can be accessed by intracellular pathogens, such as L. pneumophila, Mycobacterium tuberculosis, and Mycobacterium avium, and thus restricts their growth. [4, 5] Iron repletion with oral iron has been associated with adverse outcomes for certain infections. In a study of 137 iron-deficient Somali nomads, who were treated with placebo or oral iron (71 subjects), Murray et al. noted seven infection episodes in the placebo group and 36 in the oral iron group (including reactivation of preexisting malaria, brucellosis, and tuberculosis). [6] A large iron supplementation trial of young children on Pemba Island, was prematurely terminated as a result of an excess of serious adverse events (including deaths) in those receiving iron. [7] Ferric iron (Fe)–carbohydrate complexes are widely used for treating Fe deficiency in patients who are unable to meet their Fe requirements with oral supplements. [8] Intravenous (IV) Fe is generally well tolerated and effective in correcting Fe-deficient states. However, complexing Fe to carbohydrate polymers does not block its potent pro-oxidant effects; therefore, systemic free radical generation and, possibly, tissue damage may result. [9] On the other hand, excessive iron administration may lead to oversaturation of transferrin and the release of free, catalytically active iron into circulation (NTBI) [10] which could be readily utilized by bacteria, leading to their multiplication and the possibility of an overwhelming infection. [11, 12] The aim of the present work was to determine the short-term infection rate associated with IV iron treatment and the possible risk factors that can predict these infections in a contemporary patient cohort.

Material and methods

Patient enrollment A retrospective, longitudinal study was carried out at Marqués de Valdecilla University Hospital (Santander, Spain). The hospital records of patients who underwent ambulatory treatment with IV iron at the hospital day center between January 2011 and December 2012 were reviewed.

Patient parameter evaluation The medical records of these patients were searched after institutional review board approval was obtained for the following data fields: a) demographic features; b) principal diagnosis; c) comorbid factors, such as diabetes mellitus, arterial hypertension, and dislipemia; d) concomitant treatment as potential infection causes, such as corticosteroids, immunosuppresive therapy, and anti-TNF treatment; e) previous infection, and; f) laboratory data prior to and after IV iron treatment, such as hemoglobin concentration, mean cell hemoglobin (MCH), mean cell hemoglobin concentration (MCHC), red cell volume distribution width (RDW), serum iron, transferrin saturation, and serum ferritin.

Intravenous iron treatment One thusand mg (50 mg/ml) of ferric carboxymaltose was administered in an ambulatory regimen to the subjects at the hospital day center. The doses of this treatment were adapted subsequently according to the Ganzoni equation and their efficacy.

Case definitions We used a retrospective cohort design with a 1-month baseline period. After IV iron treatment was started, all of the patients were followed for 3 months. We looked for any infectious diseases, and we differentiated the subjects by bacterial, viral and fungal infections, as well as by gastrointestinal, genitourinary, respiratory and other diseases. If a single blood culture was positive for coagulase-negative staphylococci, Corynebacteria, or Bacillus sp., the culture was considered contaminated or representative of a transient bacteremia but not of a bacteremic episode. All other situations in patients in whom at least one blood culture was positive were defined as bacteremic episodes.

Statistical Analysis The results were analyzed with the SPSS 15.0 computer software package (Statistical Package for Social Sciences, Inc., Chicago. IL). Continuous variables were summarized as the means or as the medians and ranges. Categorical variables were compared with the chi-square and Fisher´s exact tests. Correlations between the data sets were examined using the Pearson (r) or Spearman rank (rs) correlation coefficients. Any differences were considered significant for all statistical tests at p values of less than 0.05.

Results

A total of 186 adult patients (70 men and 116 women; mean age, 63 yrs; range, 18 to 80 yrs) were enrolled. The principal diagnosis of the patients included in the study were cardiac diseases (n=41, 22%), neoplasm diseases (n=32, 17.2%), renal diseases (n=31, 16.7%), malnutrition due to bariatric surgery (n=19, 10.2%), hepatic diseases (n=15, 8.1%), and lung diseases (n=15, 8.1%) (Table 1). A comorbid condition known to increase the infection risk was found in 88 patients (47.3%), which included diabetes mellitus (n = 53), current malignancy (n = 32) (under treatment and/or not cured), being a transplant recipient (n= 3), and having both a malignancy and diabetes mellitus (n= 5). Twelve patients were treated with corticosteroids and six were treated with anti-TNF therapy. The pretreatment median hemoglobin level was 9.5 gr/dL (5.6- 12.6 gr/ dL) and after the treatment it was 11.8 gr/dL (7.4-14.1 gr/dL) (Table 2). Additionally, we found a statistically significant difference between the baseline biochemical iron metabolism parameters pre-and post IV iron treatment (Table 2). Thus, we corroborated that IV iron was an effective therapy in our patients.

Table 1. The principal diagnosis of the patients included in the study

Disease n (%)
Cardiac diseases 41 (22%)
Neoplasms diseases 32 (17.2%)
Kidney disease 31 (16.7%)
Bariatric surgery 19 (10.2%)
Liver diseases 15 (8.1%)
Lung diseases 15 (8.1%)
Others 33 (17.7%)

Table 2. The baseline laboratory iron metabolism parameters pre-and post-intravenous iron treatment

  Pre-treatment Post-treatment P
Hemoglobin (gr/dL) 9.5 (5.6-14.5) 11.8 (7.4- 14.10) <0.01
MCH (pg/RBC) 26.4 (8.6-37.5) 29.9 (21.8-37.4) <0.01
MCHC (gr/dL) 32.4 (28.6-35.3) 33.1 (25.5-35.3) <0.01
RDW (%) 18.9 (12.2-37.4) 21.4 (9-43.6) <0,01
Serum iron (mcg/dL) 25 (10-96) 69 (16-177) <0.541
Transferrin saturation (Fe/TIBC)% 7 (2-37) 23,5 (5-62) <0.01
Serum ferritin 54 (2-753) 408 (10-1492) <0.01

MCH – mean cell hemoglobin;
MCHC – mean cell hemoglobin concentration;
RDW – red cell volume distribution width.

No patients had more than one bacteremic episode within the month prior to treatment. Twenty-four patients (12.9%) suffered from one infection episode after the IV iron treatment, and 162 patients (87.1%) did not have any infection episodes. The majority of these infections were from bacterial microorganisms (75%, n=18) followed by unknown etiologies (16.7% n=4) and viral episodes (8.3%, n=2). There were no fungal infections. The urinary tract was involved in 33.3% of the cases (n=8); 25% of the cases involved therespiratory system (n=6), which was similar to the unknown focus case percentage; and 16.7% of the cases involved the gastrointestinal system (n=4). The infectious agent was unknown in 63.5% of the cases (n=15). The pathogens responsible for these episodes were E coli (16.7%, n=4) followed by E faecalis, E faecium, Lysteria and Proteus mirabilis, which all occurred in 4.2% of the cases (n=1). No intracellular microorganisms or non-capsulated bacteria were found in our series. Additionally, no serious diseases and no mortality cases were reported three months after the treatments began.

The main characteristics of the patients with and without infection episodes are shown in Table 3. More patients with an infection episode had a comorbidity (diabetes or malignancy) and ongoing immunosuppressive therapy than the patients without infections. However, this was not statistically significant. Furthermore, when we analyzed the relationship between infections with other comorbidities, such as diabetes, cancer, transplantation or treatments such as steroids or anti-TNF we did not find any association.

Table 3. Comorbidity incidences in the infection or no infection groups

  Infection No Infection p
Diabetes mellitus 8 (33.3%) 45 (28.3%) NS
Arterial hypertension 14 (58.3%) 65 (40.1%) NS
Dislipemia 5 (20.8%) 23 (14.2%) NS
Cardiac diseases 7 (29.2%) 34 (21%) NS
Renal diseases 7 (29.2%) 24 (14.8%) NS
Lung diseases 1 (4.2%) 14 (8.6%) NS
Liver diseases 1 (4.2%) 14 (8.5%) NS
Bariatric surgery 3 (12.5%) 16 (9.9%) NS
Neoplasm 7 (29.2%) 25 (15.4%) NS
Organ transplantation 0 (0%) 3 (1.9%) NS
Corticosteoids 3 (12.5%) 9 (5.6%) NS
Biologic therapy 1 (4.2%) 5 (3.1%) NS

NS – Non-Significant

Discussion

Almost all life forms require iron because of its involvement in basic cellular processes. Free iron was shown to potentiate bacterial growth in vitro. [13] In fact, during infections, pathogens use various means to acquire iron from their hosts, whereas hosts attempt to withhold it from pathogens. [14, 15] Iron therefore represents a point of conflict between the host and the pathogen, and an altered iron balance associated with poor outcomes in several infectious diseases, including malaria, [7] tuberculosis, [16] and HIV-1 infection. [17] Furthermore, certain bacterial species, including E. Coli, Klebsiella spp., and Salmonella spp., use unbound iron in the blood to enhance their growth. These organisms release siderophores, which are iron chelators, into the blood. Once these siderophores are released, bacteria can compete with proteins, such as transferrin for unbound iron in the serum. [18, 19] Staphylococcus aureus and Haemophilus influenzae do not possess siderophore; however, they do have transferrin receptors, which allow these bacteria to use iron for growth. [18] Vibrio vulnificus cannot grow if there is no iron available, and their virulence depends on the ability to obtain iron combined with transferrin. Hepcidin, which is produced during innate immune responses to infections, reduces the iron availability in the serum by inhibiting ferroportin function in macrophages and enterocytes and potentially limits extracellular pathogen growth therein. [20] Intravenous iron may also impair immune function and increase infection susceptibility. [21] Gupta et al. [22] found that exposure of mononuclear cells to IV iron agents induced significant intracellular oxidative stress and shortened CD4+ T lymphocyte survival. High doses of IV iron agents impaired the phagocytic activity and microbial killing capability of polymorphonuclear leukocytes [23-25]. Furthermore, in a recent in vitro study, iron sucrose led to impaired phagocytic function and increased polymorphonuclear leukocyte apoptosis. [26] However, the current evidence in clinical practice cannot determine whether iron supplementation increases the risk of infection or worsens outcomes due to infection. [27, 28] There is increasing data to suggest that infective and adverse-event risks may be related to the intensity and frequency of IV iron dosing. [29-31] Brookhart et al. [32] studied iron dosing patterns in a retrospective cohort of 117.050 prevalent hemodialysis (HD) patients and found that administration of large boluses of IV iron for repleting iron deficiency was associated with increased infection-related hospitalization or death compared with smaller doses of IV iron maintenance therapy. The risk of infection-related hospitalization was increased further in patients who experienced infections within the past month. Similarly, the DOPPS study showed a trend towards an increase infection.related mortality in prevalent HD patients treated with > 300 mg of IV iron. [33] A recent meta-analysis of randomized controlled trials evaluating IV iron use (often administered as frequent boluses) in patients with varying infective risk profiles found IV iron to be associated with a 30% greater risk of infection compared with oral or no iron therapy. [34] Conversely, a prospective observational study of 985 patients failed to demonstrate a relationship between infection and serum ferritin or IV iron dosing. [35] In our study, large IV iron boluses were an effective treatment for anemia that was caused by different etiologies, because hemoglobin increased at 2 or 3 points at the end of the treatment and we found any relationship between IV iron administration and infection, which was similar to the previously published data. Furthermore, in our study, transferrin saturation increased to 23.5%, thereby leaving little to no unbound iron available for bacterial utilization, as shown in previous in vitro studies. [10] For example, in the K. pneumonia case, bacterial suppression occurred throughout its incubation until the transferrin saturation exceeded 60%, at which time bacterial growth occurred. [19] Another in vitro study showed that the inhibition of Staphylococcus epidermidis growth was lost once the transferrin saturation exceeded 80%. [18] Subsequent studies had ambiguous results that left the relationship somewhat unclear. [36, 37] In a retrospective observational cohort study of 23.000 adult patients on HD hospitalized for bacterial infection, Ishida et al. found no association between iron treatment and readmission for infection. [38] Feldman’s group, in a re-analysis of their HD cohort using multivariable analysis, showed no statistically significant association between any level of iron administration and mortality. [39] Moreover, a French multicenter prospective study regarding bacteremia risk factors in HD patients (EPIBACDIAL) did not find any correlation between parenteral iron or ferritin and bacteremia. [40] Furthermore, in the Anker study, quality of life and functional status improved in heart failure (a relatively high-risk group) without an increase in infections [41].

Our study had limitations due to its retrospective nature. However, of the patients who were analyzed, we found a 12.8% infections after the beginning of iron therapy. Notably, an important difference in our study compared with the previously published studies was that we did not find any association between IV iron treatment and intracellular pathogens.

In conclusion, based on our study, iron treatment is safe and it does not associate with the development of new infections. However, the infection relationship is not clear and many studies are treating to investigate the different mechanisms regarding this issue. It is important to verify this result with other studies to improve the care and future treatment of patients because iron treatment is frequently used worldside. This knowledge can improve anemia management and avoid side effects. Thus, large multi-centered randomized controlled trials designed to evaluate both the short- and long-term safety of different IV iron dosing regimens are still required to determine the optimal iron therapy.

Conflict of interests

No conflict of interest exists and all authors had full access to all the data used in the study and take full responsibility for the decision to submit the manuscript for publication.

