Pharmacokinetics, biodistribution and antitumour effects of Sclerotium rolfsii lectin in mice

  • Authors:
    • Anupama S
    • Preeti Laha
    • Mamta Sharma
    • Kamal Pathak
    • Sanjay Bane
    • Arvind D. Ingle
    • Vikram Gota
    • Rajiv D. Kalraiya
    • Lu-Gang Yu
    • Jonathan M. Rhodes
    • Bale M. Swamy
    • Shashikala R. Inamdar
  • View Affiliations

  • Published online on: April 3, 2017     https://doi.org/10.3892/or.2017.5545
  • Pages: 2803-2810
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sclerotium rolfsii lectin (SRL) is a lectin isolated from the fungus Sclerotium rolfsii and has exquisite binding specificity towards the oncofetal Thomsen-Friedenreich antigen (TF-Ag; Galβ1-3GalNAcα-O-Ser/Thr) and its derivatives. Previous studies have shown that SRL inhibits the proliferation of human colon, breast and ovarian cancer cells in vitro and suppresses tumour growth in mice when introduced intratumourally. The present study assessed the effect of SRL on tumour growth when introduced intraperitoneally in BALB/c nude mice and investigated the pharmacokinetics and biodistribution of SRL in Swiss albino mice. When 9 doses of SRL (30 mg/kg body weight/mice) was administered to BALB/c nude mice bearing human colon cancer HT-29 xenografts, a substantial reduction in tumour size was observed. A 35.8% reduction in tumour size was noted in the treated animals after 17 days. SRL treatment also inhibited angiogenesis, and the tumours from the treated animals were observed to carry fewer blood vessels and express less angiogenesis marker protein CD31, than that from the control animals. Pharmacokinetics and biodistribution analysis revealed that SRL was detected in the serum after 1 h and its level peaked after 24 h. SRL was not detected in any of the organs apart from the kidney where a trace amount was detected after 24 h of SRL injection. No significant changes were observed in any of the biochemical parameters tested including SGOT, SGPT, LDH, CREAT and BUN in the SRL-treated mice compared to these levels in the controls. This suggests that SRL has good potential to be developed as a therapeutic agent for cancer treatment and warrant further investigations in vivo and subsequent clinical trials.

Introduction

Cancer is one of the major life threatening diseases worldwide (1). Colorectal cancer is the third leading cause of cancer-associated mortality in both men and women in the world (2). Regular screening increases the likelihood that colorectal cancer is detected at an early stage, more likely to be cured and with faster recovery (3). Although significant progress has been made over the past few decades, effective treatment of this disease is still lacking. It is believed that targeted therapy that focuses on specific molecules or signalling pathways hold the key for more effective treatment of this disease than the currently used chemotherapy.

Lectins, a group of highly diverse proteins of non-immune origin that are ubiquitous in nature, recognize specific carbohydrate structures. Tumour cells in general display different carbohydrate profiles on the cell surface compared to non-transformed cells which can be detected using lectins. Lectins can reasonably differentiate malignant tumours from normal cells, and hence are used for cancer diagnosis and some are also being studied for their possible use as therapeutic agents (4). Lectins from edible mushrooms, such as Agaricus bisporus, Agrocybe aegerita I & II, Kurkowa lectin Pleurotuseous, Tricholomamongolicum and Pholiotaadiposa, have been shown to exert antiproliferative effects in cancer in vitro and antitumor effects in vivo (511). The two main properties of lectins that is, selectivity and cytotoxicity, have been exploited for devising therapeutic strategies against cancer.

Sclerotium rolfsii, lectin (SRL) purified from the fungus Sclerotium rolfsii (12) has been shown to have exquisite binding specificity toward Thomsen-Friedenreich antigen (TF-Ag; Galβ1-3GalNAcα-O-Ser/Thr) and its derivatives (13). The TF antigen is highly expressed by >90% of tumours and rarely occurs in normal tissues (14). The crystal structure of SRL determined at 1.1 Å resolution, revealed two carbohydrate binding sites (15). Our previous studies have shown that SRL binds specifically to human cancerous colon but not to normal colon and exhibits strong antiproliferative activity in human colon cancer (HT29 and DLD1) (16), breast cancer (MCF-7 and ZR-75) and ovarian cancer (PA-1) cells by inducing cellular apoptosis (17,18). Recently, we demonstrated that the presence of SRL affects multiple cell signalling pathways with early effects on MAPK- and c-JUN-associated cell proliferation pathways followed by miRNA-associated cell activity and apoptosis pathways. Lectin affinity purification has extracted a total of 25 proteins in HT29 cells, including keratins, heat shock proteins HSP-60 and HSP-90, ATP synthase subunit α, mitochondrial retinal dehydrogenase 1, actin cytoplasmin 2, tubulins, pyruvate kinase-M, Annexin A2, peroxiredoxin-1, prohibitin, α-enolase and ADP/ATP translocase 2 that are known to be involved in cell survival, apoptosis and tumorigenesis (19,20). These effects of SRL together with its exquisite binding specificity towards TF-Ag-associated glycans prompted us to investigate the possibility of developing SRL as a therapeutic agent.

