You are on page 1of 31

364 S. Banerjee and S.B.

Paruthy

due to multiple mechanisms which include the induction of death receptors and
proapoptotic members of Bcl-2 family and inhibition of antiapoptotic Bcl-2 pro-
teins and activation of caspase(s). Also, by sulforaphane pretreatment, enhance-
ment of the inhibitory effect of TRAIL on PC-3 colony formation and sensitization
of TRAIL-resistant LNCaP colonies convey the message that sulforaphane aug-
ments the ability of TRAIL to inhibit colony growth. Additional data suggest that
upregulation of death receptors DR4 and DR5 by sulforaphane may be one of the
mechanisms that reinforce the proapoptotic effects of TRAIL in PC-3 cells and
sensitize TRAIL-resistant LNCaP cells. In vivo, sulforaphane inhibits the growth
of PC-3 cells orthotopically implanted in nude mice by inducing apoptosis and
inhibiting tumor cell proliferation. It has been recorded to inhibit the expression of
proteins related to invasion, metastasis, and angiogenesis by inhibiting phosphor-
ylation of AKT, ERK1/2, and FOXO3a and activation of NF-κB [130]. These
provide strong and compelling preclinical evidence that sulforaphane either alone
or in combination with TRAIL can be used to prevent and/or treat prostate cancer.

5 SFN, TRAIL, and Cancer Stem Cells

5.1 Prostate Cancer

An extension to the above report in support of SFN-sensitizing prostate cancer cells


to TRAIL-induced apoptosis, apoptosis-resistant cancer stem cells (CSCs) have been
identified in advanced androgen-independent prostate cancer (AIPC) that are not
eliminated by currently available therapeutics. Thus, the disease relapses due to the
ability of CSCs to self-renew and differentiate into multiple cell types populating
with all the cellular subtypes of the original tumor. In particular, these cells over-
express CSC markers including ALDH1, CD44, CD133, and c-Met [131]. Therefore,
numerous investigators pursued investigating innovative approach combining
TRAIL and bioactive natural compounds aimed to eliminate CSCs. In line with
such objective and utilizing advanced androgen-independent prostate cancer (AIPC)
cell lines – DU145 and PC3 – with highly enriched CSC characteristics and primary
patient-derived prostate CSCs, Labsch et al. revealed a synergistic action between
SFN and recombinant soluble TRAIL when used in combination [132]. The authors
built their conclusion from results comparing the effects of the drugs either as
monotherapy or in combination using multitude of in vitro assays including
NF-κB activity, self-renewal and differentiation potential, stem cell signaling via
spheroid, colony-forming assays, and antibody protein array. Mechanistically, the
dominance of a therapeutically relevant effect associated with the combination
regimen is a result of the inhibition of TRAIL-induced NF-κB activity by SFN
and reduction in the levels of Oct-3/4, HNF-3β, PDX-1, Otx2, TP63, GSC, Snail,
VEGFR-2, and HCG relative to monotherapy. These proteins relate to requirement
for self-renewal, differentiation, cell migration, and epithelial-mesenchymal transi-
tion (EMT) and tumorigenesis. Additionally, it has been noted that the expression of
CXCR4 receptor which is involved in migration and metastasis becomes inhibited
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 365

following sulforaphane-only treatment and the combination with TRAIL further


reduces the expression of this receptor. Similar results were noticed for the Notch-
1 receptor and its ligand, Jagged-1, which are regulators of asymmetric and sym-
metric division, progression, and metastasis in prostate cancer. Correspondingly, the
inhibition of SOX 2 and Nanog expression which are important regulators of self-
renewal potential was much stronger in the combination group. ALDH1 activity
which represents differentiation and self-renewal potential was also significantly
inhibited by the combination regimen attesting strong inhibition of stem cell signal-
ing potential. Furthermore, in an in vivo tumor engraftment model, TRAIL and SFN
together inhibited tumor growth, reduced CSC marker expression, and eliminated
primary prostate CSCs synergistically via the induction of apoptosis and inhibited
the proliferation and CSC marker expression. Conclusively, these findings suggest
that sulforaphane shifts the balance from (TRAIL-induced) survival signaling to
apoptosis and explains the observed synergistic effect.

5.2 Pancreatic Cancer

To address the need to eliminate oncogenically transformed CSC in pancreatic


cancer (PaCa), Kallifatidis et al. pointed that pancreatic tumors harbor tumor-initi-
ating cells (TICs; aka CSC), wherein TRAIL-activated NF-κB signaling acts as a
contributor to resistance to apoptosis induction by TRAIL [109]. The reported
anticancer activity of SFN in pancreatic cancer stem cells has been detailed in the
preceding section [109]. Of interest, sulforaphane allegedly prevents NF-κB binding
and thus is able to arrogate resistance to chemotherapy without cytotoxicity to
normal cells. In a xenograft model, sulforaphane strongly blocked tumor growth
and angiogenesis, while combination with TRAIL levied an additive effect without
any obvious cytotoxicity to normal cells [109]. Further, primary tumor cells isolated
from tumors freshly resected from patients with pancreatic cancer expressing
markers for TICs could also be sensitized by sulforaphane for TRAIL-induced
cytotoxicity; together these findings provide new insights into resistance mecha-
nisms of TICs and suggest the combination of sulforaphane with TRAIL as a
promising strategy for eliminating pancreatic TICs.