References

  • Guerinot ML (1994) Microbial iron transport. Annu Rev Microbiol 48: 743-772. [crossref]
  • Khan FA, Fisher MA, Khakoo RA (2007) Association of hemochromatosis with infectious diseases: expanding spectrum. Int J Infect Dis 11: 482-487. [crossref]
  • Wander K, Shell-Duncan B, McDade TW (2009) Evaluation of iron deficiency as a nutritional adaptation to infectious disease: an evolutionary medicine perspective. Am J Hum Biol 21:172-179.
  • Byrd TF, Horwitz MA (1989) Interferon gamma-activated human monocytes downregulate transferrin receptors and inhibit the intracellular multiplication of Legionella pneumophila by limiting the availability of iron. J Clin Invest 83:1457-1465.
  • Clemens DL, Horwitz MA (1996) The Mycobacterium tuberculosis phagosome interacts with early endosomes and is accessible to exogenously administered transferrin. J Exp Med 184:1349-1355.
  • Murray MJ, Murray AB, Murray MB, Murray CJ (1978) The adverse effect of iron repletion on the course of certain infections. Br Med J 2: 1113-1115. [crossref]
  • Sazawal S, Black RE, Ramsan M, Chwaya HM, Stoltzfus RJ, Dutta A, et al. (2006) Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: community-based, randomised, placebo-controlled trial. Lancet 367:133-1343.
  • Clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney Disease in adults (2006) Am J Kid Dis 47 (5 suppl3): S16-S85.
  • Zager RA (2006) Parenteral iron compounds: potent oxidants but mainstays of anemia management in chronic renal disease. Clin J Am Soc Nephrol 1 Suppl 1: S24-31. [crossref]
  • Parkkinen J, von Bonsdorff L, Peltonen S, Grönhagen-Riska C, Rosenlöf K. (2000) Catalytically active iron and bacterial growth in serum of haemodialysis patients after i.v. iron-saccharate administration. Nephrol Dial Transplant 15: 1827-1834.
  • Slotki I (2005) Intravenous iron supplementation in the anaemia of renal and cardiac failure–a double-edged sword? Nephrol Dial Transplant 20 Suppl 7: vii16-23. [crossref]
  • Bullen JJ, Rogers HJ, Spalding PB, Ward CG (2005) Iron and infection: the heart of the matter. FEMS Immunol Med Microbiol 43: 325-330. [crossref]
  • Fishbane S (1999) Review of issues relating to iron and infection. Am J Kidney Dis 34: S47-52. [crossref]
  • Weinberg ED (1984) Iron withholding: a defense against infection and neoplasia. Physiol Rev 64: 65-102. [crossref]
  • Armitage AE, Eddowes LA, Gileadi U, Cole S, Spottiswoode N, et al. (2011) Hepcidin regulation by innate immune and infectious stimuli. Blood 118: 4129-4139. [crossref]
  • Boelaert JR, Vandecasteele SJ, Appelberg R, Gordeuk VR (2007) The effect of the host’s iron status on tuberculosis. J Infect Dis 195: 1745-1753. [crossref]
  • McDermid JM, Jaye A, Schim van der Loeff MF, Todd J, Bates C, Austin S, et al. (2007) Elevated iron status strongly predicts mortality in West African adults with HIV infection. J Acquir Immune Defic Syndr 46:498-507.
  • Brewster UC, Perazella MA (2004) Intravenous iron and the risk of infection in end-stage renal disease patients. Semin Dial 17: 57-60. [crossref]
  • Cieri E (1999) Does iron cause bacterial infections in patients with end stage renal disease? ANNA J 26: 591-596. [crossref]
  • Rivera S, Nemeth E, Gabayan V, Lopez MA, Farshidi D, Ganz T (2005) Synthetic hepcidin causes rapid dose-dependent hypoferremia and is concentratedin ferroportin-containing organs. Blood 106:2196-2199.
  • Ishida JH, Johansen KL (2014) Iron and infection in hemodialysis patients. Semin Dial 27: 26-36. [crossref]
  • Gupta A, Zhuo J, Zha J, Reddy S, Olp J, Pai A (2010) Effect of different intravenous iron preparations on lymphocyte intracellular reactive oxygen species generation and subpopulation survival. BMC Nephrol 11:16.
  • Brewster UC (2006) Intravenous iron therapy in end-stage renal disease. Semin Dial 19: 285-290. [crossref]
  • Deicher R, Ziai F, Cohen G, Müllner M, Hörl WH (2003) High dose parenteral iron sucrose depresses neutrophil intracellular killing capacity. Kidney Int 64:728-736.
  • van Asbeck BS, Marx JJ, Struyvenberg A, Verhoef J (1984) Functional defects in phagocytic cells from patients with iron overload. J Infect 8: 232-240. [crossref]
  • Ichii H, Masuda Y, Hassanzadeh T, Saffarian M, Gollapudi S, Vaziri ND (2012) Iron sucrose impairs phagocytic function and promotes apoptosis in polymorphonuclear leukocytes. Am J Nephrol 36: 50-57.
  • Kliger AS, Foley RN, Goldfarb DS, Goldstein SL, Johansen K, et al. (2013) KDOQI US commentary on the 2012 KDIGO Clinical Practice Guideline for Anemia in CKD. Am J Kidney Dis 62: 849-859. [crossref]
  • Shah A, Roy NB2, McKechnie S3, Doree C4, Fisher SA4, et al. (2016) Iron supplementation to treat anaemia in adult critical care patients: a systematic review and meta-analysis. Crit Care 20: 306. [crossref]
  • Feldman HI, Santanna J, Guo W, Furst H, Franklin E, et al. (2002) Iron administration and clinical outcomes in hemodialysis patients. J Am Soc Nephrol 13: 734-744. [crossref]
  • Freburger JK, Ellis AR, Kshirsagar AV, Wang L, Brookhart MA (2014) Comparative short-term safety of bolus versus maintenance iron dosing in hemodialysis patients: a replication study. BMC Nephrol 15:154.
  • Agarwal R, Kusek JW, Pappas MK (2015) A randomized trial of intravenous and oral iron in chronic kidney disease. Kidney Int 88: 905-914. [crossref]
  • Brookhart MA, Freburger JK, Ellis AR, Wang L, Winkelmayer WC, et al. (2013) Infection risk with bolus versus maintenance iron supplementation in hemodialysis patients. J Am Soc Nephrol 24: 1151-1158. [crossref]
  • Bailie GR, Larkina M, Goodkin DA, Li Y, Pisoni RL, Bieber B, et al. (2015) Data from the Dialysis Outcomes and Practice Patterns Study validate an association between high intravenous iron doses and mortality. Kidney Int 87: 162-168.
  • Litton E, Xiao J, Ho KM (2013) Safety and efficacy of intravenous iron therapy in reducing requirement for allogeneic blood transfusion: systematic review and meta-analysis of randomized clinical trials. BMJ 347: f4822.
  • Hoen B, Paul-Dauphin A, Hestin D, Kessler M (2002) Intravenous iron administration does not significantly increase the risk of bacteriemia in chronic hemodialysis patients. Clin Nephrol 57:457-461.
  • Bansal A, Sandhu G, Gupta I, Kalahalli S, Nayak R, Zouain E, et al. (2014) Effect of aggressively driven intravenous iron therapy on infectious complications in end-stage renal disease patients on maintenance hemodialysis. Am J Ther 21:250-253.
  • Miskulin DC, Tangri N, Bandeen-Roche K, Zhou J, McDermott A, et al. (2014) Intravenous iron exposure and mortality in patients on hemodialysis. Clin J Am Soc Nephrol 9: 1930-1939. [crossref]
  • Ishida JH, Marafino BJ, McCulloch CE, Dalrymple LS, Dudley RA, et al. (2015) Receipt of Intravenous Iron and Clinical Outcomes among Hemodialysis Patients Hospitalized for Infection. Clin J Am Soc Nephrol 10: 1799-1805. [crossref]
  • Feldman HI, Joffe M, Robinson B, Knauss J, Cizman B, et al. (2004) Administration of parenteral iron and mortality among hemodialysis patients. J Am Soc Nephrol 15: 1623-1632. [crossref]
  • Hoen B, Paul-Dauphin A, Hestin D, Kessler M (1998) EPIBACDIAL: a multicenter prospective study of risk factors for bacteremia in chronic hemodialysis patients. J Am Soc Nephrol 9: 869-876. [crossref]
  • Anker SD, Comin Colet J, Filippatos G, Willenheimer R, Dickstein K, et al. (2009) Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med 361: 2436-2448. [crossref]

Platinum Plus Gemcitabine as the Most Effective Regimen in the First Line of Chemotherapy in Advanced Squamous Cell Lung Cancer

DOI: 10.31038/CST.2017212

Abstract

Purpose: Platinum-based doublet chemotherapy had been the standard first-line treatment for advanced NSCLC, regardless of histologic subtypes. We report overall survival (OS) and time to treatment failure (TTF) in patients with squamous cell lung cancer (SCC) receiving doublet of platinum.

Patients and Methods: Patients (N = 82) with advanced NSCLC received doublet of platinum. Sixty five (79,2%) patients were treated with a combination of platinum plus gemcitabine and 17 (20,7%) received microtubules inhibitor (3 patients were treated with vinorelbine, 3 patients with docetaxel and 11 patients with paclitaxel).

Results: Median TTF was 2,53 months CI95% [2,21 – 2,84] and median OS was 8,246 months CI95% [5,8 –2,6]. Regarding doublet of chemotherapy, in patients in which gemcitabine was used there was an improvement in TTF of 1,2 months (p= 0,107; log rank) and 4,75 months in OS (p= 0,018; log rank).

Conclusion: Gemcitabine plus platinum must be the chemotherapy of ghoice in advanced SCC. Randomized clinical trials with gemcitabine in advanced SCC are needed.

Keywords

squamous cell lung cancer, chemotherapy, gemcitabine

Introduction

Primary lung cancer is the most common malignancy and the first death related causes from cancer in the worldwide. Nowadays, it is the most important cause of cancer mortality in men and women. Lung cancer is still increasing both in incidence and mortality worldwide. In Spain, more than 21,000 men were diagnosed of lung cancer in 2012, while over 17,000 died. Lung cancer is the leading cause of dead among Spanish men. Figures in women were near 5,000 and more than 3,500 respectively. Women got into the habit of smoking some decades later than men in Spain [1]. Non-small-cell lung cancers (NSCLC) account for 85%–90% of lung cancers. NCSLC includes several histologic subtypes such as adenocarcinoma, squamous cell carcinoma (SCC), and large cell carcinoma. Platinum-based doublet chemotherapy had been the standard first-line treatment for advanced NSCLC, regardless of histologic subtypes [2].

New agents has been developed recently (anti folate, anti VEGF, antiEGFR, etc) [3]. These agents play a crucial role in first-line systemic therapy for nonsquamous histology while they do not have activity in SCC.

On the other hand, several regimens of platinum-based doublet chemotherapy are currently the standard first-line therapy for advanced lung SCC, including platinum combined with gemcitabine, docetaxel, paclitaxel, or vinorelbine and these scheludes have similar effectiveness [4].

There are not any phase III studies focused on determining what is the most active platinum-based chemotherapy regimen to treat advanced lung SCC. This study examined the comparative effectiveness of various platinum-based regimens as first-line therapy for advanced lung SCC.

Material and Methods

Patients

This is a retrospective study. The study population included patients with newly diagnosed lung SCC from 2012 to 2014 in the University General Hospital of Ciudad Real (Spain). The following inclusion criteria were used to identify eligible patients: (1) pathologically proven initial diagnosis of lung SCC as the single primary cancer; (2) age ≥ 18 years; and (3) advanced disease stage at diagnosis, which was defined as stage IIIB or stage IV disease according to the American Joint Committee on Cancer, 7th edition.5 Patients who underwent surgery during the first course of treatment and those who underwent radiotherapy with curative intent (which was defined as a cumulative dose > 50 Gy) were excluded.

All patients received chemotherapy for advanced lung SCC. Main regimens considered in our study including cisplatin (P), carboplatin (CP), gemcitabine (G), docetaxel (D), paclitaxel (T), or vinorelbine (V). Platinum agent was considered and patients were classified into patients who received cisplatin or carboplatin.

Objective

The main objective was overall survival (OS). OS was determined according to the date of diagnosis of advanced lung SCC to the date of death.

Statistical analysis

Baseline demographic and clinical variables were summarized with descriptive statistics. Regard chemotherapy, G were recoded as antimetabolite and D, T, V as microtubules inhihitor (MI).

The OS was estimated using the Kaplan-Meier method, and the differences between the study groups were compared using the log-rank test. The Cox proportional hazard model was used to estimate the univariate or adjusted hazard ratios and associated 95% confidence intervals for detecting differences in the effects of treatments on overall mortality. The sex, age, brain metastases and type of platinum were adjusted in the Cox proportional hazard model. Subgroup analyses defined according to sex, age (< 70 or ≥ 70 years) and platinum were performed as sensitivity analyses to determine whether the differences in effects on mortality of platinum + antimetabolite compared with those of P + MI. Two-sided P values of ≤ .05 were considered statistically significant. All analyses were performed by SPSS for windows v. 18.

Results

Baseline characteristics

Eighty two patients were included in this study. Among them, 79 (96.3%) were men, 41 (50%) were aged ≥ 70 years, and 2 (2,7%) had brain metastases (Table 1). Sixty five (79,2%) patients were treated with a combination of platinum plus gemcitabine and 17 (20,7%) received microtubules inhibitor (3 patients were treated with vinorelbine, 3 patients with docetaxel and 11 patients with paclitaxel). Patients aged < 70 years were more likely to receive chemotherapy with microtubules inhibitors than older patients (70,6% vs. 44,6%, P = 0.057). Up to December the 31th, 2014, 66 patients (80,5%) were with progression disease, 68 (82,2%) had died and 45 patients (54,9%) were controlled by the Palliative Care Unit. The median follow-up time was 6 months (table 1).

Table 1. Baseline characteristics of patients with advanced lung squamous cell carcinoma in our series

 

Baseline Characteristics

Gemcitabine

 

Microtubule inhibitor

 

All p
N (%) 65 (79,2%) 17 (20,7%) 82 (100%)
Age (median, range) 71 (44-88) 64 (40-81) 70 (40-84) 0,118
Years (n, %)

<70 años

>70 años

 

29 (44,6)

36 (55,4)

 

12 (70,6)

5 (29,4)

 

41 (50,0)

41 (50,0)

 

0,057

Gender (n, %)

Male

Female

 

63 (96,9)

2 (3,1)

 

16 (94,1)

1 (5,9)

 

79 (96,3)

3 (3,7)

 

0,583

Brain Metastases (n, %)

No

Yes

 

63 (96,9)

2 (3,1)

 

17 (100,0)

0 (0,0)

 

80 (97,6)

2 (2,4)

 

0,464

Platinium (n,%)

Cisplatin

Carboplatin

 

21 (32,3)

44 (67,7)

 

3 (17,6)

14 (82,4)

 

24 (29,3)

58 (70,7)

0,237
Progression first line (n, %)

No

Yes

 

11 (16,9)

54 (83,1)

 

5 (29,4)

12 (70,6)

 

16 (19,5)

66 (80,5)

0,247
Second line  (n, %)

Taxanes

Gemcitabine

TKI

27 (41,5)

21 (77,7)

0 (0,0)

6 (22,2)

  4 (23,5)

2 (50,0)

1 (25,0)

1 (25,0)

31 (37,8)

23 (74,19)

1 (3,3)

7 (22,5)

0,173
Control by Palliative Care Unit (n, %)

No

Yes

 

29 (44,6)

36 (55,4)

 

8 (47,1)

9 (52,9)

 

37 (45,1)

45 (54,9)

0,857
Estatus

Alive

Exitus

 

12 (18,5)

53 (81,5)

 

2 (11,8)

12 (88,2)

 

14 (17,1)

68 (82,9)

0,514

In patients, the main causes of death were progression disease in 59 patients (86,7%), pulmonary embolism in 2 patients (2,9%), stroke in 1 patient (1,47%), chronic obstructive pulmonary disease in 3 patients (4,4%), myocardial infarction in 1 patient (1,47%), hemoptysis in 1 patient (1,47%) and chemotherapy related toxicity in 1 patient (1,47%) (Figure 1).