Materials and methods

Materials

Protease Inhibitor Cocktail Set III was obtained from Calbiochem (Darmstadt, Germany); CD-31 (PECAM-1) was obtained from Invitrogen (Carlsbad, CA, USA) and Dulbeccos modified Eagles medium (DMEM) and fetal calf serum (FCS) were obtained from Gibco-Invitrogen (Paisley, UK). Bovine serum albumin (BSA), streptavidin-horseradish peroxidase, 3,3,5,5-tetramethylbenzidine (TMB) and poly-L-lysine solution were obtained from Sigma-Aldrich (St. Louis, MO, USA). Taq DNA polymerase was obtained from Invitrogen. Oligonucleotide primers and Tris buffer were obtained from Sigma-Aldrich and dNTPs from Fermentas (Waltham, MA, USA). 3-3-Diaminobenzidine chromogen/H2O2 substrate in buffer solution (pH 7.5) (DAB kit) and rabbit anti-SRL polyclonal antisera were obtained from Bangalore Genei India Pvt. Ltd. (Bangalore India). Caspase-3 (3G2) mouse mAb was procured from Cell Signaling Technology, Inc. (Danvers, MA, USA). Species-specific HRP-labelled secondary antibodies were procured from Bio-Rad Laboratories (Hercules, CA, USA). Anticoagulated tubes were obtained from Greiener Bio-One (1 ml K3E K3EDTA; Frickenhausen, Germany) and a flat bottom 96-well micro-test plate were obtained from Tarsons (Kolkata, India). All other reagents were of analytical grade and Milli-Q water was used for all the preparations.

Purification of Sclerotium rolfsii lectin (SRL)

Purification of SRL from the sclerotial bodies was carried out as previously described (12). Briefly, SRL was extracted from powdered sclerotial bodies with 50 mM acetate buffer containing 100 mM NaCl (pH 4.3) and subjected to 30% methanol precipitation followed by ion exchange chromatography on CM cellulose matrix and finally purified on a Superdex G-75 gel filtration column. The purified lectin was used for biodistribution and efficacy studies. The lectin activity was routinely determined by the 2-fold serial dilution method using trypsinised human erythrocytes.

Purification of polyclonal SRL antibodies from Protein A agarose column

Custom designed anti-SRL polyclonal antibodies were purified from immunized rabbit serum (from Bangalore Genei India) by subjecting to 50% ammonium sulphate precipitation, followed by affinity chromatography on a Protein A agarose column equilibrated with 100 mM phosphate buffer (pH 7.2) containing 150 mM NaCl phosphate-buffered saline (PBS). Bound SRL antibodies were eluted with 100 mM glycine-HCl (pH 2.5), dialysed against PBS and stored at −20°C.

Cell culture

Human colon cancer HT29 cells were obtained from the European Cell Culture Collection via the Public Health Laboratory Service (Porton Down, UK). HT29 cells were cultured in DMEM supplemented with 10% FBS, 100 units/ml penicillin and 100 µg/ml streptomycin (complete DMEM) at 37°C in 5% CO2.

Mice

All animal experiments were carried out at the Advanced Centre for Treatment, Research and Education in Cancer with approval from the Ethics Committee (IRB approval no. 07/2012) of Tata Memorial Centre-ACTREC-IAEC (Institutional Animal Ethics Committee). Inbred female BALB/c nude mice (6–8 weeks old, weighing 18–22 g) for efficacy studies and male Swiss albino mice (6–8 weeks old, weighing 20–25 g) for pharmacokinetics and biodistribution studies were used. The animals were housed in groups of 4–5 per polycarbonate cage under individually ventilated caging systems (IVCs) under controlled conditions of 55±5% humidity, 22±2°C temperature, and light cycle (12:12 h). All mice were provided with easy access to in-house pelleted feed and with ad libitum UV-treated water and were monitored daily for general health status.

Efficacy studies of SRL on tumour growth in BALB/c nude mice

HT29 cells were suspended at 1×107 cells/ml in DMEM and a cell suspension (0.1 ml) was injected subcutaneously into two donor mice. After 4 weeks, the tumours from these donor mice were excised, chopped into 2- to 3-mm fragments and a single piece of tumour was transplanted subcutaneously into each of 12 BALB/c nude mice. Tumours were allowed to grow to reach the appropriate size of 5–6 mm, i.e. 80–120 mm3 in volume and animals were divided randomly into 2 groups of 5 mice in each group, one for SRL treatment (T) and other used as a control (TBS, C). Mice were intraperitoneally administered SRL (30 mg/kg body weight/mouse) in TBS (pH 7.5) on every other days for 17 days for a total of 9 doses. The mice in the control group were injected intraperitoneally with TBS at the same units (5–10 UI) and maintained for 2 weeks after the last dose. The mice were weighed and tumour volume was also determined by measuring the length (L) and width (W) of the tumour after each injection. The tumour volume at day n (TVn) was calculated as TV (mm3) = (L×W2)/2. The relative tumour volume at day n (RTVn) vs. day 0 was expressed according to the formula: RTVn = TVn/TV0 (21,22). Regression of tumour T/C (%) in the treated vs. control mice was calculated using: T/C (%) = (mean RTV of treated/control group) × 100. The changes in the tumour size of the treated mice at different time-points compared to the control were used for monitoring the efficacy of the lectin. Mice were sacrificed 2 weeks after the last dose, and whole blood was collected in tubes coated with anticoagulant and used for analysis of total blood cell count, hemoglobin content, WBC and platelet count. Tumours were excised and organs such as liver, kidney and lung were collected directly in PBS to remove the blood traces, wrapped in aluminium foil and stored at −80°C until further use for immunohistological analysis. Excised tumours were also snap frozen and stored at −80°C for STR analysis.