6 Sulforaphane Combination with Other Natural Agents


in Cancer Therapy

Against the backdrop of foregoing thought-provoking reports from laboratory inves-


tigations confirming that SFN intercedes in sensitizing different treatment modalities
including TRAIL therapy, anticipations from combination of SFN and other well-
characterized chemopreventive bioactive natural compounds aiming to increase the
antitumor response became the focus of several reports. Based on scientific rationale
that health benefits of fruits and vegetables are from additive and synergistic
combination of phytochemicals, SFN when combined with other bioactive
366 S. Banerjee and S.B. Paruthy

phyto-compounds also displays promising results against various cancers. Such effect
may conceivably be due mainly because of the induction of a broad spectrum of anti-
cancer pathways. This concept is well illustrated in study reporting chemoprevention
of familial adenomatous polyposis (FAP) by combination of sulforaphane and
dibenzoylmethane in ApcMin/+ mouse colorectal cancer model that mimics the
human familial adenomatous polyposis (FAP) that progresses sporadically to colo-
rectal cancer with early Apc mutation [133]. In a prior study on human colon
carcinoma HT-29 cells, SFN shows growth inhibition by inducing G1 cell cycle
arrest and apoptosis, probably through the regulation of mitogen-activated protein
kinase (MAPK) without affecting the Akt pathway [134]. On the other hand,
dibenzoylmethane (DBM) – a minor constituent of licorice, inhibits cancer cell
growth by inhibiting Akt pathway activity. Thus, a study that included a combination
treatment regimen of SFN and DBM at half of the single dose was designed to
investigate their potential synergistic effect on colon cancer chemoprevention in
ApcMin/+ mouse model [133]. The results showed that dietary administrations of
SFN and DBM combination significantly inhibits the development of intestinal
adenomas and block the colon tumor development by inhibition of cell survival
and growth-related cell signaling pathways (such as Akt and extracellular signal-
regulated kinase) and induction of apoptosis and other related biomarkers (including
arachidonic acid metabolism, proliferating cell nuclear antigen, cleaved caspases,
cyclin D1, and p21) conceptually supporting the hypothesis that both SFN and DBM
in combination are potent natural dietary compounds in chemoprevention of gastro-
intestinal cancers. Moreover, the protective ability of glucosinolates due to syner-
gistic effect with other constituents in the diet cannot be ignored. In this direction,
SFN in combination with tea polyphenol – epigallocatechin (EGCG) – has been
testified in ovarian, prostate, and colon cancers [135–137]. Previous published
epidemiological studies indicate that consumption of green tea and cruciferous
vegetables is inversely associated with occurrence of ovarian cancer
[138–141]. Therefore, the combinatorial effect and underlying mechanism of
major components of green tea (epigallocatechin gallate – EGCG) and cruciferous
vegetables (sulforaphane) on ovarian and colon cancer cells as basis for innovative
therapeutic approach befitted as subject for further investigation [135]. Encouraging
data interestingly depicts that EGCG potentiates the inhibiting effect of SFN on
paclitaxel-sensitive and paclitaxel-resistant ovarian cancer cell lines in time- and
dose-dependent manner. Further examination points to SFN action in arresting
ovarian cancer cells in G2/M phase, while EGCG and SFN co-treatment causes
cell cycle arrest in both G2/M and S phases resulting in a significant escalation in
apoptosis. The combination also strongly inhibits hTERT (the main regulatory
subunit of telomerase) and Bcl-2 and alters the expression of phosphorylated
H2AX that promotes DNA damage response specifically in paclitaxel-resistant
ovarian cancer cell lines proposing the use of these compounds for overcoming
paclitaxel resistance in ovarian cancer treatment [135]. Furthermore, a synergistic
combination effect between sulforaphane and (-) epigallocatechin-3-gallate in
human colon carcinoma cells (HT-29 AP-1) has also been reported [136]. The
same group of investigators examined in prostate of Nrf2-knockout mice and
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 367

human prostate cancer cells the combination of EGCG and SFN with evidence of
regulatory cross talks between the transcription factors Nrf2 and AP-1 in prostate
tumors as several key Nrf2-dependent genes were found downregulated (three- to
35-fold) after in vivo administration of the combination of EGCG (100 mg/kg) and
SFN (45 mg/kg) [137]. In silico bioinformatics data revealed conserved transcription
factor binding site (TFBS) signatures in the promoter regions of Nrf2 and AP-1
suggesting the combination effect of SFN+EGCG is mediated via a concerted
modulation of Nrf2 and AP-1 pathways in the prostate [137].
Interestingly, SFN as a supportive partner with resveratrol and eugenol in induc-
ing apoptosis of tumor cells has also been reported [142, 143]. In glioma cells,
combination treatment with resveratrol inhibits cell proliferation and migration,
reduces cell viability, induces lactate dehydrogenase release, decreases
pro-survival Akt phosphorylation and increases caspase-3 activation [142]. This
points the worth of combining resveratrol and sulforaphane for the treatment of
glioma. However, more critical studies before translational implication of this study
are warranted. In human cervical cancer cells, concurrent sublethal doses of SFN and
eugenol act in a synergistic manner with gemcitabine culminating in loss of viable
cells by stimulation of apoptosis elicited by downregulation in the expression of
Bcl-2, COX-2, and IL-β [143].
Based on the notion that additional distinctive isothiocyanate constituents of
cruciferous vegetables such as indole-3-carbinol (I3C) or its condensation product
3,30 -diindolylmethane and SFN underscore any prognostic interactive influence,
their role in prostate and colon cancer models has been examined. In human colon
cancer cell lines, ITCs like sulforaphane (SFN) are cytotoxic, whereas indoles
including indole-3-carbinol and its condensation product 3,30 -diindolylmethane act
by cytostatic mechanisms. As a result, using defined combinations of SFN and
3,30 -diindolylmethane, the investigators interestingly found that at a total drug
concentration of 2.5 μM, all combinations of SFN and 3,30 -diindolylmethane were
antagonistic, but consecutively with increasing concentrations, the antagonistic
effect gradually turned into a synergistic interaction at the highest combined cyto-
toxic concentration of 40 μM [144]. Furthermore, SFN and DIM combination
(at 10 μM concentrations of each) results in strong G2/M cell cycle arrest, which
was not observed with either compound alone. Overall, the results suggest that at
low total concentrations (below 20 μM), which is physiologically more relevant, the
combined broccoli compounds display antagonistic interactions in terms of cell
growth inhibition. Building on their findings, further elucidation of mechanistic
interactions between bioactive food components is warranted for much better pre-
diction of beneficial health effects. With epidemiological findings weighing in favor
of high intake of cruciferous vegetables reducing the risk of prostate cancer
[145–147], the effect of the combination of glucosinolate hydrolysis products such
as I3C and SFN which are constituents of cruciferous vegetables seems logical and
therefore was examined mainly with reference to cell proliferation in a prostate
cancer cell line (PC-3) model. Cell proliferation in PC-3 prostate cancer cells was
significantly inhibited by I3C and sulforaphane at media concentrations of 0.2 mmol/
L and 0.02 mmol/L, respectively, which explicitly explains the observed protective
368 S. Banerjee and S.B. Paruthy