Figure 1. main causes of death of patients

Figure 1. main causes of death of patients

Time to treatment failure

Median TTF was 2,53 months CI95% [2,21 – 2,84] (Figure 2). Considering age, sex, brain metastases, platinum compound and doublet of chemotherapy, we did not find differences (Table 2). Regarding doublet of chemotherapy, in patients in which gemcitabine was used there was an improvement in TTF of 1.2 months (p= 0.107, log rank) (Figure 3).

Figure 2. Median time to treatment failure in all patients

Fig 2.Median time to treatment failure in all patients

Figure 3. Time to treatment failure regarding doublet of chemotherapy.

Fig3.Time to treatment failure regarding doublet of chemotherapy.

Table 2. Medians and hazard ratios of time to treatment failure regarding age, sex, brain metastases, platinum compound and doublet of chemotherapy

  Median months, IC95%

 

P, log rank Hazard ratio, IC95% P, cox
Age

<70 años

>70 años

 

2,9 [2,07-3,9]

2,3 [2,1-2,5]

 

0,264

 

0,757 [0,46-1,24]

 

0,269

Sex

Male

Female

 

2,5 [2,2-2,8]

1,9 [0,0-5,04]

 

0,714

 

0,806 [0,25-2,6]

 

0,718

Brain Metastases (n, %)

No

Yes

 

2,5 [2,1-2,8]

2,3 [n.r.]

 

0,771

 

0,747 [0,1-5,5]

 

0,774

Platinum (n,%)

Cisplatin

Carboplatin

 

2,3 [2,01-2,7]

2,6 [1,9- 3,2]

 

0,895

 

1,037 [0,6-1,7]

 

0,896

Doublet of CT (n, %)

Microtubules inhibitor

Gemcitabine

 

 

1,7 [0,0-4,3]

2,5 [2,2- 2,8]

 

0,107

 

1,677 [0,8-3,1]

 

0,115

Overall survival

Median OS was 8,246 months CI95% [5,8 –2,6] in all patients (Figure 4). Overall survival rate al 6, 12 and 18 months were 62,8%, 35,1% and 3,8% respectively. Considering age, sex, brain metastases, platinum compound and doublet of chemotherapy there were statistically significant differences regard sex and doublet of CT (Table 3).

Figure 4. Overall survival in all patients

Fig 4. Overall survival in all patients

Table 3. Medians and hazard ratios of time to treatment failure regarding age, sex, brain metastases, platinium compound and doublet of chemotherapy

  Median months, IC95%

 

P, log rank Hazard ratio, IC95% P, cox
Age

<70 años

>70 años

 

7,5 [4,6-10,5]

8,9 [6,4-11,3]

 

0,619

 

0,884 [0,54-1,43]

 

0,619

Sex

Male

Female

 

8,3 [6,2-10,5]

3,2 [2,4-4,1]

 

0,006

 

0,217 [0,06-0,719]

 

0,012

Brain Metastases (n, %)

No

Yes

 

8,2 [5,8-10,6]

4,3 [n.r.]

 

0,596

 

1,466 [0,35-6,1]

 

0,599

Platinium (n,%)

Cisplatin

Carboplatin

 

10,48 [3,9-17,02]

7,65 [4,9- 10,33]

 

0,260

 

0,764 [0,42-1,2]

 

0,263

Doublet of CT (n, %)

Microtubules inhibitor

Gemcitabine

 

 

5,7 [3,8-7,6]

9,45 [6,7- 12,1]

 

0,018

 

1,989 [1,1-3,5]

 

0,021

In multivariate analyses adjusted for sex, age (< 70 or ≥ 70 years) and platinium the first line chemotherapy regimen based in antimetabolites was a predictor of OS (p=0.018).

Second line

Only 31 (37,8%) patients received second line therapy of treatment. Most common agents were docetaxel (17 patients, 54,8%), paclitaxel (6 patients, 19,9%), erlotinib (6 patients, 19,4%), gefitinib (1 patient, 3,2%) and gemcitabine (1 patient, 3,2%).

Median OS posprogression (OSpp) was 6,637 months CI95% [5,4 –7,8]. With respect to type of treatment, patients treated with docetaxel had a median of 9,01 months CI95% [5,1 –12,9], patients treated with paclitaxel 6,6 months CI95% [0,5 –12,7] and patients treated with erlotinib 5,8 months CI95% [4,6 –7,1]. Patient treated with gefitinib and gemcitabina were alive while 5,4 months and 3,2 months respectively. There were statistically significant differences in median OSpp regarding type of treatment (p=0,026 log rank). Hazard ratio for use of taxanes was 0,695, CI95% [0,26 –1,83], p=0,4.

Discussion

Squamous cell NSCLC is a particularly aggressive type of lung cancer, and few treatments are effective [6]. The NCCN and ESMO frontline recommendations include chemotherapy doublets, which are considered the cornerstone of initial therapy for squamous NSCLC [7,8]. To our knowledge, no prospective clinical trial has specifically compared cytotoxic chemotherapies for advanced lung SCC so there is is uncertainty about the best option of treatment.

Respect gender, women is represented in 3,7% in our study. In most published studies, little is reported about women diagnosed of advanced squamous NCSLC treated with CT. Overall survival among women in our study is poor (3,2 months versus 8,3). Recently, it has been described that females patients with squamous NCSLC had a significantly higher rate of human papillomavirus (HPV) infection compared to males with SCC [9] and HPV infection appears to be involved in cancer progression in SCC by promoting the expression of p53. On the other hand, patterns of mutation in SCC are unknown being KRAS, FGFR1 and PIK3CA most frequently reported and women seem to have less PIK3CA mutation than men. Moreover, there may exist unknown endocrine mechanisms this particularly lack of response and bad prognosis in women. To our knowledge, there are no differences in smoking patters that could explain this fact [10].

Elderly and young patients, defined by >70 years or <70 years, have similar proportion in our series. Although doublets with microtubule inhibitors are less used in old patients overall survival is similar in both groups. The definition of elderly patients varies widely across trials and and an uniform definition is lacking. Particularly in the actual world setting, older patients are often untreated even even when free of comorbid illnesses. Most studies in advanced NSCLC define elderly patients as those older than 70 years for the treatment of advanced NSCLC [11] and the benefit of platinum-based doublet regimens in this population seems to be greater than single-agent chemotherapy. So avoiding the use of doublets in elderly  is not justified in absence of comorbid status. Studies in elderly patients show similar response rates than in younger ones even with aggressive regimens of chemotherapy.

Palliative Care Unit (PCU) has played a preeminent role in the management of patients. More than half of our patients were remitted to PCU (45 patients, 54,9%) This fact allowed better control of symptoms, especially at home, and ultimately improved patients’ quality of life [12]. Early integration of palliative care into the treatment strategy should be mandatory because it may improve quality of life, particularly end-of-life care and improve overall survival. Our institution is working to make success this target.

Median time to treatment failure was 2,53 months and overall survival 8,24 months. The use of cisplatin or carboplatin has no impact on TTF or on OS, although use of cisplatin has a tendency to improve overall survival in 3 months over the use of carboplatin. Our data are similar to the reported about Veterans Health Administration data [12]. The use of gemcitabine as platinum doublet has demonstrated increased survival in our patients over microtubule inhibitors (9,45 months versus 4,7 months). A phase III trial of cisplatin and pemetrexed compared with cisplatin and gemcitabine revealed a statistically significant improvement in OS with cisplatin and gemcitabine in patients with squamous cell histology (10.8 vs 9.4 months) [13,14]. A retrospective study in patients with advanced lung SCC, revealed the use of various regimens did not have a significant effect on survival outcomes [15].

Finally, 31 patients (37,8%) received second line of treatment being docetaxel the most common agent used in this setting followed by paclitaxel and erlotinib. Although the number of patients is small, there is an improvement in survival in the group treated with taxanes respect to the group treated with tyrosine kinase inhibitor TKI). In TITAN phase III study, [16] patients were randomized to receive TKI or CT (taxanes or pemetrexed) and both treatments were similar in efficacy with different toxicity profiles.

We conclude that gemcitabine must be the CT of choice in advanced SCC. On the other hand, female patients had worse prognosis, patients aged >70 years old must receive a double of platinum, cisplatin is similar to carboplatin and cisplatin must be used when there is no contraindication for its use. Aditional agents and strategies must be developed in this setting to improve quality of life and survival.

References

  • Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, et al. (2013) Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49: 1374-1403. [crossref]
  • Le Chevalier T, Scagliotti G, Natale R, Danson S, Rosell R, et al. (2005) Efficacy of gemcitabine plus platinum chemotherapy compared with other platinum containing regimens in advanced non-small-cell lung cancer: a meta-analysis of survival outcomes. Lung Cancer 47: 69-80.
  • Reck M, Popat S, Reinmuth N, De Ruysscher D, Kerr KM, et al. (2014) Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25 Suppl 3: iii27-39. [crossref]
  • Schiller JH, Harrington D, Belani CP, et al. (2002) Comparison of four chemotherapy regimens for advanced non small-cell lung cancer. N Engl J Med 346: 92-8.
  • Edge SB, Compton CC (2010) The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17: 1471-1474. [crossref]
  • McKeage MJ, Jameson MB, et al. (2010) Comparative outcomes of squamous and non-squamous non-small cell lung cancer (NSCLC) patients in phase II studies of ASA404 (DMXAA): retrospective analysis of pooled data. J Thorac Dis 2:199-204.
  • Ettinger DS, Wood DE, Akerley W, Bazhenova LA, Borghaei H, et al. (2014) Non-small cell lung cancer, version 1.2015. J Natl Compr Canc Netw 12: 1738-1761. [crossref]
  • Reck M, Popat S, Reinmuth N, De Ruysscher D, Kerr KM, Peters S (2014) ESMO Guidelines Working Group. Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25 Suppl 3: iii27-39.
  • Fan X, Yu K, Wu J, Shao J, Zhu L, et al. (2015) Correlation between squamous cell carcinoma of the lung and human papillomavirus infection and the relationship to expression of p53 and p16. Tumour Biol 36: 3043-3049. [crossref]
  • Qiong Z, Na WY, Bo W2, Zhu Z, Ling P, et al. (2015) Alterations of a spectrum of driver genes in female Chinese patients with advanced or metastatic squamous cell carcinoma of the lung. Lung Cancer 87: 117-121. [crossref]
  • Owonikoko TK, Ramalingam SS2 (2015) Minimize toxicity or preserve efficacy? A delicate trade-off in the management of older patients with lung cancer. J Clin Oncol 33: 534-536. [crossref]
  • Wagner B, Meffert C2, Becker G2 (2015) [Availability and integration of palliative medicine at certified lung cancer centers]. Pneumologie 69: 218-224. [crossref]
  • Santana-Davila R, Szabo A, Arce-Lara C, Williams CD, et al. (2014) Cisplatin versus carboplatin-based regimens for the treatment of patients with metastatic lung cancer. An analysis of Veterans Health Administration data. J Thorac Oncol 9: 702-9.
  • Scagliotti GV, Parikh P, von Pawel J, et al. (2008) Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 26:3543-3551.
  • Liao BC, Shao YY, Chen HM, Shau WY, Lin ZZ, et al. (2015) Comparative effectiveness of first-line platinum-based chemotherapy regimens for advanced lung squamous cell carcinoma. Clin Lung Cancer 16: 137-143. [crossref]
  • Ciuleanu T, Stelmakh L, Cicenas S (2012) Efficacy and safety of erlotinib versus chemotherapy in second-line treatment of patients with advanced, non-small-cell lung cancer with poor prognosis (TITAN): a randomised multicentre, open-label, phase 3 study. Lancet Oncol 13:300-8.

Humanized Monoclonal Antibodies in Pulmonology: An Integrated Review

DOI: 10.31038/IMROJ.2017213

Abstract

Asthma is an important chronic disease affecting a lot of people worldwide. Treatment options for asthma like biological agents are being developed more frequently nowadays. Despite a lot of treatment options, some patients still remain symptomatic. As more and more practitioners choose treatment with biologic agents as a convenient way of therapy, biologic agents and other valuable methods must be discovered in order to cope with a growing number of treatment agents. This manuscript emphasizes on new generation monoclonal human (ized) antibodies in asthmatics and off-label use. The first developed biologic agent is the anti- immunoglobulin E monoclonal antibody called omalizumab. Currently it is an approved treatment option for asthma.

Introduction

Asthma is an important chronic disease affecting a lot of people worldwide [1]. Treatment options for asthma like biological agents are being developed more frequently nowadays. Despite a lot of treatment options, some patients still remain symptomatic. As more and more practitioners choose treatment with biologic agents as a convenient way of therapy, biologic agents and other valuable methods must be discovered in order to cope with a growing number of treatment agents. This special issue emphasizes on new generation monoclonal human (ized) antibodies in asthmatics [2-4].

The pathophysiological mechanisms underlying asthma, which is a heterogeneous disease, are characterized by interactive responses among various cell types and the hematopoietic cells of the adaptive and innate immune systems. Frequently is conventional therapy like inhaled steroids and beta-agonists sufficient for asthma symptoms. However, a little minority of the asthmatics is not controlled with conventional therapy. Therefore, are new treatment options essential for severe asthmatic patients [5].