PCR analysis of short tandem repeat (STR)

In order to ascertain the human origin of the tumours using short tandem repeat (STR) method (23), a small piece of the tumour tissues was collected from BALB/c mice with HT-29 xenograft, excised after 2 weeks from the last dose of injection. These tissues were snap frozen and stored at −80°C for extraction of DNA. DNA was extracted from the samples using phenol chloroform extraction followed by ethanol precipitation. The DNA precipitates were suspended in Tris-EDTA (TE) buffer, pH 8.4. The extracted DNA was subjected to PCR amplification using human-specific microsatellite primers (forward, 5′-TGCCATTTGTGGGTACATTC-3′ and reverse, 5′-TTGTGTTTCTTTTTCTGTTCCTACA-3′). In brief, 100 ng of target DNA was amplified in a 15-µl reaction volume containing 5 IU of Taq DNA polymerase, 0.4 pM each oligonucleotide primer, 10 mM Tris buffer, 3.5 mM MgCl2, 0.4 mM each of dNTPs. PCR was performed with a 5-min denaturation at 94°C followed by 34 cycles of a 20-sec denaturation at 94°C, a 20-sec annealing at 58°C, and a 30-sec elongation at 72°C in a thermal cycler (Eppendorf, Hauppauge, NY, USA). Amplicon was separated on 2% agarose gel and visualized by UV light.

Pharmacokinetics and biodistribution studies

Swiss albino mice were randomized into two groups, the control group (TBS) and the SRL-treated group which included 4 time-points (1, 6, 24 and 48 h) with 3 animals for each time-point. Lectin was prepared in TBS (pH 7.5), hence TBS served as the vehicle control. Animals were injected intraperitoneally with TBS and SRL (24 mg/kg body weight/mouse) and maintained for different time-points and blood was collected from the treated and control mice by retro-orbital bleeding. After incubation of the blood at room temperature (RT) for 15 min, serum was collected by centrifuging the sample at 2,500 rpm for 15 min at 4°C and stored at −80°C until further use. The pharmacokinetic profile of the serum samples was evaluated by ELISA. Serum samples collected at 24 h were also analysed for biochemical parameters of toxicity such as serum liver enzymes (aspartate transaminase and alanine transaminase), lactate dehydrogenase (LDH), blood urea nitrogen (BUN) and creatinine levels [carried out at Unique Bio Diagnostics Enterprises (Mumbai, India)]. Animals were then sacrificed by cervical dislocation, and the organs (liver, kidney, heart, stomach, lungs, spleen, intestine and brain) were collected directly in PBS to remove the blood traces, wrapped in aluminium foil and stored at −80°C until further use for biodistribution studies by ELISA or histology.

Tissue homogenization

Tissue homogenate was prepared by taking a part of the tissue from each organ, thawed on ice, weighed in a 2-ml Eppendorf tube and extracted in extraction buffer, PBS pH 7.4 (four volume/g of organ weight). Protease Inhibitor Cocktail Set III was added to the extraction buffer (1/20th volume of the tissue weight) and subjected to homogenization using an electric homogenizer (24,25). Prior to homogenization, the probe was dipped in ice and the tissue was homogenized at 4°C with 8–10 oscillate up- and down-strokes for soft tissues derived from organs such as the liver, stomach, small intestine, lung, spleen, kidney and brain; whereas, hard tissues from the heart required more strokes and agitation. After homogenization, each sample was centrifuged for 20 min at 16,000 rpm at 4°C and the supernatant was collected and stored at −80°C until further use for analysis.

Bioanalytical validation

A calibration curve was constructed using different concentrations of SRL (0.002–0.312 µg/ml and 0.002–0.078 µg/ml) including a lower limit of quantification (LLOQ) for evaluating SRL in organ and serum extracts, respectively. Analytical recovery by ELISA was evaluated by adding serum and organ extracts at a 1:100 dilution and using the respective calibration curves and measuring its recovery using the same procedure and conditions as described in the ELISA experiments. The absorbance was measured at 450 nm and values were plotted against the concentrations. The LLOQ was set as the lowest measurable concentration with acceptable accuracy and precision not >20% of the expected values (26,27). Precision and accuracy were determined by analyzing quality control samples (QCs). Recovery was computed by comparing responses in triplicate of extracted QCs (28,29).