effect of cruciferous vegetables in relation to the risk of developing prostate


cancer [148].
At present time, there is no strong evidence for an inverse association between
consumption of cruciferous vegetables and pancreatic cancer risk, except a prospec-
tive study reporting cabbage consumption and statistically significant lower risk of
pancreatic cancer (one or more servings/week versus never consumption)
[149]. However, in support of the notion that combination of chemopreventive
bioactive compounds may afford protection against pancreatic cancer, few studies
have been conducted as presented herein. In related context, Hutzen et al. appraised
and reported bioactive isothiocyanates from cruciferous vegetables such as benzyl
isothiocyanate (BITC) and sulforaphane, both being capable of inhibiting cell
viability and inducing apoptosis in PANC-1 by mechanistically targeting inhibition
of tyrosine phosphorylation of signal transducer and activator of transcription factor-
3 (STAT-3) [150]. The STAT proteins comprise a family of latent transcription factors
with well-established roles in cell proliferation, growth, and survival and are consti-
tutively active in many human cancers; thus, inactivation of STAT-3 signaling has
emerged as an attractive target in cancer therapeutics. Interestingly, SFN has minimal
effect on the direct inhibition of STAT-3 tyrosine phosphorylation; however, combi-
nations of BITC and SFN inhibited cell viability and STAT-3 phosphorylation more
radically than either agent alone signifying that the combination of these bioactive
agents, BITC and sulforaphane, carries immense translational potential as a thera-
peutic and interventional agent. Moreover, overlapping with this report, in a 16-week
medium-term pancreatic carcinogenesis model in hamsters, the protective effects of
benzyl isothiocyanate and sulforaphane against initiation of pancreatic carcinogen-
esis have been acknowledged [151]. The multiplicity of combined pancreatic lesions
including atypical hyperplasias and adenocarcinomas was significantly decreased by
feeding a diet supplemented with 80 ppm BITC and 80 ppm SFN in the initiation but
not the post-initiation stage underpinning the fact that isothiocyanates BITC and SFN
together can block benzo(a)pyrene initiation of hamster pancreatic carcinogenesis.
Relating to pancreatic cancer therapy, another published study in literature docu-
ments that low doses of aspirin (ASP, 1 mM), curcumin (CUR,10 μM), and sulfo-
raphane (SFN, 5 μM) (ACS) combination statistically reduce cell viability and
concurrently induce apoptosis facilitated by activation of caspase-3 and poly
(ADP-ribose) polymerase (PARP) cleavage, considered as hallmark of apoptosis
[152]. Further mechanistic probing reveals that ACS inhibits NF-κB DNA binding
activity and promotes sustained activation of phosphor-extracellular signal-regulated
kinase 1/2 (p-ERK1/2), c-Jun, p38 MAPK signaling, and p53 proteins as probable
mechanism [152]. In addition, the feasibility of using solid lipid nanoparticulate
system to deliver this combination chemopreventive regimen has been investigated
for pancreatic cancer chemoprevention [153]. Referencing previously described
concept of cancer stem cells backed by in vitro data that SFN can eliminate
pancreatic CSC by inhibiting NF-κB activity alongside self-renewal capability
sensitizes the cells to undergo apoptosis [72, 109, 110]. An extension to this
observation, SFN, quercetin, and a natural mixture of a complete set of green tea
catechins (combination of green tea-derived catechins – epicatechin-2-gallate (ECG)
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 369

and catechin gallate (CG)) have been found to complement each other in the
elimination of advanced pancreatic cancer by miR-let-7 induction and K-ras inhibi-
tion [154]. Additionally, this novel combination of quercetin, sulforaphane, and GTC
also inhibits the self-renewal potential, apoptosis resistance, and migratory potential
of tumor cells; of significant interest is the fact that the combination of bioactive
agents is more effective than each single agent despite current therapeutics do not
adequately target the CSCs and novel therapeutic options are critically warranted.
Additional citation in the literature opinions sulforaphane synergizing with quercetin
to inhibit self-renewal capacity of pancreatic cancer stem cells by inhibiting stem cell
pluripotent transcription factor Nanog which could be a novel strategy to eliminate
cancer stem cell characteristics and destroy CSCs [155].

7 Sulforaphane and Clinical Trials

Given the exceptional inciting laboratory research conclusions projecting SFN as a


multifaceted bioactive natural compound that targets multiple processes associated
with tumor growth and survival and further outcome of experimental laboratory
studies in context of combined novel therapies, clinical trials are expected to be
initiated in the future to confirm therapeutic benefit of its combination with standard
chemotherapeutics for chemo- and radiosensitization of cancer. The toxicity and
effects of SFN in healthy men and patients with cancer have been recorded. In a
phase I clinical trial, SFN from broccoli sprouts has been evaluated for safety,
tolerance, and metabolism and conveyed a good safety profile [156]. In this protocol,
following a 5-day acclimatization period on a crucifer-free diet, broccoli sprout
extracts were administered orally at 8-h intervals for 7 days (21 doses). Most clinical
trials utilize broccoli sprouts or their hot water infusion as a rich and consistent
source of glucoraphanin because the young florets reportedly contain glucoraphanin
levels 20–50 times more than mature market-stage broccoli [13]. Current search for
sulforaphane and cancer at http://www.clinicaltrials.gov indicates a total of 13 trials
either ongoing, complete, or currently recruiting (Table 1). Phase II clinical trial for
treating patients with recurrent prostate cancer is currently being performed
(ClinicalTrials.gov, NCT01228084). Unfortunately, contextually relating to focus
of this chapter, none of the projected trials is currently inclined to sensitization
aspect of cancer therapy. However, the POUDER study objective which is currently
recruiting participants will provide data on the clinical feasibility and acceptability
of an supportive treatment option accompanying palliative chemotherapy
[NCT01879878]. This pilot study, POUDER trial, determines the feasibility of a
randomized controlled trial regarding the application of freeze-dried broccoli sprouts
rich in sulforaphane and quercetin in patients with advanced surgically
non-resectable pancreatic ductal adenocarcinoma (PDA) under palliative chemother-
apy. The patients in the experimental arm will receive 15 capsules containing a total
of 90 mg of active sulforaphane during the chemotherapy treatment course as a
nutrition supplement, whereas patients assigned to the placebo group will receive
inactive substance (methylcellulose) with identical capsule and potion distribution.
370 S. Banerjee and S.B. Paruthy