Anti-IL-5 Molecules

Interleukins derived from T-helper-2 (Th2) cells and innate lymphoid cells play an important role in the pathogenesis of asthma. Monoclonal antibodies targeting these cytokines as treatment for severe asthma are expected to be beneficial [6]. Eosinophilic inflammation is an important event in the pathogenesis of asthma. IL-5 is a key cytokine that arranges eosinophil production, survival, maturation and recruitment of eosinophils to the inflammation [7]. Mepolizumab, reslizumab, and benralizumab are new developed monoclonal antibodies that target the cytokine IL-5. Mepolizumab and reslizumab have been approved by the US Food and Drug Administration (FDA) for the treatment of patients with severe asthma with an eosinophilic phenotype [8,9]. Mepolizumab and reslizumab binds directly to IL-5 ligand. These molecules effectively decreased circulating and sputum eosinophil counts, but they failed to improve airway mucosal eosinophilia, acute exacerbation rates, lung function and symptom scores in several studies. These disappointing results may be affected from inappropriate selection of the patients. In order to overcome the probable mechanistic limitations of early anti-IL-5 agents, an anti-IL5R monoclonal antibody was developed and called as benralizumab. Benralizumab, previously known as MEDI-563, is a humanized recombinant IgG1-k isotype monoclonal antibody. It was constructed from the mouse anti-human IL-5Rα mAbs generated by mice immunized with recombinant human IL- 5Rα [10]. Clinical studies revealed that anti-interleukin 5 monoclonal therapies for asthma could be safe for slightly improving FEV1 (or FEV1% of predicted value), quality of life, and reducing exacerbations risk and blood and sputum eosinophils. However these drugs have no significant effect on PEF, and SABA rescue use. These may be a result of patient selection. Further trials are required to clarify the optimal antibody for different patients [7].

Anti-IL-4/IL-13 Molecules

Another investigated cytokine important in the inflammatory pathways in the pathogenesis of asthma is anti-IL-4. IL-4 is a pleiotropic cytokine secreted mainly by activated T cells. Mast cells, basophils, and eosinophils can also secret IL-4 [10,11]. IL-4 is important in inducing IgE isotype switching, T cell polarization into Th2 cells, and generation of IL-4, IL-5, and IL-13 by Th2 cells. IL-4Rα is expressed on CD4+ and CD8+ T cells, B cells, macrophages, lung epithelial cells, airway goblet cells, and smooth muscle cells [11]. There is a functional homogeniety between IL-4 and IL-13. IL-4 can activate a heterodimeric receptor complex consisting of the IL-4 receptor α- subunit (IL-4Rα) and a γC subunit. IL-13 can activate the IL-4Rα and the IL-13 receptor α1-subunit (IL-13Rα1) [5]. Both IL-4 and IL-13 can bind to heterodimeric combination of the α-subunit of the IL-13 receptor and the α-subunit of the IL-4 receptor. And this leads to signaling of both IL-4 and IL-13. Therefore, will blocking IL-4R α with an antibody of this receptor chain expected to block the effects of both IL-4 and IL-13 [6].

Pascolizumab and VAK694 are anti-IL-4 neutralizing monoclonal antibodies. Also, IL-4 receptor antagonist drugs like dupilumab, pitrakinra and AMG-317 have been discovered. Even a recombinant IL-4Rα that captures soluble IL-4 and prevents their binding to IL-4 receptors, has been developed. It is called altrakincept. However, further research on this drug was discontinued by its manufacturer, since the phase 3 clinical trial failed to confirm its earlier promising results. Since there is a high redundancy of IL-4 and IL-13 signaling, blocking of both IL-4 and IL-13 has been expected to be more efficient [11]. Dupilumab is a drug that inhibits signaling from IL- 4 and IL-13 concomitantly. It is a molecule that binds to the alpha subunit of the IL-4 receptor. Phase II trials for dupilumab showed that asthma exacerbations were decreased in patients using this drug [12,13]. Dupilumab also improved lung function. It reduced the inhaled corticosteroid dose in the patient group. There was also an associated reduction in fractional exhaled nitric oxide with reduced serum concentrations of Th2-associated inflammatory markers such as CCL17 (TARC), CCL26 (eotaxin-3), and IgE [6]. These results are promising and further clinical trials will us show us the long-term efficacy of dupilumab [11].

Another similar drug that targets IL-4 is pitrakinra. It is a recombinant human IL-4 variant that competitively inhibits IL-4Ra to interfere with the actions of both IL-4 and IL-13. Studies on this competitive antagonist called pitrakinra revealed that it leads to significant reduction in asthma exacerbations and improves asthma symptoms in patients with eosinophilia [14]. Pitrakinra also attenuated the late-phase asthmatic response to allergen challenge in patients with mild atopic asthma [6]. The other drug called AMG317 was evaluated in another phase II trial in approximately 300 patients with moderate to severe asthma. Weekly injections over 12 weeks were well tolerated but did not have significant effects on the Asthma Control Questionnaire score (ACQ score; the primary outcome) [14].

Interleukin 13 shares 30% homology with interleukin 4. IL-13 is secreted by Th2 cells, ILC2s, mast cells, basophils, and eosinophils. IL-13 has the potential to increase goblet-cell differentiation, and activation of fibroblasts. IL-13 production can induce an increase in bronchial hyperresponsiveness, and switching of B-cell antibody production towards IgE [6]. IL-13 is similar to IL-4 and uses the same signaling pathways. The high-affinity receptor of IL-13 is a heterodimer of IL-4Rα/IL- 13Rα1. IL-13Rα1 is present on eosinophils, B cells, monocytes, macrophages, smooth muscle cells, lung epithelial cells, airway goblet cells, and endothelial cells. Biologicals that target IL-13 are anti-IL-13 mAbs: anrukinzumab, dectrekumab, GSK679586, IMA-026, lebrikizumab, RPC-4046, and tralokinumab [11].

Patients with severe asthma often have elevated levels in sputum despite therapy with high dose inhaled or oral corticosteroids. Lebrikizumab is one of the many humanized monoclonal antibodies that have been developed to specifically bind to IL-13 and inhibit its function [14]. In a randomized, double-blind, placebo- controlled study were 219 asthma patients observed and evaluated whether the drug lebrikizumab could alter the course of asthma. Lebrikizumab treatment was associated with improved lung function. Patients with high pretreatment levels of serum periostin had greater improvement in lung function with lebrikizumab. This therapy needs futher evaluation before being utilized in clinics [15]. Another monoclonal antibody called tralokinumab, an investigational human IL-13-neutralising immunoglobulin G4 monoclonal antibody, has been evaluated in adults with moderate to severe uncontrolled asthma despite controller therapies. Patients were randomly assigned to receive tralokinumab or placebo subcutaneously every 2 weeks for 13 weeks. Although it had an acceptable safety and tolerability, it did not reduce asthma exacerbations [16].

Antithymic stromal lymphopoietin

Thymic stromal lymphopoietin (TSLP) is an epithelial cell derived cytokine that may trigger allergic inflammation and, thus, play a role in allergic asthma [17]. It is an epithelial-derived cytokine and makes its effect through its receptor, TSLP-R, which is a heterodimeric receptor that consists of the IL-7 receptor alpha chain (IL-7Rα) and the TSLP receptor alpha chain 1 (TSLPRα). In hematopoietic cells, TSLP-R is mainly expressed in DCs, monocytes, B cells, T cells, NK cells, invariant natural killer T (iNKT) cells, eosinophils, basophils, and mast cells [11].

A human anti TSLP monoclonal immunoglobulin G2 lambda (AMG 157) that binds human TSLP and prevents receptor interaction was assessed in a trial. Randomly assigned 31 patients with mild allergic asthma received AMG 157 (700 mg) or placebo intravenously, once a month for three doses. The primary outcome, the maximum percentage decrease in the FEV during the late asthmatic response was 45.9 percent less in the AMG 157 group than the placebo group on day 84. AMG 157 reduced allergen induced bronchoconstriction and airway inflammation. No serious adverse effects were reported. Further studies on this drug are planned to clarify its use in clinical practice [17].

Anti-IL-9 Monoclonal Antibody

IL-9 is a Th2 cytokine and a T cell and mast cell growth factor. Anti-IL-9 antibody-treatment has been shown to protect from allergen-induced airway remodeling, with a concomitant reduction in mature mast cell numbers and activation. It can also decrease expression of the profibrotic mediators transforming growth factor (TGF)-b1, vascular endothelial growth factor (EGF), and fibroblast growth factor-2 (FGF-2) in the lung. The function of IL-9 in allergy has been investigated for its pleiotropic activities on cell types associated with allergic diseases including Th2 lymphocytes, mast cells, B cells, eosinophils, and airway epithelial cells. An anti-IL-9 monoclonal antibody (MEDI-528) has been studied in a clinical trial on 327 asthmatic subjects. Patients were randomized to receive placebo or one of three doses of MEDI-528 (dosage 30, 100, or 300 mg s.c. twice weekly for 4 weeks) in addition to their usual asthma medications. The addition of MEDI-528 to existing asthma controller medications did not improve ACQ-6 scores, asthma exacerbation rates, or FEV1 values. Further clinical trials are needed to explore this drug for altering the course of asthma. Thus, the potential clinical benefit of targeting IL-9 or its receptor in the treatment of asthma remains to be shown in further studies [5].

Anti-IL-2 antibody

Allergen exposure can stimulate IL-2 and its receptor expression (IL -2R) a chain (sCD25) in airways of patients with severe asthma. Daclizumab is a humanized monoclonal antibody that binds specifically to a subunit (CD25) of the high-affinity IL-2R, and inhibits IL-2 binding and its biological activity. Daclizumab can inhibit various T cell functions, including T cell proliferation and cytokine production. It has been investigated in a randomized controlled study. The drug has the potential to improve pulmonary function and asthma control in patients moderate to severe chronic asthma [18]. The risk of immunosuppression in clinical practice needs to be clarified.

Anti-GATA3-spesific DNAzyme

Approximately half of the asthmatic patients exhibit a Th2 type in response to allergen exposure. This Th2 endotype is characterized by a predominant activation of Th2 cells that produce cytokines such as interleukins 4, 5, and 13. The expression and production of all these Th2 cytokines have been shown to be controlled by the zinc finger transcription factor GATA3, which is essential for Th2-cell differentiation and activation. It is considered to be the master transcription factor of the Th2 pathway of immune activation. Therefore, could be interventions to disrupt this immune network, a synthetic DNA molecule (DNAzyme), that binds to GATA3 messenger RNA and cleaves it, a solution. This synthetic molecule called SB010 could significantly attenuate both late and early asthmatic responses after allergen provocation in patients with allergic asthma. Biomarker analysis after this drug showed an attenuation of Th2-regulated inflammatory responses [19].

Anti-IL-17 antibody

Although half of the asthma patients exhibit a Th2 type endotype, some remaining patients exhibit a Th17 driven endotype. This subpopulation is characterized with a Th17 driven inflammation. Th17 cells can contribute to airway hyperresponsiveness by recruiting both eosinophils and neutrophils. Therefore, has been IL-17 receptor blocking suggested to beneficial in asthma treatment [20]. Biologicals targeting IL-17 include an anti-IL-17A mAb: secukinumab and an anti-IL- 17 receptor mAb: brodalumab. Although the inhibition of IL-17 receptor A had no effect on subjects with asthma as a whole, a subgroup analysis showed an effect with uncertain significance. Further studies are needed to determine the role of secukinumab in asthma [11]. Brodalumab (AMG 827) is a human, anti–IL-17RA immunoglobulin G2 (IgG2) monoclonal antibody that binds with high affinity to human IL-17RA, blocking the biologic activity of IL- 17A, -17F, -17A/F heterodimer, and -17E (IL-25). Brodalumab can block IL-25 activity and IL-17A and IL-17F. In a randomized controlled study were 302 patients taking this drug evaluated and at the end of the study there was no evidence for an effect of brodalumab in these patients. Further studies may clarify the potential of this drug [20].

Anti TNF antibodies

In addition, human(ized) monoclonal antibodies (HMA) evaluated for the treatment of severe persistent asthma (SPA), but not approved after Phase II trial are as follows; Infliximab (Recombinant human–murine chimeric anti-TNFα monoclonal antibody Infliximab), etanercept (Soluble TNFα receptor fusion protein), and golimumab (Fully human TNFα-blocking antibody) [5]. The expression of TNF alfa is increased in asthma in association with airway neutrophilia. Berry and colleagues have demonstrated that the TNF-α axis is upregulated in patients with refractory asthma, as evidenced by the increased expression of membrane-bound TNF-α, TNF receptor 1, and TNF-α– converting enzyme by peripheral-blood monocytes [21]. Treatment with golimumab did not demonstrate a favorable risk-benefit profile in patients with severe persistent asthma [22]. A study with etanercept showed a small decrease in asthma exacerbations was observed in a randomized placebo controlled study [23]. In a case-series report was it told that in severe, uncontrolled, steroid-dependent asthma infliximab could reduce exacerbations and hospitalizations [24]. In some severe refractory asthma endotypes anti-TNFα therapy may have a role. However, it should be kept in mind that these agents have some safety concerns and should use carefully only in some severe refractory asthma endotypes [5].

ANTI-IgE

The first developed biologic agent is the anti-immunoglobulin E monoclonal antibody called omalizumab. Currently it is an approved treatment option for asthma [5].

The other human(ized) promising monoclonal antibody drug developed, but not approved yet is ligeluzimab. Ligeluzimab binds with very high affinity to the Cε3 domain of IgE. Ligeluzumab may provide longer supression of IgE. Trials with this biologic agent are ongoing [5].

Anti-ige: off-label use non-atopic asthma

The off-label use of omalizumab in patients with uncontrolled non-atopic asthma has resulted in a decrease in exacerbation rates and improvement in asthma symptom scores. In a study conducted in 2013, omalizumab was administered to 266 patients with severe allergic asthma and 29 patients with non-atopic severe asthma for two years, and the study found a decline in the exacerbation rate, increase in the quality of life, and significantly improved disease control in both groups [25-27]. In two studies which used omalizumab in a group of patients with non-atopic severe asthma, the authors observed downregulation of FcRI expression in the basophils and increased FEV1.

Nasal polyposis, allergic rhinitis, and allergic bronchopulmonary aspergillosis

Allergic bronchopulmonary aspergillosis affects 7 to 9% of patients with cystic fibrosis (CF) and 1 to 2% of patients with asthma, posing a diagnostic challenge [28,29]. Cases series related to ABPA were first published in 2007. In a series of eight cases with cystic fibrosis and ABPA published by Tanou et al. [30] in 2014, the authors reported increased FEV1, improved respiratory symptoms, and reduced steroid consumption. In a series of six cases with CF diagnosed with ABPA published by Lehman et al. [31], the authors reported improved symptoms in patients receiving omalizumab, whereas the efficacy of the treatment was less pronounced in patients diagnosed with ABPA and long disease duration and in patients who developed progressive lung problems [31]. In a series of 14 patients with severe asthma and ABPA in 2015, Aydın et al. [32] showed that 11 patients achieved complete and three patients achieved partial response. The authors also reported an overall improvement in the pulmonary functions and respiratory symptoms with a statistically significant reduction in the use of oral corticosteroids (OCS) and number of disease episodes. Also, the patients with a total immunoglobulin E (IgE) level of <1,000 IU showed a better response to omalizumab, compared to those with a total IgE level of >1,000 IU. However, this finding was found to have a low-evidence level, considering the lack of large-scale, prospective case series and randomized and placebo- controlled studies.