ELISA

Indirect ELISA was performed using 96-well plates. The plates were pre-coated with a known concentration of SRL (0.002–0.3 µg/100 µl/well), serum samples or tissue homogenate (1:100 dilution) in coating buffer (15 mM sodium carbonate and 17 mM bicarbonate buffer, pH 9.6, containing 3 mM sodium azide) and incubated overnight at 4°C. Wells were washed thrice with 50 mM PBS, at pH 7.4 containing 0.1% (v/v) Tween-20 (PBST) to remove the unbound proteins. Non-specific binding was blocked by incubating with 100 µl of 1% BSA in PBS for 2 h at RT at 37°C, followed by washing twice with PBST to remove excess blocking agent. Wells were incubated with rabbit anti-SRL polyclonal antibodies (100 µl/well, 1:10,000 dilutions in PBS) overnight at 4°C. After washing, HRP-conjugated anti-rabbit IgG secondary antibodies (Sigma-Aldrich) at 1:10,000 dilutions were added and incubated for 2 h at RT at 37°C. The colour was developed by the addition of 3,3,5,5-tetramethylbenzidine (TMB; Sigma-Aldrich). The reaction was stopped using 4 M H2SO4 (100 µl/well) and the absorbance was recorded at 450 nm on a microtiter plate reader (Tecan, Crailsheim, Germany).

Histological analysis

Histochemical examinations were performed using the tissue samples of the tumors and organs including liver, spleen, kidney, heart, lung, stomach, small intestine and brain. Excised tumours were collected and fixed in 10% formalin and embedded in paraffin blocks, and 5-µm sections were used for hematoxylin and eosin (H&E) staining and for immunohistochemistry (IHC). The histological sections were observed under an optical microscope and photographed.

Immunohistochemical staining

Sections (5-µm thick) of the SRL-treated tumours with the controls were prepared by using the tumour tissues collected from the snap-frozen samples stored at −80°C and stained with a specific mouse anti-CD31 (PECAM-1) antibody (clone MEM-05) according to the manufacturers instructions. The primary antibody (1:500) was diluted in PBS and incubated with samples at RT for 1 h followed by washing with TBST and then with HRP-conjugated goat anti-mouse IgG (H+L) secondary antibodies (1:3,000) diluted in PBS and incubated at RT for 1 h. Finally the slides were developed using 3,3-diaminobenzidine (DAB) substrate and counterstained with hematoxylin. Images (magnification, ×10) were captured to monitor CD31 expression.

Statistical analysis

The antitumor activity (as measured by differences in tumor mass) was expressed as mean ± standard deviation (SD). In vivo data and ELISA calibration curve with unknowns was analyzed using GraphPad Prism 6. The statistical significance of differences in tumor volume was determined by one-way analysis of variance (ANOVA), followed by Sidak test for three or more groups; a value of P<0.05 was considered as significant.

Results

SRL inhibits tumour growth in BALB/c nude mice bearing HT-29 xenografts

Mice injected intraperitoneally with SRL (30 mg/kg body weight/mouse) showed significant inhibition (35.8%, P<0.0001) of tumour growth when compared with that observed in the control group (Fig. 1). The average initial tumour volume in the TBS and SRL-treated groups was 155±4.7 and 147±10.9 mm3, respectively before tumour cell treatment and the average tumour volume after completion of 9 doses was 353±71.2 and 171±16 mm3, respectively (P<0.001). No noticeable change in the weight and tumour volume of either control or treated group was observed after maintaining treatment for 14 days. The histology of the tumour sections of the SRL-treated and untreated animals showed marked differences. The SRL-treated mouse tumours appeared more like normal ones while the TBS-treated control mouse tumours showed a characteristic tumour appearance. The histology of liver sections showed that SRL-treated mice appeared normal compared to the control-treated mice bearing tumours, indicating the tumour-regressing effect of SRL (Fig. 2). Moreover, all the treated animals were healthy without any significant changes in body weight compared to the controls. Complete blood count (CBC) revealed no detectable changes in the SRL-treated and TBS-treated groups (Table I). PCR analysis of the tumours obtained from the experimental mice showed similar product size of 275 bp as that of the implanted HT29 cells, indicating that the tumours which developed in nude mice were from the implanted human cancer cells and not from the mice (Fig. 3).

Table I.

Toxicity studies of SRL in BALB/c nude mice.

Table I.

Toxicity studies of SRL in BALB/c nude mice.

ParametersSRL (750 µg/mice)Control (TBS)
Initial body weight (g)20.3120.62
Final body weight (g)19.9720.94
MortalityNoneNone
RBC (106/µl)9.6910.33
WBC (103/µl)1.811.38
Platelet count (103/µl)1009990
Hemoglobin (g/dl)14.415.73

[i] RBC, red blood corpuscles; WBC, white blood corpuscles.

Anti-angiogenic effect of SRL

SRL-treated animals also showed clear reduction in the number of blood vessels in the excised tumours compared with that from the control-treated animals (Fig. 1), indicating an effect of SRL treatment on suppression of angiogenesis. The anti-angiogenesis effect of SRL was also supported by the immunohistological analysis in which the expression of PECAM (CD31), an angiogenesis marker, was observed to be much lower in the SRL-treated animals than that in the control-treated animals (Fig. 2B).