Table 1 Clinical trials with sulforaphane in cancer (http://www.clinicaltrials.gov)


Identifier
Official title # Study design Status
Pilot study evaluating broccoli sprouts in NCT Intervention Recruiting
advanced pancreatic cancer [POUDER trial] 01879878 participants
The effects of sulforaphane in patients with NCT Interventional Study completed
biochemical recurrence of prostate cancer 01228084
Chemoprevention of prostate cancer, HDAC NCT Interventional Not known
inhibition, and DNA methylation 01265953
Evaluating the effect of broccoli sprouts NCT Interventional Study completed
(sulforaphane) on cellular proliferation, an 00982319
intermediate marker of breast cancer risk
A human dietary intervention study to NCT Interventional Ongoing, but not
investigate the effect of sulforaphane on 01950143 recruiting
prostate cancer interception
Effect of topical application of sulforaphane NCT Observational Ongoing, but not
containing broccoli sprout extracts on 00894712 recruiting
radiation dermatitis during external beam participants
radiation therapy for breast cancer
Sulforaphane: a dietary histone deacetylase NCT Interventional Study completed
(HDAC) inhibitor in ductal carcinoma in situ 00843167
(DCIS)
Broccoli sprout extracts in healthy NCT Interventional Study completed
volunteers: a pilot study of Nrf2 pathway 02023931
modulation in oral mucosa
An intervention study to assess NCT Interventional Completed
bioavailability of sulforaphane delivered by 02300324
glucoraphanin-enriched broccoli soups in
healthy subjects
A pilot study evaluation of sulforaphane in NCT Interventional Ongoing, but not
atypical nevi-precursor lesions: assessment 01568996 recruiting
of STAT1 and STAT3 risk markers of participants
melanoma
Broccoli sprouts intervention in Qidong, PR, NCT Interventional Completed
China 01437501
Cruciferous vegetable intake and histone NCT Observational Recruiting
status in screening colonoscopy patients 01344330 participants
A randomized, double-blind, placebo- NCT Interventional Study completed
controlled, multiple ascending dose study to 02055716
evaluate the safety, tolerance,
pharmacokinetics, and pharmacodynamics
®
of Sulforadex in healthy male subjects
following daily dosing for 7 days

The outcome of this study will provide data on the clinical feasibility and accept-
ability of supportive treatment option accompanying palliative chemotherapy.
Importantly, from the outcome of the results, future clinical studies to create further
awareness for therapeutic benefit of sulforaphane in combination with chemothera-
peutic agents and radiotherapy in the field of oncology are anticipated.
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 371

8 Conclusions and Perspective

Based on the above overwhelming evidence supporting preclinical cognizance of


SFN as an adjuvant combination partner in a spectrum of organ specific antitumor
treatment modalities (including targeted therapy) currently in use, emerging per-
spective justifies practical clinical application of SFN on cancer patients in future.
However, more robust preclinical data using SFN or its analogs using available
genetically modified animal models are warranted. This will add value to combined
novel therapies and additionally prove an indisputable multipronged broad research-
based strategy toward stronger and efficacious therapeutic outcome in the field of
cancer treatment. Secondly, it is critical to explore and synthesize novel synthetic
analogs with improved bioavailability and extending their retention time in tumor
tissue with added benefit of capitalizing as innovative research strategy-based
prescription drug for prognostic anticancer treatment benefits.

References
1. Siegel RL, Miller KD, Jemal A (2016) Cancer statistics, 2016. CA Cancer J Clin 66:7–30
2. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674
3. Steinmetz KA, Potter JD (1991) Vegetables, fruit, and cancer. I. Epidemiology. Cancer Causes
Control 2:325–357
4. Potter JD, Steinmetz K (1996) Vegetables, fruit and phytoestrogens as preventive agents.
IARC Sci Publ 139:61–90
5. Liu RH (2003) Health benefits of fruit and vegetables are from additive and synergistic
combinations of phytochemicals. Am J Clin Nutr 78:517S–520S
6. Block G, Patterson B, Subar A (1992) Fruit, vegetables, and cancer prevention: a review of the
epidemiological evidence. Nutr Cancer 18:1–29
7. D’Incalci M, Steward WP, Gescher AJ (2005) Use of cancer chemopreventive phytochemicals
as antineoplastic agents. Lancet Oncol 6:899–904
8. Sarkar FH, Li Y (2006) Using chemopreventive agents to enhance the efficacy of cancer
therapy. Cancer Res 66:3347–3350
9. van Poppel G, Verhoeven DT, Verhagen H, Goldbohm RA (1999) Brassica vegetables and
cancer prevention. Epidemiology and mechanisms. Adv Exp Med Biol 472:159–168
10. Bosetti C, Filomeno M, Riso P, Polesel J, Levi F, Talamini R, Montella M, Negri E,
Franceschi S, La Vecchia C (2012) Cruciferous vegetables and cancer risk in a network of
case-control studies. Ann Oncol 23:2198–2203
11. IARC (2004) Cruciferous vegetables, isothiocyanates and indoles. International Agency for
Research on Cancer, Lyon
12. Navarro SL, Li F, Lampe JW (2011) Mechanisms of action of isothiocyanates in cancer
chemoprevention: an update. Food Funct 2:579–587
13. Fahey JW, Zhang Y, Talalay P (1997) Broccoli sprouts: an exceptionally rich source of
inducers of enzymes that protect against chemical carcinogens. Proc Natl Acad Sci U S A
94:10367–10372
14. Kushad MM, Brown AF, Kurilich AC, Juvik JA, Klein BP, Wallig MA, Jeffery EH (1999)
Variation of glucosinolates in vegetable crops of Brassica oleracea. J Agric Food Chem
47:1541–1548
15. Gasper AV, Al-Janobi A, Smith JA, Bacon JR, Fortun P, Atherton C, Taylor MA, Hawkey
CJ, Barrett DA, Mithen RF (2005) Glutathione S-transferase M1 polymorphism and
372 S. Banerjee and S.B. Paruthy