Efficacy and safety of omalizumab were first evaluated in a randomized, double-blind, placebo-controlled study of 221 patients with seasonal allergic rhinitis in 2002 [33]. This study reported a significant symptomatic relief up to 48% in the combination treatment group (specific immunotherapy [SIT]+omalizumab), compared to SIT group alone. A randomized study in Japan reported a significant improvement in daily nasal and eye symptoms in patients with seasonal allergic rhinitis receiving omalizumab [34].

In 2007, a randomized placebo-controlled study of eight patients was the first to report reduced rates of postoperative polyp recurrence in patients with atopic asthma and nasal polyps (NP) [35]. In a study of 19 patients with severe asthma and NP in 2011, Vennera et al. [36] reported symptom reduction and disease stabilization with the use of omalizumab treatment. In addition, Tajiri et al. [37] evaluated omalizumab in patients with severe asthma and NP, and reported significant improvements in nasal symptoms, asthma control, and sinus tomography results. However, not all studies were able to show the beneficial effects of the treatment. In a randomized, double-blind, placebo-controlled study of patients with chronic rhinosinusitis receiving omalizumab, Pinto et al. [38] showed improvement in the Sino-Nasal Outcome Test (SNOT-20) scores at three, five, and six months, although there was no significant difference in the scores compared to the control group. The aforementioned study did not observe any changes in the quality of life, symptom scores, cellular inflammation, nasal passage, and olfactory test parameters.

Atopic Dermatitis And Food Allergy

In a series of three patients published in 2005, the authors reported no response after four months of treatment [39], while Lane et al. [40] published a series of three patients in the same study period and reported successful treatment outcomes of severe AD using omalizumab [40]. In addition, a pilot study of 21 patients published in 2009 found a statistically significant clinical improvement in all patients [41]. Another series of three patients published in 2011 reported significant improvement in the Eczema Area and Severity Index (EASI) and itching severity score in patients with severe AD unresponsive to conventional treatment and those with elevated IgE levels [42]. A study of 11 patients published in the same year reported reduced SCORing Atopic Dermatitis (SCORAD) scores, reduced symptoms, and significant improvement in the quality of life (3). The efficacy of omalizumab was also evaluated in 20 adults with severe AD in a prospective, 28-week, open-label study conducted by Hotze et al. [43] in 2014. The authors reported no response to treatment in seven patients harboring filaggrin gene mutation (FLG), while there was a significant improvement in the remaining eight patients. The authors also concluded that the patients with FLG gene mutation were prone to achieve lower response to omalizumab.

Furthermore, in food-related immunotherapy (IT) studies, omalizumab initiated before or received simultaneously with the treatment facilitated the development of tolerance. In a series of 11 patients with cow milk allergy scheduled for IT, 10 patients tolerated daily intake of 8 g cow milk after the initiation of omalizumab, nine weeks before IT, and combination with IT treatment, thereafter [44]. Another use of this treatment is to facilitate a rapid and safe transition to the maintenance phase in patients with food allergy receiving oral IT [45]. Another use of the treatment in food allergy is eosinophillic esophagitis developing in association with multiple food allergies. Also, in a study administered omalizumab to patients with eosinophillic esophagitis, the authors found reduced allergic symptoms and improved quality of life, although there was no change in endoscopic and histologic characteristics of the disease [46].

Food Allergy and Anaphylaxis

Anaphylaxis can occur as a result of exposure to various allergens such as food, drug, and venom; however, no triggering factor can be shown in some cases, of which the latter is known as idiopathic anaphylaxis. The patients with elevated baseline tryptase levels or those diagnosed with mastocytosis are expected to have higher rates of anaphylaxis with a more aggressive course of disease. In particular, patients with venom allergy on IT may experience some difficulties in switching to maintenance dose. Severe anaphylactic episodes can be observed in patients diagnosed with mastocytosis on IT due to venom allergy. In addition, IT combined with omalizumab has enabled a safe transition to the maintenance phase in this group of patients [47-49]. Addition of omalizumab to rush and ultra-rush venom IT protocols has increased the success of IT and enabled a safer transition to the maintenance phase. Another use of omalizumab is to prevent recurrent anaphylactic episodes in patients who are unable to be controlled with conventional therapies [50- 55]. There are case reports on the role of this treatment in preventing idiopathic anaphylactic episodes [56-58].

Several studies have reported asthma symptom control, improved quality of life, and development of tolerance to aspirin in two patients with the use of omalizumab in patients with aspirin-induced airway disease, nasal polyps, and severe asthma [59, 60]. Two patients with recurrent insulin allergy, despite the use of desensitization protocols, and one patient of carboplatin allergy during carboplatin therapy due to ovarian cancer successfully continued their treatment with the addition of omalizumab to the treatment [61-63].

In conclusion, although omalizumab has been approved for the treatment of severe allergic asthma and chronic idiopathic urticaria, it offers an off-label use as a final resort in many allergic diseases. Recent studies have shown that omalizumab is effective in treating bullous pemphigoid, Stevens-Johnson syndrome (SJS)/toxic
epidermal necrolysis (TEN, Type-III/IV reaction, such as graft versus host disease), Netherton syndrome, asthma and chronic obstructive pulmonary disease overlap syndrome (ACOS). Its effects on soluble inflammatory markers, such as sCD200, sTRAIL, hematopoietic cells, Th1/2 cytokines (CXCL8; IL-1β; IL-4; IL-5; IL17A), total antioxidant capacity, hydrogen peroxide, malondialdehyde and total nitric oxide concentrations were demonstrated in several studies [64-70].

Monoclonal antibodies are a candidate for use in several indications with the contribution of large-scale studies to the literature in the near future.

Declaration of Interest

All authors declare that they have no conflict of interest.

Acknowledgements

Prof.Dr. Tse Wen Chang, Prof.Dr.Saadet Gumuslu, Prof.Dr. Fatih Uz, Prof.Dr Arzu Mirici.

References

  • Beasley R, Semprini A, Mitchell EA (2015) Risk factors for asthma: is prevention possible? Lancet 386: 1075-1085. [crossref]
  • Yalcin AD, Celik B, Gumuslu S (2014) D-dimer levels decreased in severe allergic asthma and chronic urticaria patients with the omalizumab treatment. Expert Opin Biol Ther 14:283-6.
  • Yalcin AD (2015) Advances in anti-IgE therapy. Biomed Res Int 2015: 317465. [crossref]
  • Yalcin AD, Cilli A, Bisgin A, et al. (2013) Omalizumab is effective in treating severe asthma in patients with severe cardiovascular complications and its effects on sCD200, d-dimer, CXCL8, 25-hydroxyvitamin D and IL-1ß levels. Expert Opin Biol Ther 13:1335-41.
  • Mitchell PD, El-Gammal AI, O’Byrne PM (2016) Emerging monoclonal antibodies as targeted innovative therapeutic approaches to asthma. Clin Pharmacol Ther 99:38-48.
  • Chung KF (2015) Targeting the interleukin pathway in the treatment of asthma. Lancet 386: 1086-1096. [crossref]
  • Wang FP, Liu T, Lan Z, Li SY, Mao H (2016) Efficacy and Safety of Anti-Interleukin-5 Therapy in Patients with Asthma: A Systematic Review and Meta-Analysis. PLoS One 11: e0166833. [crossref]
  • Ortega HG, Liu MC, Pavord ID, Brusselle GG, FitzGerald JM, et al. (2014) Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med 371: 1198-1207. [crossref]
  • Castro M, Zangrilli J, Wechsler ME et al. (2015) Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med 3:355-66.
  • Khorasanizadeh M, Eskian M, Assa’ad AH, Camargo CA Jr, Rezaei N,  et al. (2016) Efficacy and Safety of Benralizumab, a Monoclonal Antibody against IL-5Rα, in Uncontrolled Eosinophilic Asthma. Int Rev Immunol 35: 294-311. [crossref]
  • Tan HT, Sugita K, et al. (2016) Novel Biologicals for the Treatment of Allergic Diseases and Asthma. Curr Allergy Asthma Rep 16: 70. [crossref]
  • Wenzel S, Ford L, Pearlman D, Spector S, Sher L, et al. (2013) Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med 368: 2455-2466. [crossref]
  • Wenzel S, Castro M, Corren J, et al. (2016) Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high- dose inhaled corticosteroids plus a long-acting ß2 agonist: a randomised double-blind placebo-controlled pivotal phase 2b dose-ranging trial. Lancet 388:31-44.
  • Hambly N, Nair P (2014) Monoclonal antibodies for the treatment of refractory asthma. Curr Opin Pulm Med 20: 87-94. [crossref]
  • Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, et al. (2011) Lebrikizumab treatment in adults with asthma. N Engl J Med 365: 1088-1098. [crossref]
  • Piper E, Brightling C, Niven R, Oh C, Faggioni R, et al. (2013) A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Eur Respir J 41: 330-338. [crossref]
  • Gauvreau GM, O’Byrne PM, Boulet LP, et al. (2014) Effects of an anti-TSLP antibody on allergen-induced asthmatic responses. N Engl J Med 370:2102-10.
  • Busse WW, Israel E, Nelson HS, et al. (2008) Daclizumab improves asthma control in patients with moderate to severe persistent asthma: a randomized, controlled trial. Am J Respir Crit Care Med 178:1002-8.
  • Krug N, Hohlfeld JM, Kirsten AM, et al. (2015) Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme. N Engl J Med 372:1987-95.
  • Busse WW, Holgate S, Kerwin E, et al. (2013) Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med 188:1294-302.
  • Berry MA, Hargadon B, Shelley M, Parker D, Shaw DE, et al. (2006) Evidence of a role of tumor necrosis factor alpha in refractory asthma. N Engl J Med 354: 697-708. [crossref]
  • Wenzel SE, Barnes PJ, Bleecker ER, et al. (2009) A randomized, double-blind, placebo-controlled study of tumor necrosis factor-alpha blockade in severe persistent asthma. Am J Respir Crit Care Med 179:549-58.
  • Morjaria JB, Chauhan AJ, Babu KS, et al. (2008) The role of a soluble TNFalpha receptor fusion protein (etanercept) in corticosteroid refractory asthma: a double blind, randomised, placebo controlled trial. Thorax 63:584-91.
  • Taillé C, Poulet C, Marchand-Adam S, Borie R, Dombret MC, et al. (2013) Monoclonal Anti-TNF-α Antibodies for Severe Steroid-Dependent Asthma: A Case Series. Open Respir Med J 7: 21-25. [crossref]
  • Domingo C, Pomares X, Angril N, Rudi N, Amengual MJ, et al. (2013) Effectiveness of omalizumab in non-allergic severe asthma. J Biol Regul Homeost Agents 27: 45-53. [crossref]
  • Garcia G, Magnan A, Chiron R, et al. (2013) A proof-of-concept, randomized, controlled trial of omalizumab in patients with severe, difficult-to-control, nonatopic asthma. Chest 144:411-9.
  • de Llano LP, Vennera Mdel C, Álvarez FJ, Medina JF, Borderías L, et al. (2013) Effects of omalizumab in non-atopic asthma: results from a Spanish multicenter registry. J Asthma 50: 296-301. [crossref]
  • Laufer P, Fink JN, Bruns WT, Unger GF, Kalbfleisch JH, et al. (1984) Allergic bronchopulmonary aspergillosis in cystic fibrosis. J Allergy Clin Immunol 73: 44-48. [crossref]
  • Greenberger PA, Patterson R (1988) Allergic bronchopulmonary aspergillosis and the evaluation of the patient with asthma. J Allergy Clin Immunol 81: 646-650. [crossref]
  • Tanou K, Zintzaras E, Kaditis AG (2014) Omalizumab therapy for allergic bronchopulmonary aspergillosis in children with cystic fibrosis: a synthesis of published evidence. Pediatr Pulmonol 49:503-7
  • Lehmann S, Pfannenstiel C, Friedrichs F, Kröger K, Wagner N, et al. (2014) Omalizumab: a new treatment option for allergic bronchopulmonary aspergillosis in patients with cystic fibrosis. Ther Adv Respir Dis 8: 141-149. [crossref]
  • Aydin Ö, Sözener ZÇ, Soyyigit S et al. (2015) Omalizumab in the treatment of allergic bronchopulmonary aspergillosis: One center’s experience with 14 cases. Allergy Asthma Proc 36:493-500.
  • Kuehr J, Brauburger J, Zielen S, et al. (2002) Efficacy of combination treatment with anti-IgE plus specific immunotherapy in polysensitized children and adolescents with seasonal allergic rhinitis. J Allergy Clin Immunol 109:274-80.
  • Okubo K, Nagakura T (2008) Anti-IgE antibody therapy for Japanese cedar pollinosis: omalizumab update. Allergol Int 57: 205-209. [crossref]
  • Penn R, Mikula S (2007) The role of anti-IgE immunoglobulin therapy in nasal polyposis: a pilot study. Am J Rhinol 21: 428-432. [crossref]
  • Vennera Mdel C, Picado C, Mullol J, Alobid I, Bernal-Sprekelsen M (2011) Efficacy of omalizumab in the treatment of nasal polyps. Thorax 66: 824-825. [crossref]
  • Tajiri T, Matsumoto H, Hiraumi H, Ikeda H, Morita K, et al. (2013) Efficacy of omalizumab in eosinophilic chronic rhinosinusitis patients with asthma. Ann Allergy Asthma Immunol 110: 387-388. [crossref]
  • Pinto JM, Mehta N, DiTineo M, et al. (2010) A randomized, double-blind, placebo- controlled trial of anti-IgE for chronic rhinosinusitis. Rhinology 48:318-24.
  • Krathen RA, Hsu S (2005) Failure of omalizumab for treatment of severe adult atopic dermatitis. J Am Acad Dermatol 53: 338-340. [crossref]
  • Lane JE, Cheyney JM, Lane TN, Kent DE, Cohen DJ (2006) Treatment of recalcitrant atopic dermatitis with omalizumab. J Am Acad Dermatol 54: 68-72. [crossref]
  • Sheinkopf LE, Rafi AW, Do LT, Katz RM, Klaustermeyer WB (2008) Efficacy of omalizumab in the treatment of atopic dermatitis: a pilot study. Allergy Asthma Proc 29: 530-537. [crossref]
  • Thaiwat S, Sangasapaviliya A (2011) Omalizumab treatment in severe adult atopic dermatitis. Asian Pac J Allergy Immunol 29: 357-360. [crossref]
  • Hotze M, Baurecht H, Rodríguez E, et al. (2013) Increased efficacy of omalizumab in atopic dermatitis patients with wild-type filaggrin status and higher serum levels of phosphatidylcholines. Allergy 69:132-5.
  • Nadeau KC, Schneider LC, Hoyte L, Borras I, Umetsu DT (2011) Rapid oral desensitization in combination with omalizumab therapy in patients with cow’s milk allergy. J Allergy Clin Immunol 127: 1622-1624. [crossref]
  • Bégin P, Dominguez T, Wilson SP, Bacal L, Mehrotra A, et al. (2014) Phase 1 results of safety and tolerability in a rush oral immunotherapy protocol to multiple foods using Omalizumab. Allergy Asthma Clin Immunol 10: 7. [crossref]
  • Rocha R, Vitor AB, Trindade E, Lima R, Tavares M, et al. (2011) Omalizumab in the treatment of eosinophilic esophagitis and food allergy. Eur J Pediatr 170: 1471-1474. [crossref]
  • Kontou-Fili K. High omalizumab dose controls recurrent reactions to venom immunotherapy in indolent systemic mastocytosis. Allergy 63:376-8.
  • Sokol KC, Ghazi A, Kelly BC, Grant JA (2014) Omalizumab as a desensitizing agent and treatment in mastocytosis: a review of the literature and case report. J Allergy Clin Immunol Pract 2: 266-270. [crossref]
  • da Silva EN, Randall KL (2013) Omalizumab mitigates anaphylaxis during ultrarush honey bee venom immunotherapy in monoclonal mast cell activation syndrome. J Allergy Clin Immunol Pract 1:687- 8.
  • Douglass JA, Carroll K, Voskamp A, Bourke P, Wei A, et al. (2010) Omalizumab is effective in treating systemic mastocytosis in a nonatopic patient. Allergy 65: 926-927. [crossref]
  • Jagdis A, Vadas P (2014) Omalizumab effectively prevents recurrent refractory anaphylaxis in a patient with monoclonal mast cell activation syndrome. Ann Allergy Asthma Immunol 113:115-6.
  • Paraskevopoulos G, Sifnaios E, Christodoulopoulos K, Mantopoulou F, Papakonstantis M, et al. (2013) Successful treatment of mastocytic anaphylactic episodes with reduction of skin mast cells after anti-IgE therapy. Eur Ann Allergy Clin Immunol 45: 52-55. [crossref]
  • Matito A, Blázquez-Goñi C, Morgado JM, Alvarez-Twose I, Mollejo M, et al. (2013) Short-term omalizumab treatment in an adolescent with cutaneous mastocytosis. Ann Allergy Asthma Immunol 111: 425-426. [crossref]
  • Bell MC, Jackson DJ (2012) Prevention of anaphylaxis related to mast cell activation syndrome with omalizumab. Ann Allergy Asthma Immunol 108: 383-384. [crossref]
  • Kibsgaard L, Skjold T, Deleuran M, et al. (2014) Omalizumab induced remission of idiopathic anaphylaxis in a patient suffering from indolent systemic mastocytosis. Acta Derm Venereol 94:363-4.
  • Lee J (2014) Successful prevention of recurrent anaphylactic events with anti-immunoglobulin E therapy. Asia Pac Allergy 4: 126-128. [crossref]
  • Demirtürk M, Gelincik A, ColakoÄŸlu B, Dal M, Büyüköztürk S (2012) Promising option in the prevention of idiopathic anaphylaxis: omalizumab. J Dermatol 39: 552-554. [crossref]
  • Jones JD, Marney SR Jr, Fahrenholz JM (2008) Idiopathic anaphylaxis successfully treated with omalizumab. Ann Allergy Asthma Immunol 101:550-1.
  • Bobolea I, Barranco P, Fiandor A, et al. (2010) Omalizumab: a potential new therapeutic approach for aspirin-exacerbated respiratory disease. J Investig Allergol Clin Immunol 20:448-9.
  • Matheu V, Franco A, Perez E, Hernández M, Barrios Y (2007) Omalizumab for drug allergy. J Allergy Clin Immunol 120: 1471-1472. [crossref]
  • Cavelti-Weder C, Muggli B, Keller C, et al. Successful use of omalizumab in an inadequately controlled type 2 diabetic patient with severe insulin allergy. Diabetes Care 35:e41.
  • Yong PF, Malik R, Arif S, Peakman M, Amiel S, et al. (2009) Rituximab and omalizumab in severe, refractory insulin allergy. N Engl J Med 360: 1045-1047. [crossref]
  • Ojaimi S, Harnett PR, Fulcher DA (2013) Successful carboplatin desensitization by using omalizumab and paradoxical diminution of total IgE levels. J Allergy Clin Immunol Pract 2:105-6.
  • Uzun R, Yalcin AD, Celik B, et al. (2016) Levofloxacin Induced Toxic Epidermal Necrolysis: Successful Therapy with Omalizumab (Anti-IgE) and Pulse Prednisolone. Am J Case Rep 17:666-71.
  • Yalcin AD (2015) A case of netherton syndrome: successful treatment with omalizumab and pulse prednisolone and its effects on cytokines and immunoglobulin levels. Immunopharmacol Immunotoxicol 38:162- 6.
  • Yalcin AD, Celik B, Yalcin AN (2016) Omalizumab (anti-IgE) therapy in the asthma-COPD overlap syndrome (ACOS) and its effects on circulating cytokine levels. Immunopharmacol Immunotoxicol 38:253-6.
  • Yalcin AD, Genc GE, Celik B, Gumuslu S (2014) Anti-IgE monoclonal antibody (omalizumab) is effective in treating bullous pemphigoid and its effects on soluble CD200. Clin Lab 60: 523-524. [crossref]
  • Yalcin AD, Bisgin A, Gorczynski RM (2012) IL-8, IL10, TGF-ß and GCSF levels were increased in severe persistent allergic asthma patients with the anti-IgE treatment. Mediators inflamation
  • Yalcin AD, Bisgin A (2012) The Relation of sTRAIL Levels and Quality of Life In Omalizumab Using Severe Persistent Allergic Asthma Patients. (WISC 2012: WAO Outstanding Abstract Award) Med Sci Monit 18: LE9-10.
  • Yalcin AD, Ucar S, Gumuslu S, Strauss L (2015) Effects of Omalizumab on Eosinophil Cationic Peptid, 25-Hydroxyvitamin-D, IL-1ß, and sCD200 in a cases of Samter’s syndrome: 36 Months follow-up. Immunopharmacology And Immunotoxicology.