Pharmacokinetic profile

Pharmacokinetic profile of SRL in Swiss albino mice was obtained by evaluation of serum samples from the SRL-treated mice by ELISA. Mice injected with SRL (24 mg/kg body weight/mouse) intraperitoneally showed maximal SRL concentrations in serum at 24 h (8.02 µg/ml) (Fig. 4A). These results indicate that SRL was not accumulated in the body and was eliminated. Biochemical parameters in serum samples collected at 24 h and analysed for hepatic function namely SGPT, SGOT, bilirubin and cholesterol exhibited no significant alterations between the SRL-treated mice and control-treated mice. Biochemical parameters related to kidney functions including urea, uric acid creatinine and total protein also showed no significant differences between the SRL-treated and control group (Table II). These results indicate the non-toxic nature of SRL treatment in the mice.

Table II.

Toxicity studies of SRL in Swiss albino mice.

Table II.

Toxicity studies of SRL in Swiss albino mice.

ParametersStd. valuesTBSSRL
OT IU/l54–29898.664.5
PT IU/l17–774325.5
CREAT mg/dl0.3–10.430.4
BUN mg/dl8–3340.733.6
LDH g/dl159–10451132.3557.5

[i] OT, aspartate aminotransferase; PT, alanine aminotransferase; CREAT, creatinine; BUN, blood urea nitrogen; LDH, lactate dehyrogenase.

In vivo biodistribution

The presence of SRL in various organs including the liver, kidney, lung, spleen, stomach, small intestine, heart and brain, respectively was analysed. The lectin was detected in trace amounts in the kidney but not in any other organ (Fig. 4B). SRL at 76, 113, 79 and 22 µg/g was detected in the kidney at 1, 6, 24 and 48 h, respectively. H&E staining of tissue sections of the different organs showed no morphological changes at 24 h between the SRL-treated or control-treated animals (Fig. 5). These results suggest that SRL is effectively cleared in the body and is non-toxic to mice.

Discussion

The present study reports that when administered intraperitoneally, SRL reduces tumour growth and inhibits angiogenesis in human colon cancer HT-29 cell xenografts in mice and is non-toxic. SRL was demonstrated to have good renal clearance and was eliminated without causing any sign of toxicity or significant changes in biochemical parameters of the mice.

Previous studies have shown that SRL induces cell apoptosis and inhibits cell growth in human colon cancer cells in vitro and suppresses tumour growth in NOD-SCID mice bearing HT-29 xenografts in vivo when injected intratumourally (16). SRL was shown to affect multiple signalling pathways in HT-29 cells by interacting with cell membrane proteins involved in cell survival, apoptosis and tumorigenesis (19,20). The present study revealed that intraperitoneal introduction of SRL suppressed tumour growth in the xenograft mouse model. H&E staining of the liver sections of the SRL-treated and control-treated mice showed no difference in morphology of the liver but the expression of CD-31 was substantially lower in the SRL-treated mice. This suggests that SRL treatment also inhibits angiogenesis. After SRL intraperitoneal injection, the level of SRL in the blood was seen to be peaked at 24 h. It is known that almost all drugs, chemicals and xenobiotics are eliminated through renal excretion; hence it was necessary to estimate the effects of SRL on kidney function (30,31). There were no changes in biochemical parameters in the SRL-treated mice compared to the control-treated mice group indicating no alterations in the animal hepatic and renal functions. Biodistribution analysis showed that SRL did not accumulated in any organ apart from its temporary appearance in the kidney. Pharmacokinetic and biodistribution patterns indicate that SRL has a very fast rate of absorption and is rapidly distributed to the entire body through the circulation. SRL appears to be quickly eliminated from the body through the kidney, indicating a beneficial property of SRL as a therapeutic agent.

SRL binds to the oncofetal TF antigen and TF-related carbohydrate structures (12,13). In colorectal cancer, increased expression of oncofetal TF and sialylated Tn is common (3237) and can be detected using lectins including peanut lectin, Arachis hypogea (PNA) (38) and Sambucus nigra lectin (SNA). Many lectins have shown anticancer effects when tested in vitro and in some cases also in vivo but not many have reached clinical trials partly due to their cytotoxicity (39). Tn antigen (GalNAc-Ser/Thr)-binding Helix pomatia lectin (HPA) identifies a subset of metastatic colorectal cancer. HPA binding also reveals overexpression of sialyl-Lewis X carbohydrate antigen which is correlated with poor prognosis of colorectal cancer (40,41). Viscum album var. coloratum agglutinin (VCA), binding lectin from Korean mistletoe has been used in human clinical trials at low doses for the treatment of different cancers without serious side-effects and the action seems to be beneficial (39). A mannose/sialic acid-binding plant lectin from Polygonatum cyrtonema lectin (PCL) has shown antitumour effects by inducing apoptosis and autophagy in cancer cells (42,43). Two cytotoxic mistletoe isolectins designated as KML-IIU and KML-IIL were found to exhibit cytotoxicity in various human and mouse cancer cell lines (44). Mistletoe lectin has been extensively studied for its clinical use and has reached clinical trials (45,46). Reports are available on the antitumor effects of Concanavalin A lectin, one of the most extensively studied lectin. Clinical use of Con A is questionable due to its strong cytotoxic effects including the induction of hepatitis (47,48).