metabolism of sulforaphane from standard and high-glucosinolate broccoli. Am J Clin Nutr


82:1283–1291
16. Hu R, Khor TO, Shen G, Jeong WS, Hebbar V, Chen C, Xu C, Reddy B, Chada K, Kong AN
(2006) Cancer chemoprevention of intestinal polyposis in ApcMin/+ mice by sulforaphane, a
natural product derived from cruciferous vegetable. Carcinogenesis 27:2038–2046
17. Kassahun K, Davis M, Hu P, Martin B, Baillie T (1997) Biotransformation of the naturally
occurring isothiocyanate sulforaphane in the rat: identification of phase I metabolites and
glutathione conjugates. Chem Res Toxicol 10:1228–1233
18. Clarke JD, Dashwood RH, Ho E (2008) Multi-targeted prevention of cancer by sulforaphane.
Cancer Lett 269:291–304
19. Cornblatt BS, Ye L, Dinkova-Kostova AT, Erb M, Fahey JW, Singh NK, Chen MS, Stierer T,
Garrett-Mayer E, Argani P et al (2007) Preclinical and clinical evaluation of sulforaphane for
chemoprevention in the breast. Carcinogenesis 28:1485–1490
20. Zhang Y, Tang L (2007) Discovery and development of sulforaphane as a cancer chemopre-
ventive phytochemical. Acta Pharmacol Sin 28:1343–1354
21. Juge N, Mithen RF, Traka M (2007) Molecular basis for chemoprevention by sulforaphane: a
comprehensive review. Cell Mol Life Sci 64:1105–1127
22. Fimognari C, Hrelia P (2007) Sulforaphane as a promising molecule for fighting cancer. Mutat
Res 635:90–104
23. Awasthi YC, Jaiswal S, Sahu M, Sharma A (2014) Mechanisms of chemopreventive activity of
Sulforaphane. Springer, India
24. Fahey JW, Talalay P (1999) Antioxidant functions of sulforaphane: a potent inducer of Phase II
detoxication enzymes. Food Chem Toxicol 37:973–979
25. Myzak MC, Dashwood RH (2006) Chemoprotection by sulforaphane: keep one eye beyond
Keap1. Cancer Lett 233:208–218
26. Jakubikova J, Sedlak J, Mithen R, Bao Y (2005) Role of PI3K/Akt and MEK/ERK signaling
pathways in sulforaphane- and erucin-induced phase II enzymes and MRP2 transcription,
G2/M arrest and cell death in Caco-2 cells. Biochem Pharmacol 69:1543–1552
27. Tortorella SM, Royce SG, Licciardi PV, Karagiannis TC (2015) Dietary sulforaphane in cancer
chemoprevention: the role of epigenetic regulation and HDAC inhibition. Antioxid Redox
Signal 22:1382–1424
28. Rajendran P, Delage B, Dashwood WM, Yu TW, Wuth B, Williams DE, Ho E, Dashwood RH
(2011) Histone deacetylase turnover and recovery in sulforaphane-treated colon cancer cells:
competing actions of 14-3-3 and Pin1 in HDAC3/SMRT corepressor complex dissociation/
reassembly. Mol Cancer 10:68
29. Myzak MC, Karplus PA, Chung FL, Dashwood RH (2004) A novel mechanism of
chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res
64:5767–5774
30. Ambrosone CB, McCann SE, Freudenheim JL, Marshall JR, Zhang Y, Shields PG (2004)
Breast cancer risk in premenopausal women is inversely associated with consumption of
broccoli, a source of isothiocyanates, but is not modified by GST genotype. J Nutr
134:1134–1138
31. Fowke JH, Chung FL, Jin F, Qi D, Cai Q, Conaway C, Cheng JR, Shu XO, Gao YT, Zheng W
(2003) Urinary isothiocyanate levels, brassica, and human breast cancer. Cancer Res
63:3980–3986
32. Terry P, Wolk A, Persson I, Magnusson C (2001) Brassica vegetables and breast cancer risk.
JAMA 285:2975–2977
33. Pawlik A, Slominska-Wojewodzka M, Herman-Antosiewicz A (2016) Sensitization of estro-
gen receptor-positive breast cancer cell lines to 4-hydroxytamoxifen by isothiocyanates
present in cruciferous plants. Eur J Nutr 55:1165–1180
34. O’Brien NA, Browne BC, Chow L, Wang Y, Ginther C, Arboleda J, Duffy MJ, Crown J,
O’Donovan N, Slamon DJ (2010) Activated phosphoinositide 3-kinase/AKT signaling confers
resistance to trastuzumab but not lapatinib. Mol Cancer Ther 9:1489–1502
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 373