The Usefulness of the Rate Pressure Product (RPP) for Cardiac Rehabilitation Exercise Prescription

DOI: 10.31038/IMROJ.2017212

Introduction

The autonomic nervous system (ANS) is an arm of the nervous system surrounded by the peripheral nervous system (PN) and the central nervous system (CNS). It is responsible for the regulation of involuntary bodily functions such as the beating of the heart to the way in which food is digested [1-4]. This system further separates into two division: the sympathetic nerves (SNs) and the parasympathetic nerves (PNs) with each carrying efferent (motor) signals to the heart and afferent responses to the brain [2,3]. In maintaining the body’s homeostasis, each nerve fibre triggers internal or external stimulus. Stimulations coming from the SNs releases epinephrine and norepinephrine prepare the body for stressful or emergency situations or what is best known as the fight or flight state [2]. SNs activities increase heart rate, cardiac output, contractility, conduction velocity and blood pressure during physical stimuli such as exercise [2]. SNs also makes the palm of the hand sweat, the pupils to dilate, and causes the hair on the body to stand on end [2].

In contrast, the PNs which originate from the brain stem and the sacral portion of the spinal cord releases acetylcholine to conserve energy during normal relaxed situations [2]. The efferent outflow termed the vagus nerve operates the parasympathetic to transmit nerves fibres to the lung, heart and other organs [2, 3,]. These nerves work to lower blood pressure (BP), to slow the heart rate (HR) down and to control digestive functions [2].

The SNs and PNs work in opposite direction of each other and as such the SNs enhance automaticity, while the PNs inhabit it [5]. A good demonstration of SNs and PNs operating in opposed action would be if the heart receives a neural stimulus from the parasympathetic branch; it would slow the heart down whereas sympathetic activities would speed up the heart. There is a wide consent suggesting that any changes between the systems play a role in pathological dysfunctions of the ANS [3-5]. For example, a cascade of adverse cardiac events takes place if parasympathetic vagal tone decreases [3, 5-6]. Hypertension-related diseases, coronary heart disease, heart failure and myocardial ischemia are various heart conditions caused by chronic SNs activation [5-8]. The shifts to a more sympathetic overdrive are a catastrophe for ANS impairment. As such any treatment whether by drug action or with exercise training that tilts the autonomic balance toward greater parasympathetic dominance and less sympathetic activity significantly improve prognosis [3].

Moreover, cardiovascular autonomic functions or dysfunctions are clinically evaluated by measuring resting heart rate (RHR), heart rate (HR), BP or heart rate recovery (HRR) [6]. Whether done directly or indirectly these autonomic parametres are good indicators in determining how the heart is working during conditions like exercise or stress [6,7,5,9]. In clinical practices, more specifically in cardiac rehabilitation (CR), several autonomic parametres are used in assessing patients ANS function and their physical capacity. Unfortunately, because of the difficulties and the lack of experience to perform some tests, and the time it takes to do the test, some methods are not applicable in CR setting [4].

In looking at different nerve simulations, clinical research studies found that ANS parametres were risk markers for cardiovascular diseases [5,7,9]. A decline in heart rate variability (HRV), for example, was associated with many cardiac conditions including sudden death [9]. Indeed clinical procedures like HRV in monitoring autonomic processes are necessary with patients. They are practical to check if the ANS is operating normally or to see if a disease or disorders are attacking the system. In this paper, we briefly look at the RPP and how the usefulness of this autonomic test is to CR. The RPP has been quoted in the literature, but it is now accepted as a reliable tool for making clinical decisions for exercise prescription.

Cardiac Rehabilitation and Exercise Testing (ET)

Cardiac rehabilitation is an outpatient health programme delivered by a multidisciplinary team of health professionals (i.e. physicians, nurses, exercise physiologist, dieticians) following cardiac incidents [10-13]. The plan typically provides a multifaceted offering of health services such as low to moderate exercise training, health education, risk factor modifications, counselling and social services [10-12]. The objective of CR is to enhance secondary prevention by lessening cardiac symptoms thereby reducing cardiac mortalities and morbidities for patients with cardiovascular disease [13]. Evidence demonstrate the efficacy of CR interventions where these schemes have improved patient’s quality of life (i.e. reduce depression, better risk profile, enhanced functional status) [12-14]. In one study a CR exercise-based programme was safe to improve cardiopulmonary function with patients who had preserved left ventricular ejection fraction (LVEF) and reduced LVEF [15]. In another research, the authors suggested following coronary artery bypass surgery exercise has the potential to better the long-term prognosis and lower the need for hospital care in cardiac patients [16]. CR is indicative in supporting cardiac autonomic functions to improve the long-term health and well-being of cardiac patients and their families.

Exercise prescription in CR is a determinant on patient’s ET results. Before the start of a CR programme, it is standard practice for all patients to undergo clinical assessments which include ET [11]. With a goal to boost patient’s clinical outcome, ET is done by evaluating left ventricular function (LVF) using an echocardiography or with a maximal exercise test limited by symptoms [11,17-20]. Before the beginning and ending of the programme, ET is the most critical testing component in CR. It provides plenty of information about patients’ functional capacity, their hemodynamic adaptation to maximal and submaximal levels of exercise HR and BP, their residual myocardial ischemia, and their cardiac arrhythmias which can be either induced or worsens with activity [20]. CR exercise testing also let us knows the amount needed to calculate patients training heart rate (THR) for the aerobic exercise [20].

The cardiopulmonary graded test or CPX is the gold standard and approved method used for CR exercise testing. Testing is conducted by treadmill walking, ergometre cycling, stepping, or performing a 12 minute timed walking test [11, 18-20]. During the CPX, patient’s peak oxygen uptake (VO2peak), their anaerobic threshold, their VE/VCO2 and O2 are observed as well as other parametres such as their maximal workload, and their resting and exercised BP and HR [20]. VO2peak is the most frequently analysed CPX parametre as it determines patients’ functional capacity, and it is the strongest prognostic for cardiovascular disease [20]. VO2peak provides information on exercise intensity with a percentage of 50% to 70% the most acceptable [18, 20]. Under the supervision of a healthcare professional patient’s workload is monitored at various exercise stage [18, 20]. They are asked about the perception of exercise intensity using the well-known Borg Rating of Perceived Exertion Scale (RPE) [11, 20]. Furthermore, it is advisable the patient completes each stages of exercise [11]. However, with their discretion, the physician or cardiologist could terminate the test at a particular heart rate or at the request of the patient [11].

Following the completion of ET patients HR, BP and their total VO2peak are recorded and analysed [20]. As a component of functional capacity, the VO2peak decides exercise prescription and is cited as an independent predictor of all-cause mortality in patients with cardiac conditions [11, 20]. After they have been discharged from CR, studies show patient’s functional capacity gets better [20]. The VO2peak test appears to be a valuable clinical assessment in the planning of patients’ management. If the test is not available to measure patients’ fitness capacity, the one metabolic equivalent (MET) formula is applied [11, 20]. The one METs is a very simple procedure to express the energy cost of physical activities as multiples of resting metabolic rate [11]. It is a measurement of the exertion intensity of physical activity, and it is defined by the amount of oxygen consumed while sitting quietly at rest and is equal to 3.5 ml O2 per kg body weight x min (i.e. 3.5 ml O2/kg/min) [11, 20]. For example, a physical activity requiring an 8-MET resting metabolic rate represents a VO2 of 28 ml • kg-1 • min-1. In calculating the absolute oxygen requirement of the activity with 8 MET the individual’s body weight is multiplied by the VO2 (kg-1 • min-1) (i.e. VO2 (kg-1 • min-1 =28 kg-1 • min-1 x 70 kg = 1.960 ml • min-1). A noted feature of the metabolic equivalent is that men and women do not produce the same values (i.e. METs= 14.7 – 0.11 x age for males and respectively 14.7 – 0.13 x age for females.) [20]. This gender difference in computation accounts for women’s having lower level of work capacity [11].