The strong anti-proliferative, antitumour and anti-angiogenesis effects of SRL shown in vitro and in vivo in the present and previous studies with no detectable cytotoxic effects confirm the high potential of SRL as a therapeutic agent for cancer treatment.

Acknowledgements

The present study was supported by funding from the British Council under UKIERI, UGC under UPE and CPEPA and DBT-BIRAC programme.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

SRL

Sclerotium rolfsii lectin

HT-29

human colorectal adenocarcinoma grade II cell line

TF

Thomsen-Friedenreich antigen

AAL

Agrocybe aegerita lectin

ABL

Agaricus bisporus lectin

PNA

peanut agglutinin

SNA

Sambucus nigra lectin

HPA

Helix pomatia lectin

VCA

Viscum album var. coloratum agglutinin

PCL

Polygonatum cyrtonema lectin

Con A

Concanavalin A lectin

BSA

bovine serum albumin

DMEM

Dulbeccos modified Eagles medium

FCS

fetal calf serum

GalNAc

N-acetyl-D-galactosamine

GlcNAc

N-acetyl-D-glucosamine

HRP

horseradish peroxidase

PBS

phosphate-buffered saline

SDS

sodium dodecyl sulphate

TBS

Tris-buffered saline

RT

room temperature

PBST

phosphate-buffered saline Tween-20

References

1 

World Health Organization: Global Health Observatory data repository 2011. Number of deaths (world) by cause; http://apps.who.int/gho/data/node.main.CODWORLD?lang=enAccessed. October 31–2013

2 

American Cancer Society: Cancer Facts & Figures 2015. US Mortality Data, National Center for Health Statistic Center for Disease, Control & Prevention. 2015.https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2015.html

3 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin MD, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Bies C, Lehr CM and Woodley JF: Lectin-mediated drug targeting: History and applications. Adv Drug Deliv Rev. 56:425–435. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Yu LG, Fernig DG, White MR, Spiller DG, Appleton P, Evans RC, Grierson I, Smith JA, Davies H, Gerasimenko OV, et al: Edible mushroom (Agaricus bisporus) lectin, which reversibly inhibits epithelial cell proliferation, blocks nuclear localization sequence-dependent nuclear protein import. J Biol Chem. 274:4890–4899. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Yu L, Fernig DG, Smith JA, Milton JD and Rhodes JM: Reversible inhibition of proliferation of epithelial cell lines by Agaricus bisporus (edible mushroom) lectin. Cancer Res. 53:4627–4632. 1993.PubMed/NCBI

7 

Zhao C, Sun H, Tong X and Qi Y: An antitumour lectin from the edible mushroom Agrocybe aegerita. Biochem J. 374:321–327. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Jiang S, Chen Y, Wang M, Yin Y, Pan Y, Gu B, Yu G, Li Y, Wong BH, Liang Y, et al: A novel lectin from Agrocybe aegerita shows high binding selectivity for terminal N-acetylglucosamine. Biochem J. 443:369–378. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Wang HX, Liu WK, Ng TB, Ooi VE and Chang ST: The immunomodulatory and antitumor activities of lectins from the mushroom Tricholoma mongolicum. Immunopharmacology. 31:205–211. 1996. View Article : Google Scholar : PubMed/NCBI

10 

Li YR, Liu QH, Wang HX and Ng TB: A novel lectin with potent antitumor, mitogenic and HIV-1 reverse transcriptase inhibitory activities from the edible mushroom Pleurotus citrinopileatus. Biochim Biophys Acta. 1780:51–57. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Zhang GQ, Sun J, Wang HX and Ng TB: A novel lectin with antiproliferative activity from the medicinal mushroom Pholiota adiposa. Acta Biochim Pol. 56:415–421. 2009.PubMed/NCBI

12 

Swamy BM, Hegde GV, Naik RS and Inamdar SR: T-antigen binding lectin from the phytopathogenic fungus Sclerotium rolfsii. Lect. Biol Biochem Clin Biochem. 15:45–55. 2001.