35. Pawlik A, Wiczk A, Kaczynska A, Antosiewicz J, Herman-Antosiewicz A (2013)


Sulforaphane inhibits growth of phenotypically different breast cancer cells. Eur J Nutr
52:1949–1958
36. Kaczynska A, Swierczynska J, Herman-Antosiewicz A (2015) Sensitization of HER2
positive breast cancer cells to Lapatinib using plants-derived isothiocyanates. Nutr Cancer
67:976–986
37. Kaczynska A, Herman-Antosiewicz A (2016). Combination of lapatinib with isothiocyanates
overcomes drug resistance and inhibits migration of HER2 positive breast cancer cells. Breast
Cancer
38. Lubecka-Pietruszewska K, Kaufman-Szymczyk A, Stefanska B, Cebula-Obrzut B,
Smolewski P, Fabianowska-Majewska K (2014) Clofarabine, a novel adenosine analogue,
reactivates DNA methylation-silenced tumour suppressor genes and inhibits cell growth in
breast cancer cells. Eur J Pharmacol 723:276–287
39. Hussain A, Mohsin J, Prabhu SA, Begum S, Nusri Qel A, Harish G, Javed E, Khan MA,
Sharma C (2013) Sulforaphane inhibits growth of human breast cancer cells and augments the
therapeutic index of the chemotherapeutic drug, gemcitabine. Asian Pac J Cancer Prev
14:5855–5860
40. Erzinger MM, Bovet C, Hecht KM, Senger S, Winiker P, Sobotzki N, Cristea S,
Beerenwinkel N, Shay JW, Marra G et al (2016) Sulforaphane preconditioning sensitizes
human colon cancer cells towards the bioreductive anticancer prodrug PR-104A. PLoS One
11:e0150219
41. Wang X, Doherty GP, Leith MK, Curphey TJ, Begleiter A (1999) Enhanced cytotoxicity of
mitomycin C in human tumour cells with inducers of DT-diaphorase. Br J Cancer
80:1223–1230
42. Rahmati-Yamchi M, Zarghami N, Nozad Charoudeh H, Ahmadi Y, Baradaran B, Khalaj-
Kondori M, Milani M, Akbarzadeh A, Shaker M, Pourhassan-Moghaddam M (2015)
Clofarabine has apoptotic effect on T47D breast cancer cell line via P53R2 gene expression.
Adv Pharm Bull 5:471–476
43. Lubecka-Pietruszewska K, Kaufman-Szymczyk A, Stefanska B, Cebula-Obrzut B,
Smolewski P, Fabianowska-Majewska K (2015) Sulforaphane alone and in combination
with clofarabine epigenetically regulates the expression of DNA methylation-silenced
tumour suppressor genes in human breast cancer cells. J Nutrigenet Nutrigenomics 8:
91–101
44. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner
JA, Mason W, Mariani L et al (2005) MGMT gene silencing and benefit from temozolomide in
glioblastoma. N Engl J Med 352:997–1003
45. Stedt H, Samaranayake H, Pikkarainen J, Maatta AM, Alasaarela L, Airenne K, Yla-Herttuala
S (2013) Improved therapeutic effect on malignant glioma with adenoviral suicide gene
therapy combined with temozolomide. Gene Ther 20:1165–1171
46. Zhang Z, Li C, Shang L, Zhang Y, Zou R, Zhan Y, Bi B (2016) Sulforaphane induces apoptosis
and inhibits invasion in U251MG glioblastoma cells. SpringerPlus 5:235
47. Karmakar S, Weinberg MS, Banik NL, Patel SJ, Ray SK (2006) Activation of multiple
molecular mechanisms for apoptosis in human malignant glioblastoma T98G and U87MG
cells treated with sulforaphane. Neuroscience 141:1265–1280
48. Li C, Zhou Y, Peng X, Du L, Tian H, Yang G, Niu J, Wu W (2014) Sulforaphane inhibits
invasion via activating ERK1/2 signaling in human glioblastoma U87MG and U373MG cells.
PLoS One 9:e90520
49. Lan F, Yang Y, Han J, Wu Q, Yu H, Yue X (2016) Sulforaphane reverses chemo-resistance to
temozolomide in glioblastoma cells by NF-kappaB-dependent pathway downregulating
MGMT expression. Int J Oncol 48:559–568
50. Lan F, Pan Q, Yu H, Yue X (2015) Sulforaphane enhances temozolomide-induced apoptosis
because of down-regulation of miR-21 via Wnt/beta-catenin signaling in glioblastoma.
J Neurochem 134:811–818
374 S. Banerjee and S.B. Paruthy