As mentioned earlier, patients’ exertion level in CR is estimated from the RPE scale. The scale ranges from 6 to 20, but the American Association of Cardiovascular and Pulmonary Rehabilitation (AACPR) suggest a RPE of 11 to 15 as a safe zone for patients [11]. An important characteristic of the RPE is that it works linearly with HR and with exercise intensity [11,21]. As such you can estimate the HR value of various levels of work intensity by adding a zero to each point on the PRE [21]. For instance, RPE of 6 becomes 60 and represents HR at rest, and 20 becomes 200, which may represent patients maximal HR [21]. Subsequently, you can use the RPP to know patients’ maximal HR and training workload. Case in point, if a patient develops some discomfort in the chest (i.e. angina pectoris) at a given level of exercise intensity, for safety the CR health professional should advise the patient to workout at a lower intensity. In this way, it would help to keep his training HR below the threshold where he may experience physical symptoms. The RPP is complementary to the RPE while having the ability to support safe CR exercise prescription for cardiac patients who might experience mild chest pain while they are exercising.

Rate Pressure Product (RPP)

To determine the energy requirement and establish the amount of stress put on the heart during exercise, cardiac specialist or exercise physiologists use the RPP. It is an observation of myocardial oxygen consumption (MVO2) [22-26] representing the internal myocardial workload when the heart beats while the external myocardial work is a reflection of different stages of exercise. [27]. Expressed as the product of systolic blood pressure (SBP) and resting heart rate, you can calculate the RPP by multiplying the SBP by the RHR and dividing by 100 (i.e. RPP = SBP x HR/100) [21-27]. PNs and SNs mediate both HR and SBP with SBP only affected by SN [25]. What’s more, depending on the individual physical or health condition RPP score may vary. Fornitano and de Godoy suggested RPP above 30,000 mmHg bpm are good values to predict the absence of obstructive coronary artery disease in patients with positive ET [27].

Heart Rate and Blood Pressure on Exercise Training

The heart needs sufficient amount of oxygen to work properly, and if there is not enough supply, it will cause the heart to weaken (e.g. heart failure) [22]. In this case, the RRP is important in providing information on patients’ myocardial oxygen consumption [20]. Blood pressure and HR is a determinant of physical fitness since they both increase during exercise, but not at the same pace [20, 24, 25]. In subjects with BP between 110 to 120 systolic and 60-80 diastolic whereas resting heart rate (RHR) is 65 to 70 beats per minute (bpm) is considered normal [26]. Under these conditions, the heart does not need to work as hard because the oxygen demand is less [26]. Conversely, in patients with BP over 140/90 mm Hg and an RHR of 85 bpm or higher the heart works harder as it requires more oxygen [26].

Typically, an increase in HR during exercise is a sign that more blood and oxygen is travelling to the working muscles, while elevated BP indicates more blood gets pumped to the heart [24, 26]. As noted, increased BP and HR do not occur at the same time. Thus, a rise in HR triggers blood vessels to widen which in turn helps to keep BP under control [24]. This situation is why healthy people can recover much faster from exercise as compared to someone with a medical condition [20, 22]. The quick recovery is also a sign that there is more parasympathetic vagal tone and less sympathetic activity, which also accounts for the reduction in HR [22].

Extensive clinical and rehabilitation studies on the impact of RPP noted its efficacy as a reliable index to assess patients’ with cardiovascular conditions or related complications on myocardial oxygen consumption during their exercise. Coelho and colleague identified positive changes in MVO2 values following training in patients with ischemic heart disease [28]. Keyhani and co-authors investigated the effects of an eight-week CR aerobic exercise programme on BP, HR, and RPP in patients with congestive heart failure (CHF) found their cardiac functional capacity improved as well as their autonomic function [29]. Still, Adams et al. compared peak RPP values with various modes of aerobic exercise after CR training discovered treadmill walking to associate with a higher score while resistance training produced a much lower number [21].

Looking at the RPP and autonomic responses of Tai Chi practitioners and non-practitioners at rest and using two different stressors: hand gripping and standing Figueroa and colleagues saw improved autonomic function (i.e. parasympathetic tone) with the Tai Chi group [27]. The Tai Chi practitioners’ sympathetic outflow and RPP were also significantly lower at rest suggesting they were better efficient in myocardial oxygen use during resting and pathological stress [27]. The positive outcomes are a testament that the RPP is a reliable tool and an acceptable approach in observing patient’s cardiac autonomic exercise responses that favour greater parasympathetic tone.
The magnitude and the time BP and HR changes after the cessation of exercise are not without discrepancies [28]. When compared to pre-exercise during the first hour of recovery, Somers et al. [30] found lower BP levels, whilst Pescatello et al. [30] saw a significant fall that was up to 12 hours following exercise. Equally, post-exercise HR was reported to enhance, cause no change or decrease [30]. These observations give us a hint that different exercise intensity, duration, and mode significantly influence BP and HR responses following training. Forjaz et al. [31] study recognised that exercise training at a lower intensity, does not only generate a small increase in RPP during exercise, but it also decreases post-exercise rate at rest. By doing so, this reduces myocardial oxygen consumption and lower cardiovascular risks after exercise. In regards to exercise at moderate or high intensity, RPP appears to be greater during training but it decreases below baseline following the recovery period [31]. As cited RPP varies with exercise and there is evident it has clinical implications in providing exercise prescription with those experiencing medical conditions [30].

Conclusion

In addition to standardised CR exercise tests, the RPP supplements with other ET. This autonomic measurement is efficient as gives clues and evaluates patient’s physical or cardiac functional capacity, exercise tolerance and oxygen demand during CR exercise testing and training. Importantly, its utilisation offers support to CR health professionals in selecting the right exercise intensity or training method for those patients whom may show cardiac risk.

References

  • Shields RW Jr (1993) Functional anatomy of the autonomic nervous system. J Clin Neurophysiol 10: 2-13. [crossref]
  • Freeman JV, Dewey FE, Hadley DM, Meyers J, Froelicher VF (2006) Autonomic nervous system interaction with the cardiovascular system during exercise. Pro ardiovasc Dis 48: 342-362.
  • Curtis BM, O’Keefe JH Jr (2002) Autonomic tone as a cardiovascular risk factor: the dangers of chronic fight or flight. Mayo Clin Proc 77: 45-54. [crossref]
  • Zygmunt A, Stanczyk J (2010) Methods of evaluation of autonomic nervous system function. Arch Med Sci 6: 11-18. [crossref]
  • Sztajzel J (2004) Heart rate variability: a noninvasive electrocardiographic method to measure the autonomic nervous system. Swiss Med Wkly 134: 514-522.
  • Martin B, Papelier Y, Laursen PB, Ahmaidi S (2007) Noninvasive assessment of cardiac parasympathetic function: postexercise heart rate recovery or heart rate variability? Am Jf Physiol Heart Circ Physiol 293: H8-H10.
  • Ruediger H, Seibt R, Scheuch K, Krause M, Alam S (2004) Sympathetic and parasympathetic activation in heart rate variability in male hypertensive patients under mental stress. J Hum Hypertens 18: 307-315.
  • Palatini P, Julius S (2009) The role of cardiac autonomic function in hypertension and cardiovascular disease. Curr Hypertens Rep 11: 199-205. [crossref]
  • Del Pozo JM, Gevirtz RN, Scher B, Guarneri E (2004) Biofeedback treatment increases heart rate variability in patients with known coronary artery disease. Am Heart J 147: 545.
  • National Health and Medical Research Council: Strengthening cardiac rehabilitation and secondary prevention for Aboriginal and Torres Strait Islander Peoples: a guide for health professionals. Canberra, Australia: Australian Government; 2005.
  • Goble AJ, Worcester M (1999) Best practice guidelines for cardiac rehabilitation and secondary prevention. Victoria, Australia: Heart Research Centre – on behalf of Department of Human Services Victoria
  • Cardiac rehabilitation: a model of care for South Australia. Australia: South Australia Department of Health, Statewide Service Strategy division.
  • Cardiac rehabilitation: evidence-based best practice guidelines. New Zealand: Heart Foundation; 2002.
  • Williams MA, Ades PA, Hamm LF, Keteyian SJ, LaFontaine TP, et al. (2006) Clinical evidence for a health benefit from cardiac rehabilitation: an update. Am Heart J 152: 835-841. [crossref]
  • Chul K, Choi HE, Lim YJ (2016) The effect of cardiac rehabilitation exercise training on cardiopulmonary function in ischemic cardiomyopathy with reduced left ventricular ejection fraction. Ann Rehabil Med 40: 647-656.
  • Hedbäck B, Perk J, Hörnblad M, Ohlsson U (2001) Cardiac rehabilitation after coronary artery bypass surgery: 10-year results on mortality, morbidity and readmissions to hospital. J Cardiovasc Risk 8: 153-158.
  • Zoneraich S (1983) Exercise testing and cardiac rehabilitation in patients with coronary artery disease. Bull N Y Acad Med 59: 635-659. [crossref]
  • Pina IL, Balady GJ, Hanson P, Labovitz AJ, Madonna DW, Myers J (1995) Guidelines for Clinical Exercise Testing Laboratories A Statement for Healthcare Professionals From the Committee on Exercise and Cardiac Rehabilitation, American Heart Association. Circulation 91:912-21.
  • Fletcher GF, Balady GJ, Amsterdam EA, Chaitman B, Eckel R, et al. (2001) Exercise standards for testing and training a statement for healthcare professionals from the American Heart Association. Circulation 104: 1694-1740.
  • Niebauer J (2011) Cardiac rehabilitation manual.
  • Adams J, Hubbard M, McCullough-Shock T, Simms K, et al. (2010) Myocardial work during endurance training and resistance training: a daily comparison, from workout session 1 through completion of cardiac rehabilitation. In Baylor University Medical Center.
  • Rishu S, Gupta V, Walia L, Mittal N (2013) Rate pressure product predicts cardiovascular risk in type 2 diabetics with cardiac autonomic neuropathy. Nat J Physiol, Pharma and Pharmacology 3: 43-47
  • Miyai N, Arita M, Miyashita K, Morioka I, Shiraishi T, Nishio I (2002) Blood pressure response to heart rate during exercise test and risk of future hypertension. Hypertension 39:761-6.
  • Gobel FL, Norstrom LA, Nelson RR, Jorgensen CR, Wang Y (1978) The rate-pressure product as an index of myocardial oxygen consumption during exercise in patients with angina pectoris. Circulation 3: 549-556.
  • Figueroa MA, Demeersman RE, Manning J (2012) The autonomic and rate pressure product responses of tai chi practitioners. N Am J Med Sci 4: 270-275. [crossref]
  • Sembulingam P, Sembulingam K, Ilango S, Sridevi G (2015) Rate pressure product as a determinant of physical fitness in normal young adults. IOSR-JDMS 1: 8-12.
  • Fornitano LD, Godoy MF (2006) Increased rate-pressure product as predictor for the absence of significant obstructive coronary artery disease in patients with positive exercise test. Arquivos brasileiros de cardiologia 86:138-44. 28
  • Coelho EM, Monteiro F, Da Conceição JM, Cruz J, Cunha D, et al. (1982) The rate pressure product: fact of fallacy? Angiology 33: 685-689. [crossref]
  • Keyhani D, Kargarfard M, Sarrafzadegan N, Sadeghi M (2013) Autonomic function change following a supervised exercise program in patients with congestive heart failure. ARYA atherosclerosis 9:150.
  • Al-Shamma YMH, Mhous AJ, Mousawi AM (2008) Heart Rate and Blood Pressure Changes After Exercise In Normal Adolescent In Relation To Body Mass Index. Kufa J Med 11: 437-447
  • Forjaz CL, Matsudaira Y, Rodrigues FB, Nunes N, Negrão CE (1998) Post-exercise changes in blood pressure, heart rate and rate pressure product at different exercise intensities in normotensive humans. Braz J Med Biol Res 31: 1247-1255. [crossref]

Revisiting ‘what causes cancer?’

DOI: 10.31038/CST.2017211

Letter to the Editor

A thoughtful editorial previously published in Cancer Studies and Therapeutics pondered the question of “What is the Main Cause of Cancer?’ [1]. Certainly there are no simple answers.

Perhaps a study by Poutahidis et al (2015) [2] provides some clues to this ‘What Causes Cancer?’ enigma. Their studies in animal models revealed multigenerational cancer phenomena that were transplantable using fecal microbiota alone. These findings raise the possibility that disrupted microbiota, arising from societal practices such as refined diets or antibiotics during earlier generations, may have carcinogenic consequences in subsequent generations. The authors postulated that detrimental microbial effects in utero and during infancy lead to a dysregulated host immune system featuring premature thymic involution, possibly via epigenetic mechanisms. Under these immune-suppressed conditions, future infant mucosal surfaces become more permeable to environmental threats including sepsis [3]. Extrapolating across generations, microbiota may function as part of a quorum sensing mechanism ultimately influencing host immune and hormonal homeostasis, thus altering cancer susceptibility of progeny animals [4]. In those studies, grandchildren of mice consuming ‘fast food’ diets were at high risk to develop cancer at a young age, even without other predisposing genetic or environmental risks.

These intriguing data are supported by other findings suggesting that bacteria should be on our ‘What Causes Cancer?’ radar screen [5-7]. Firstly, direct evidence exists in humans with Helicobacter pylori infection and inflammation-associated gastric cancer [8]. Likewise, in the lower bowel, infection with a related microbe H. hepaticus leads to inflammation-associated colon and mammary tumors in mice [9, 10]. Further, certain pathogenic Escherichia coli organisms are shown to cause DNA damage in gut epithelia [6], and even to invade the bloodstream and extra-intestinal tissues. Indeed, E. coli has been implicated in mastitis and breast cancer in women [11], whereas Lactobacillus sp apparently inhibits mammary cancer development [12]. This raises the possibility that certain microbiota serve as invisible mutagens or guardians that help to explain the enigma.

And there’s more. Many studies have now shown that cancer-fighting capacity of our immune system can be mobilized or inhibited by our gut bacteria [10, 13-15]. Animal model systems mimicking complex cancer processes in human subjects reveal that microbes indirectly modulate tissue injury repair capacity and risk for tumor development and progression [3, 7, 9, 13-17]. For example, Poutahidis et al (2013) found that microbe therapy in mice led to proficient wound repair occurring twice-as-fast as in untreated controls [16]. Another study by Varian et al (2016) showed that microbe monotherapy was sufficient to increase thymus gland size, inhibit intestinal polyp formation, and increase lifespan in mouse models [18]. The proposed immune mechanisms involved microbial up-regulation of transcription factor Forkhead box protein N1 (FoxN1), the protein that is entirely lacking in athymic nude mice rendering them without T lymphocytes and as a result highly permissive to cancer growth [18, 19].