13 

Chachadi VB, Inamdar SR, Yu LG, Rhodes JM and Swamy BM: Exquisite binding specificity of Sclerotium rolfsii lectin toward TF-related O-linked mucin-type glycans. Glycoconj J. 28:49–56. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yu LG: The oncofetal Thomsen-Friedenreich carbohydrate antigen in cancer progression. Glycoconj J. 24:411–420. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Leonidas DD, Swamy BM, Hatzopoulos GN, Gonchigar SJ, Chachadi VB, Inamdar SR, Zographos SE and Oikonomakos NG: Structural basis for the carbohydrate recognition of the Sclerotium rolfsii lectin. J Mol Biol. 368:1145–1161. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Inamdar SR, Savanur MA, Eligar SM, Chachadi VB, Nagre NN, Chen C, Barclays M, Ingle A, Mahajan P, Borges A, et al: The TF-antigen binding lectin from Sclerotium rolfsii inhibits growth of human colon cancer cells by inducing apoptosis in vitro and suppresses tumor growth in vivo. Glycobiology. 22:1227–1235. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Savanur MA, Eligar SM, Pujari R, Chen C, Mahajan P, Borges A, Shastry P, Ingle A, Kalraiya RD, Swamy BM, et al: Sclerotium rolfsii lectin induces stronger inhibition of proliferation in human breast cancer cells than normal human mammary epithelial cells by induction of cell apoptosis. PLoS One. 9:e1101072014. View Article : Google Scholar : PubMed/NCBI

18 

Eligar SM, Pujari R, Swamy BM, Shastry P and Inamdar SR: Sclerotium rolfsii lectin inhibits proliferation and induces apoptosis in human ovarian cancer cell line PA-1. Cell Prolif. 45:397–403. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Barkeer S, Guha N, Hothpet V, Adavigowda Saligrama D, Hegde P, Padmanaban A, Yu LG, Swamy BM and Inamdar SR: Molecular mechanism of anticancer effect of Sclerotium rolfsii lectin in HT29 cells involves differential expression of genes associated with multiple signaling pathways: A microarray analysis. Glycobiology. 25:1375–1391. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Barkeer S, Gudihal R, Eligar SM, Hegde P, Lu GY, Bale MS and Inamdar SR: Identification and characterization of Sclerotium rolfsii lectin (SRL) binding proteins from human colon epithelial cancer HT29 cells. Translational Biomedicine ISSN 2172-0479. 2015.

21 

Sharma SK, Pedley RB, Bhatia J, Boxer GM, El-Emir E, Qureshi U, Tolner B, Lowe H, Michael NP, Minton N, et al: Sustained tumor regression of human colorectal cancer xenografts using a multifunctional mannosylated fusion protein in antibody-directed enzyme prodrug therapy. Clin Cancer Res. 11:814–825. 2005.PubMed/NCBI

22 

Vallespí MG, Pimentel G, Cabrales-Rico A, Garza J, Oliva B, Mendoza O, Gomez Y, Basaco T, Sánchez I, Calderón C, et al: Antitumor efficacy, pharmacokinetic and biodistribution studies of the anticancer peptide CIGB-552 in mouse models. J Pept Sci. 20:850–859. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ingle AD and Hosetti BB: Use of immuno-compromised mouse model for establishment and study of human animal tumours. Indian J Vet Pathol. 34:156–161. 2010.

24 

Simpson RJ: Homogenization of mammalian tissue. Cold Spring Harb Protoc pdb prot 5455. 2010.https://doi.org/10.1101/pdb.prot5455https://doi.org/10.1101/pdb.prot5455. View Article : Google Scholar

25 

Burden DW: Guide to the Homogenization of Biological Samples. Random Primers. 7:2008.1–14, 2008. http://www.opsdiagnostics.com/notes/ranpri/Homogenization%20Guide%20ver.pdf

26 

Hartmann C, Smeyers-Verbeke J, Massart DL and McDowall RD: Validation of bioanalytical chromatographic methods. J Pharm Biomed Anal. 17:193–218. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Ahn JE, Karlsson MO, Dunne A and Ludden TM: Likelihood based approaches to handling data below the quantification limit using NONMEM VI. J Pharmacokinet Pharmacodyn. 35:401–421. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Jusko WJ: Use of pharmacokinetic data below lower limit of quantitation values. Pharm Res. 29:2628–2631. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Beal SL: Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn. 28:481–504. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Sallam H, El-Serafi I, Meijer L and Hassan M: Pharmacokinetics and biodistribution of the cyclin-dependent kinase inhibitor -CR8- in mice. BMC Pharmacol Toxicol. 14:502013. View Article : Google Scholar : PubMed/NCBI

31 

Khlebtsov N and Dykman L: Biodistribution and toxicity of engineered gold nanoparticles: A review of in vitro and in vivo studies. Chem Soc Rev. 40:1647–1671. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Ghazarian H, Idoni B and Oppenheimer SB: A glycobiology review: Carbohydrates, lectins and implications in cancer therapeutics. Acta Histochem. 113:236–247. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Campbell BJ, Finnie IA, Hounsell EF and Rhodes JM: Direct demonstration of increased expression of Thomsen-Friedenreich (TF) antigen in colonic adenocarcinoma and ulcerative colitis mucin and its concealment in normal mucin. J Clin Invest. 95:571–576. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Singh R, Campbell BJ, Yu LG, Fernig DG, Milton JD, Goodlad RA, FitzGerald AJ and Rhodes JM: Cell surface-expressed Thomsen-Friedenreich antigen in colon cancer is predominantly carried on high molecular weight splice variants of CD44. Glycobiology. 11:587–592. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Hanisch FG and Baldus SE: The Thomsen-Friedenreich (TF) antigen: A critical review on the structural, biosynthetic and histochemical aspects of a pancarcinoma-associated antigen. Histol Histopathol. 12:263–281. 1997.PubMed/NCBI