51. Shi L, Chen J, Yang J, Pan T, Zhang S, Wang Z (2010) MiR-21 protected human glioblastoma
U87MG cells from chemotherapeutic drug temozolomide induced apoptosis by decreasing
Bax/Bcl-2 ratio and caspase-3 activity. Brain Res 1352:255–264
52. Kotowski U, Heiduschka G, Brunner M, Czembirek C, Eder-Czembirek C, Schmidt R,
Fahim T, Thurnher D (2011) Radiosensitization of head and neck cancer cells by the phyto-
chemical agent sulforaphane. Strahlenther Onkol 187:575–580
53. Biel MA (2010) Photodynamic therapy of head and neck cancers. Methods Mol Biol
635:281–293
54. Lee SJ, Hwang HJ, Shin JI, Ahn JC, Chung PS (2015) Enhancement of cytotoxic effect on
human head and neck cancer cells by combination of photodynamic therapy and sulforaphane.
Gen Physiol Biophys 34:13–21
55. Laurie SA, Licitra L (2006) Systemic therapy in the palliative management of advanced
salivary gland cancers. J Clin Oncol 24:2673–2678
56. Chu WF, Wu DM, Liu W, Wu LJ, Li DZ, Xu DY, Wang XF (2009) Sulforaphane induces
G2-M arrest and apoptosis in high metastasis cell line of salivary gland adenoid cystic
carcinoma. Oral Oncol 45:998–1004
57. Wang XF, Wu DM, Li BX, Lu YJ, Yang BF (2009) Synergistic inhibitory effect of sulforaph-
ane and 5-fluorouracil in high and low metastasis cell lines of salivary gland adenoid cystic
carcinoma. Phytother Res 23:303–307
58. Chang CC, Hung CM, Yang YR, Lee MJ, Hsu YC (2013) Sulforaphane induced cell cycle
arrest in the G2/M phase via the blockade of cyclin B1/CDC2 in human ovarian cancer cells.
J Ovarian Res 6:41
59. Chuang LT, Moqattash ST, Gretz HF, Nezhat F, Rahaman J, Chiao JW (2007) Sulforaphane
induces growth arrest and apoptosis in human ovarian cancer cells. Acta Obstet Gynecol Scand
86:1263–1268
60. Bryant CS, Kumar S, Chamala S, Shah J, Pal J, Haider M, Seward S, Qazi AM, Morris R,
Semaan A et al (2010) Sulforaphane induces cell cycle arrest by protecting RB-E2F-1 complex
in epithelial ovarian cancer cells. Mol Cancer 9:47
61. Hunakova L, Gronesova P, Horvathova E, Chalupa I, Cholujova D, Duraj J, Sedlak J (2014)
Modulation of cisplatin sensitivity in human ovarian carcinoma A2780 and SKOV3 cell lines
by sulforaphane. Toxicol Lett 230:479–486
62. Sharma C, Sadrieh L, Priyani A, Ahmed M, Hassan AH, Hussain A (2011) Anti-carcinogenic
effects of sulforaphane in association with its apoptosis-inducing and anti-inflammatory
properties in human cervical cancer cells. Cancer Epidemiol 35:272–278
63. Wang X, Govind S, Sajankila SP, Mi L, Roy R, Chung FL (2011) Phenethyl isothiocyanate
sensitizes human cervical cancer cells to apoptosis induced by cisplatin. Mol Nutr Food Res
55:1572–1581
64. Yu D, Sekine-Suzuki E, Xue L, Fujimori A, Kubota N, Okayasu R (2009) Chemopreventive
agent sulforaphane enhances radiosensitivity in human tumor cells. Int J Cancer 125:1205–1211
65. Zak-Prelich M, Narbutt J, Sysa-Jedrzejowska A (2004) Environmental risk factors pre-
disposing to the development of basal cell carcinoma. Dermatol Surg 30:248–252
66. Knatko EV, Ibbotson SH, Zhang Y, Higgins M, Fahey JW, Talalay P, Dawe RS, Ferguson J,
Huang JT, Clarke R et al (2015) Nrf2 activation protects against solar-simulated ultraviolet
radiation in mice and humans. Cancer Prev Res 8:475–486
67. Berg D, Otley CC (2002) Skin cancer in organ transplant recipients: epidemiology, pathogen-
esis, and management. J Am Acad Dermatol 47:1–17, quiz 18-20
68. Benedict AL, Knatko EV, Dinkova-Kostova AT (2012) The indirect antioxidant sulforaph-
ane protects against thiopurine-mediated photooxidative stress. Carcinogenesis
33:2457–2466
69. Kalra S, Zhang Y, Knatko EV, Finlayson S, Yamamoto M, Dinkova-Kostova AT (2011) Oral
azathioprine leads to higher incorporation of 6-thioguanine in DNA of skin than liver: the
protective role of the Keap1/Nrf2/ARE pathway. Cancer Prev Res 4:1665–1674
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 375

70. Devesa SS, Blot WJ, Fraumeni JF Jr (1998) Changing patterns in the incidence of esophageal
and gastric carcinoma in the United States. Cancer 83:2049–2053
71. Qazi A, Pal J, Maitah M, Fulciniti M, Pelluru D, Nanjappa P, Lee S, Batchu RB, Prasad M,
Bryant CS et al (2010) Anticancer activity of a broccoli derivative, sulforaphane, in barrett
adenocarcinoma: potential use in chemoprevention and as adjuvant in chemotherapy. Transl
Oncol 3:389–399
72. Kallifatidis G, Labsch S, Rausch V, Mattern J, Gladkich J, Moldenhauer G, Buchler MW,
Salnikov AV, Herr I (2011) Sulforaphane increases drug-mediated cytotoxicity toward cancer
stem-like cells of pancreas and prostate. Mol Ther 19:188–195
73. Walther DJ, Peter JU, Bader M (2002) 7-Hydroxytryptophan, a novel, specific, cytotoxic agent
for carcinoids and other serotonin-producing tumors. Cancer 94:3135–3140
74. Cianchi F, Vinci MC, Supuran CT, Peruzzi B, De Giuli P, Fasolis G, Perigli G, Pastorekova S,
Papucci L, Pini A et al (2010) Selective inhibition of carbonic anhydrase IX decreases cell
proliferation and induces ceramide-mediated apoptosis in human cancer cells. J Pharmacol
Exp Ther 334:710–719
75. Potter C, Harris AL (2004) Hypoxia inducible carbonic anhydrase IX, marker of tumour
hypoxia, survival pathway and therapy target. Cell Cycle 3:164–167
76. Mastrangelo L, Cassidy A, Mulholland F, Wang W, Bao Y (2008) Serotonin receptors, novel
targets of sulforaphane identified by proteomic analysis in Caco-2 cells. Cancer Res
68:5487–5491
77. Mokhtari RB, Kumar S, Islam SS, Yazdanpanah M, Adeli K, Cutz E, Yeger H (2013)
Combination of carbonic anhydrase inhibitor, acetazolamide, and sulforaphane, reduces the
viability and growth of bronchial carcinoid cell lines. BMC Cancer 13:378
78. Chauhan D, Hideshima T, Anderson KC (2008) Targeting proteasomes as therapy in multiple
myeloma. Adv Exp Med Biol 615:251–260
79. Orlowski RZ, Kuhn DJ (2008) Proteasome inhibitors in cancer therapy: lessons from the first
decade. Clin Cancer Res 14:1649–1657
80. Oakervee HE, Popat R, Curry N, Smith P, Morris C, Drake M, Agrawal S, Stec J, Schenkein D,
Esseltine DL et al (2005) PAD combination therapy (PS-341/bortezomib, doxorubicin and
dexamethasone) for previously untreated patients with multiple myeloma. Br J Haematol
129:755–762
81. Jakubikova J, Cervi D, Ooi M, Kim K, Nahar S, Klippel S, Cholujova D, Leiba M, Daley JF,
Delmore J et al (2011) Anti-tumor activity and signaling events triggered by the iso-
thiocyanates, sulforaphane and phenethyl isothiocyanate, in multiple myeloma.
Haematologica 96:1170–1179
82. Chou TC (2006) Theoretical basis, experimental design, and computerized simulation of
synergism and antagonism in drug combination studies. Pharmacol Rev 58:621–681
83. He X, Yang K, Chen P, Liu B, Zhang Y, Wang F, Guo Z, Liu X, Lou J, Chen H (2014) Arsenic
trioxide-based therapy in relapsed/refractory multiple myeloma patients: a meta-analysis and
systematic review. OncoTargets Ther 7:1593–1599
84. Takahashi S (2010) Combination therapy with arsenic trioxide for hematological malignan-
cies. Anticancer Agents Med Chem 10:504–510
85. Rollig C, Illmer T (2009) The efficacy of arsenic trioxide for the treatment of relapsed and
refractory multiple myeloma: a systematic review. Cancer Treat Rev 35:425–430
86. Munshi NC, Tricot G, Desikan R, Badros A, Zangari M, Toor A, Morris C, Anaissie E,
Barlogie B (2002) Clinical activity of arsenic trioxide for the treatment of multiple myeloma.
Leukemia 16:1835–1837
87. Doudican NA, Wen SY, Mazumder A, Orlow SJ (2012) Sulforaphane synergistically enhances
the cytotoxicity of arsenic trioxide in multiple myeloma cells via stress-mediated pathways.
Oncol Rep 28:1851–1858
88. Greenstein S, Krett NL, Kurosawa Y, Ma C, Chauhan D, Hideshima T, Anderson KC, Rosen
ST (2003) Characterization of the MM.1 human multiple myeloma (MM) cell lines: a model
376 S. Banerjee and S.B. Paruthy