This leads us back to the original question of ‘What causes Cancer?’. The original author posited that for a theory of to be widely accepted, the answer should explain the striking differences in cancer risk by age and among tissues [1]. We should at least consider the possibility that our modernized lifestyle practices using antibiotics, Caesarian births, and refined diets have depleted valuable diversity and beneficial organisms in our microbiome with carcinogenic consequences to future generations. After all, oral supplementation with a model organism Lactobacillus reuteri, once believed to be widespread in humans but now dwindling to <4% of people worldwide [4], was sufficient to rescue multigenerational health impairments in infant mice [2, 20].

Further research is needed to better understand the roles of microbiota among the many possibilities for “What is the Main Cause of Cancer?’. However, based on existing data, opportunities abound for engineering diets and microbe cocktails to reinforce host balance and extinguish cancer for generations to come.

References

  • López-Lázaro M (2016) What is the Main Cause of Cancer? Can Stu The J 1:1-2.
  • Poutahidis T, Varian BJ, Levkovich T, Lakritz JR, Mirabal S, et al. (2015) Dietary microbes modulate transgenerational cancer risk. Cancer Res 75: 1197-1204. [crossref]
  • Erdman SE, Poutahidis T (2015) Gut bacteria and cancer. Biochim Biophys Acta 1856: 86-90. [crossref]
  • Erdman SE, Poutahidis T (2016) Microbes and Oxytocin: Benefits for Host Physiology and Behavior. Int Rev Neurobiol 131: 91-126. [crossref]
  • Rao VP, Poutahidis T, Fox JG and Erdman SE (2007) Breast cancer: should gastrointestinal bacteria be on our radar screen? Cancer Res 67:847-850.
  • Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, et al. (2012) Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338:120-123.
  • Erdman SE (2016) Gut microbiota: Microbes offer engineering strategies to combat cancer. Nat Rev Gastroenterol Hepatol 13:125-126.
  • Wroblewski LE, Peek RM, Jr. and Wilson KT (2010) Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clinical microbiology reviews 23:713-739.
  • Erdman SE, Poutahidis T, Tomczak M, Rogers AB, Cormier K, et al. (2003) CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol 162: 691-702. [crossref]
  • Rao VP, Poutahidis T, Ge Z, Nambiar PR, et al. (2006) Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res 66:7395-7400.
  • Urbaniak C, Gloor GB, Brackstone M, Scott L, Tangney M, et al. (2016) The Microbiota of Breast Tissue and Its Association with Breast Cancer. Appl Environ Microbiol 82: 5039-5048. [crossref]
  • Lakritz JR, Poutahidis T, Levkovich T, Varian BJ, et al. (2014) Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice. Int J Cancer 135:529-540.
  • Erdman SE, Rao VP, Olipitz W, Taylor CL, Jackson EA, et al. (2010) Unifying roles for regulatory T cells and inflammation in cancer. Int J Cancer 126: 1651-1665. [crossref]
  • Vétizou M, Pitt JM,  Daillère R,  Lepage P2, Waldschmitt N3, et al. (2015) Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350: 1079-1084. [crossref]
  • Sivan A, Corrales L, Hubert N, Williams JB, et al. (2015) Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350:1084-1089.
  • Poutahidis T, Kearney SM, Levkovich T, Qi P, Varian BJ, et al. (2013) Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. PLoS One 8: e78898. [crossref]
  • Dvorak HF (2015) Tumors: wounds that do not heal-redux. Cancer Immunol Res 3: 1-11. [crossref]
  • Varian BJ, Goureshetti S, Poutahidis T, Lakritz JR,  et al. (2016) Beneficial bacteria inhibit cachexia. Oncotarget 7: 11803-11816. [crossref]
  • Erdman SE (2016) Microbes and healthful longevity. Aging (Albany NY) 8: 839-840. [crossref]
  • Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, et al. (2016) Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 165:1762-1775.

Basic Food Safety Practise of Open-Air-Market Vending In The Eastern Highlands Of Papua New Guinea

DOI: 10.31038/IMROJ.2017211

Abstract

A cross-sectional survey was conducted to appraise basic food safety practises by street or open-air market vendors about the township of Goroka in late 2014. Study subjects were randomly selected over a period of two months. Of the one hundred and twenty food vendors observed in the survey, most had unconventional methods of handling food or allowed consumers to pick up food with bare hands if they choose. Use of gloves was hardly observed. Only thirty-two percent handled cooked food with thongs or fork which was apparent at barbeque sites. Twenty-one percent of the subjects in the survey did not provide proper food wrappers. Water use was limited to fresh fruit sale but obviously lacking in all others. Practically all cooked-food vendors did not use covers on food containers consequently increasing the risk of environmental or fly contamination. This survey clearly demonstrates that street food vendors either possess limited knowledge of food safety and hygiene, or just improvise to earn a living. The activity is widespread and unregulated. However, such practise can easily set the stage for an outbreak of food-borne infection. Vulnerable consumers could be at particular risk and the practise could also help maintain and spread existing infections.

Key Words

Street food safety, sanitation, street food, vending and Papua New Guinea

Background

Easy access to clean water and proper sanitation relates to better health as much they are essential preconditions for promoting basic food safety [1-4]. Maintenance of hand hygiene through washing and use of gloves can lessen the level of contamination thereby ensuring food safety [4-7]. Contamination can occur during handling, processing or storage but can be minimised if appropriate safety measures are observed and those involved have a clear knowledge of basic food safety practises [8-10]. Some studies have found that the level of food contamination is associated with hygiene awareness and the attitudes of people involved in the business [11-13]. Contamination risk is nevertheless heightened if there is a notable gap in basic food safety knowledge and practise. [1, 3]. In Papua New Guinea, most diarrhoeal infections originate from contaminated food or water subsequent to poor sanitation or inaccessibility to clean water [14]. Cooked food sale in the open environment in the highlands is common. It is an informal sector practised by locals including the unemployed. Food such as cooked vegetables, meat, rice or floor is often prepared at homes and brought to the vending site. The food is usually packed in large bowls, or cooking pots and transported to vending site as opposed to static food warmers used in restaurants or other established outlets where a certain level of food safety is assured. Ready-to-eat food sale in open- air-market or street could be possibly compromised if there is a lax in food safety during preparation or storage for the duration of vending [15, 16]. On the other hand, vendors may not be obliged to adhere to basic food safety measures as the activity is not regulated [17, 18]. This survey aims to identify key gaps in basic food safety practise with respect to handling, protection from environmental contamination and easy access to clean water.

Materials and Methods

A cross-sectional survey was conducted in late 2014. It involved observing and recording the sale of ready-to-eat food, particularly cooked food by open-air-market or street vendor about the township of Goroka in the eastern highlands of Papua New Guinea. Sample size determination was not necessary as target subjects were limited and variable at the different sites visited. Data was obtained by observing key questions in a brief questionnaire without having to making verbal contact or interviewing subjects involved in the cooked-food sale. The anonymous approach by this this survey guaranteed confidential use of data. Questions were rather basic with the assumption that most of the vendors would possess basic concepts of hygiene and food safety despite having no formal training on food safety. Different sites were visited including the main market in town where such activities are competitive and frequent. Each questionnaire was completed through up-close examination of food containers, covers used on containers, serving utensils such as thongs, food wrapping and use of gloves. Observations were done on different occasions but it is possible some subjects may have appeared more than once in the survey as cooked food sale is common in the area. Necessary details of the vending events were captured in the questionnaire and analysed using Ms Excel.

Figure 1. Observed food safety measures

Figure 1. Observed food safety measures

Results

Following the survey, majority of the street and open-air-market vendors failed to maintain basic food safety. Categories of food observed in the survey were basically local, cooked food (Table 1). The hundred and twenty (n=120) vendors observed apparently demonstrated a low level of hygiene or food safety practise. Glove use (0%) was not observed through the entire duration of the survey although most vendors applied irregular measures of food handling (32%). It is probable that such food were prepared in similar manner and transported. Food wrapping (21%) appeared to be improvised in some instances where old newspapers were used. On the other hand, local consumers had a tendency to pick up food with bare hand or simply choose to eat despite the obvious. Hygiene about the vending sites was questionable as running water or basic sanitation infrastructure was non-existent with periodic waste management. It is a widely known concept that hand hygiene is maintained through washing particularly during food preparation. However, water use was only observed in peeled, fresh-fruit sale (31%). Virtually all food containers or boxes had covers stripped off (16%) purposely to allow visibility of food to prospective consumers. This practise exposed food to environmental or fly contamination. In contrast to barbeques, most other cooked food sale did not maintain heat so evidently food went cold after a while. The practise appeared to be regular throughout the duration of the survey with similar method of food preparation. All measures undertaken by vendors to ensure food safety were documented (Figure 1).

Table 1. Types of ready-to-eat food sold at streets or open-air-markets

Types of ready-to-eat food
Barbeques (sausages, pork, lamb etc,.)
Deep fried floor balls
Fried potatoes
Peeled fresh fruits
Plain bread balls/sandwiches
Boiled vegetables
Fried eggs

Discussion

Sale and consumption of ready-to-eat food in streets or open-air-markets in the highlands of Papua New Guinea is common amongst locals simply because they are within a consumer’s means and convenient than restaurants or established fast-food outlets [16]. However, open-markets are makeshift and mobile, therefore lack the necessary structure to protect and maintain food safety [17]. Poor regulation of the practise and overall insanitary conditions of the environment and vending sites raises serious issues of both health and food safety [18]. The apparent lack of water and sanitation infrastructure at designated markets should be also be a concern for overall public health safety [13, 18]. It was evident that individuals involved in this trade were semi-educated and possessed limited knowledge of basic food safety or would have had partial exposure to such information including personal hygiene [15]. The activity cannot be banned as it is an income-generating for many locals and unemployed town residents. The consistent demand for these easily accessible and cheap foods will certainly encourage and sustain the trade. This survey emphasizes the need to develop street-food safety by initiating awareness and education on basic food safety and raises the need to formulate appropriate codes of practise in accordance with the Hazard Analysis Critical Control Point (HACCP) guidelines [19, 20]. Moreover, public health authorities could consider developing policies aimed at training and registering or licensing street-food vending.

Conclusion

This survey clearly demonstrates that there is a poor level of food safety observance by vendors and consumer alike. In a region where diarrhoeal diseases are endemic and sanitation is deplorable, such vending practise has the potential to trigger enteric disease outbreaks [14, 21]. However, these vending practises are likely to continue unhindered unless relevant authorities step up. Contamination of food is likely and can easily set the stage for an outbreak of food-borne infection or associated diarrhoeal illness particularly amongst the vulnerable populations [13]. In addition, the practise could also help maintain and spread existing infections. Further study is required to determine the extent of microbial contamination on food samples and hands of food handlers in the open-air-market vending business. A major limitation to this study is the obvious lack of a structured interview to establish individual knowledge of food safety and use of raw materials for food preparation.

References

  • Fewtrell L, et al. (2005) Water, sanitation, and hygiene interventions to reduce diarrhoea in less developed countries: a systematic review and meta-analysis. The Lancet Infectious Diseases 5:42-52.
  • Annette Prüss, D.K., Lorna Fewtrell, and Jamie Bartram, Estimating the Burden of Disease from Water, Sanitation, and Hygiene at a Global Level. Environmental Health Perspectives 110: 6.
  • Bartram J, Cairncross S (2010) Hygiene, sanitation, and water: forgotten foundations of health. PLoS Med 7: e1000367. [crossref]
  • Curtis V, Schmidt W, Luby S, Florez R, Touré O, et al. (2011) Hygiene: new hopes, new horizons. Lancet Infect Dis 11: 312-321. [crossref]
  • Ayçiçek H, et al. (2004) Assessment of the bacterial contamination on hands of hospital food handlers. Food Control 15: 4253-259.
  • Malhotra R, et al. (2006) Study of hand hygiene and enteroparasite infestation among food handlers working in a medical college of North India. Indian Journal of Pathology & Microbiology 49: 296-301.
  • Green LRR, Vincent Mason, Ryan Bushnell, Lisa Reimann, David W, Mack James C (2007) Motsinger, Michelle D.; Stigger, Tammi; Selman, Carol A., Factors Related to Food Worker Hand Hygiene Practices. Journal of Food Protection
  • Lues JFR and I Van Tonder (2007) The occurrence of indicator bacteria on hands and aprons of food handlers in the delicatessen sections of a retail group. Food Control 18:326-332.
  • Brown KL and S Wray (2014) Control of airborne contamination in food processing, in Hygiene in Food Processing (Second edition), HLM Lelieveld, Holah JT, and Napper D, Editors, Woodhead Publishing. 174-202.
  • Skovgaard N (2007) New trends in emerging pathogens. Int J Food Microbiol 120: 217-224. [crossref]
  • Angelillo IF, Viggiani NM, Rizzo L, Bianco A (2000) Food handlers and foodborne diseases: knowledge, attitudes, and reported behavior in Italy. J Food Prot 63: 381-385. [crossref]
  • Bas M, Azmi Safak Ersun and Kivanç G (2006) The evaluation of food hygiene knowledge, attitudes, and practices of food handlers’ in food businesses in Turkey. Food Control 17:
  • Isara AR, Isah EC, Lofor PV, Ojide CK (2010) Food contamination in fast food restaurants in Benin City, Edo State, Nigeria: Implications for food hygiene and safety. Public Health 124: 467-471. [crossref]
  • Horwood P, Greenhill A (2012) Cholera in Papua New Guinea and the importance of safe water sources and sanitation. Western Pac Surveill Response J 3: 3-5. [crossref]
  • FAO (2014) Food for the Cities: Food processing and street foods.
  • FAO (2014) Food for the Cities: Street Foods, 2014.
  • Chander Pal Thakur, Chahat Narula RM, Swati Mahapatra and Kalita TJ (2013) Food safety and hygiene practices among street food vendors in Delhi, India. International Journal of Current Research
  • Regulators F.W.G.F.o.F.S (2002) The experience of improving the safety of street food via international technical assistance.
  • Ekanem EO (1998) The street food trade in Africa: safety and socio-environmental issues. Food Control 9: 211-215.
  • Unnevehr LJ and Jensen H (1999) The economic implications of using HACCP as a food safety regulatory standard. Food Policy 24: 625-635.
  • Bukenya, Nwokolo GB, Compound Hygiene N (1991) Presence of Standpipe and the Risk of Childhood Diarrhoea in an Urban Settlement of Papua New Guinea. International Journal of Epidemiology 20: 534-539.