36 

Almogren A, Abdullah J, Ghapure K, Ferguson K, Glinsky VV and Rittenhouse-Olson K: Anti-Thomsen-Friedenreich-Ag (anti-TF-Ag) potential for cancer therapy. Front Biosci (Schol Ed). 4:840–863. 2012.PubMed/NCBI

37 

Belov L, Zhou J and Christopherson RI: Cell surface markers in colorectal cancer prognosis. Int J Mol Sci. 12:78–113. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Singh R, Subramanian S, Rhodes JM and Campbell BJ: Peanut lectin stimulates proliferation of colon cancer cells by interaction with glycosylated CD44v6 isoforms and consequential activation of c-Met and MAPK: Functional implications for disease-associated glycosylation changes. Glycobiology. 16:594–601. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Yau T, Dan X, Ng CC and Ng TB: Lectins with potential for anti-cancer therapy. Molecules. 20:3791–3810. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Mitchell BS and Schumacher U: The use of the lectin Helix pomatia agglutinin (HPA) as a prognostic indicator and as a tool in cancer research. Histol Histopathol. 14:217–226. 1999.PubMed/NCBI

41 

Peiris D, Ossondo M, Fry S, Loizidou M, Smith-Ravin J and Dwek MV: Identification of O-linked glycoproteins binding to the lectin Helix pomatia agglutinin as markers of metastatic colorectal cancer. PLoS One. 10:e01383452015. View Article : Google Scholar : PubMed/NCBI

42 

Liu B, Wu JM, Li J, Liu JJ, Li WW, Li CY, Xu HL and Bao JK: Polygonatum cyrtonema lectin induces murine fibrosarcoma L929 cell apoptosis and autophagy via blocking Ras-Raf and PI3K-Akt signaling pathways. Biochimie. 92:1934–1938. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Wang SY, Yu QJ, Bao JK and Liu B: Polygonatum cyrtonema lectin, a potential antineoplastic drug targeting programmed cell death pathways. Biochem Biophys Res Commun. 406:497–500. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Kang TB, Song SK, Yoon TJ, Yoo YC, Lee KH, Her E and Kim JB: Isolation and characterization of two Korean mistletoe lectins. J Biochem Mol Biol. 40:959–965. 2007.PubMed/NCBI

45 

Yoon TJ, Yoo YC, Kang TB, Song SK, Lee KB, Her E, Song KS and Kim JB: Antitumor activity of the Korean mistletoe lectin is attributed to activation of macrophages and NK cells. Arch Pharm Res. 26:861–867. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Rostock M, Huber R, Greiner T, Fritz P, Scheer R, Schueler J and Fiebig HH: Anticancer activity of a lectin-rich mistletoe extract injected intratumorally into human pancreatic cancer xenografts. Anticancer Res. 25:1969–1975. 2005.PubMed/NCBI

47 

Miyagi T, Takehara T, Tatsumi T, Suzuki T, Jinushi M, Kanazawa Y, Hiramatsu N, Kanto T, Tsuji S, Hori M, et al: Concanavalin a injection activates intrahepatic innate immune cells to provoke an antitumor effect in murine liver. Hepatology. 40:1190–1196. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Lei HY and Chang CP: Lectin of Concanavalin A as an anti-hepatoma therapeutic agent. J Biomed Sci. 16:102009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 37 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Anupama S , Laha P, Sharma M, Pathak K, Bane S, Ingle AD, Gota V, Kalraiya RD, Yu L, Rhodes JM, Rhodes JM, et al: Pharmacokinetics, biodistribution and antitumour effects of Sclerotium rolfsii lectin in mice. Oncol Rep 37: 2803-2810, 2017
APA
Anupama S, ., Laha, P., Sharma, M., Pathak, K., Bane, S., Ingle, A.D. ... Inamdar, S.R. (2017). Pharmacokinetics, biodistribution and antitumour effects of Sclerotium rolfsii lectin in mice. Oncology Reports, 37, 2803-2810. https://doi.org/10.3892/or.2017.5545
MLA
Anupama S, ., Laha, P., Sharma, M., Pathak, K., Bane, S., Ingle, A. D., Gota, V., Kalraiya, R. D., Yu, L., Rhodes, J. M., Swamy, B. M., Inamdar, S. R."Pharmacokinetics, biodistribution and antitumour effects of Sclerotium rolfsii lectin in mice". Oncology Reports 37.5 (2017): 2803-2810.
Chicago
Anupama S, ., Laha, P., Sharma, M., Pathak, K., Bane, S., Ingle, A. D., Gota, V., Kalraiya, R. D., Yu, L., Rhodes, J. M., Swamy, B. M., Inamdar, S. R."Pharmacokinetics, biodistribution and antitumour effects of Sclerotium rolfsii lectin in mice". Oncology Reports 37, no. 5 (2017): 2803-2810. https://doi.org/10.3892/or.2017.5545