system to elucidate the characteristics, behavior, and signaling of steroid-sensitive and -resis-
tant MM cells. Exp Hematol 31:271–282
89. Fox E, Razzouk BI, Widemann BC, Xiao S, O’Brien M, Goodspeed W, Reaman GH, Blaney
SM, Murgo AJ, Balis FM et al (2008) Phase 1 trial and pharmacokinetic study of arsenic
trioxide in children and adolescents with refractory or relapsed acute leukemia, including acute
promyelocytic leukemia or lymphoma. Blood 111:566–573
90. Doudican NA, Bowling B, Orlow SJ (2010) Enhancement of arsenic trioxide cytotoxicity by
dietary isothiocyanates in human leukemic cells via a reactive oxygen species-dependent
mechanism. Leuk Res 34:229–234
91. Miller WH Jr, Schipper HM, Lee JS, Singer J, Waxman S (2002) Mechanisms of action of
arsenic trioxide. Cancer Res 62:3893–3903
92. Lunghi P, Tabilio A, Lo-Coco F, Pelicci PG, Bonati A (2005) Arsenic trioxide (ATO) and
MEK1 inhibition synergize to induce apoptosis in acute promyelocytic leukemia cells. Leu-
kemia 19:234–244
93. Deininger MW, Druker BJ (2003) Specific targeted therapy of chronic myelogenous leukemia
with imatinib. Pharmacol Rev 55:401–423
94. Lin LC, Yeh CT, Kuo CC, Lee CM, Yen GC, Wang LS, Wu CH, Yang WC, Wu AT (2012)
Sulforaphane potentiates the efficacy of imatinib against chronic leukemia cancer stem
cells through enhanced abrogation of Wnt/beta-catenin function. J Agric Food Chem
60:7031–7039
95. Capdevila J, Elez E, Peralta S, Macarulla T, Ramos FJ, Tabernero J (2008) Oxaliplatin-based
chemotherapy in the management of colorectal cancer. Expert Rev Anticancer Ther
8:1223–1236
96. Rixe O, Ortuzar W, Alvarez M, Parker R, Reed E, Paull K, Fojo T (1996) Oxaliplatin,
tetraplatin, cisplatin, and carboplatin: spectrum of activity in drug-resistant cell lines and in
the cell lines of the National Cancer Institute’s Anticancer Drug Screen panel. Biochem
Pharmacol 52:1855–1865
97. Kaminski BM, Weigert A, Brune B, Schumacher M, Wenzel U, Steinhilber D, Stein J, Ulrich S
(2011) Sulforaphane potentiates oxaliplatin-induced cell growth inhibition in colorectal cancer
cells via induction of different modes of cell death. Cancer Chemother Pharmacol
67:1167–1178
98. Guise CP, Abbattista MR, Singleton RS, Holford SD, Connolly J, Dachs GU, Fox SB,
Pollock R, Harvey J, Guilford P et al (2010) The bioreductive prodrug PR-104A is activated
under aerobic conditions by human aldo-keto reductase 1C3. Cancer Res 70:1573–1584
99. Singleton RS, Guise CP, Ferry DM, Pullen SM, Dorie MJ, Brown JM, Patterson AV, Wilson
WR (2009) DNA cross-links in human tumor cells exposed to the prodrug PR-104A:
relationships to hypoxia, bioreductive metabolism, and cytotoxicity. Cancer Res
69:3884–3891
100. Gu Y, Patterson AV, Atwell GJ, Chernikova SB, Brown JM, Thompson LH, Wilson WR
(2009) Roles of DNA repair and reductase activity in the cytotoxicity of the hypoxia-activated
dinitrobenzamide mustard PR-104A. Mol Cancer Ther 8:1714–1723
101. Jameson MB, Rischin D, Pegram M, Gutheil J, Patterson AV, Denny WA, Wilson WR
(2010) A phase I trial of PR-104, a nitrogen mustard prodrug activated by both hypoxia and
aldo-keto reductase 1C3, in patients with solid tumors. Cancer Chemother Pharmacol
65:791–801
102. Abou-Alfa GK, Chan SL, Lin CC, Chiorean EG, Holcombe RF, Mulcahy MF, Carter WD,
Patel K, Wilson WR, Melink TJ et al (2011) PR-104 plus sorafenib in patients with advanced
hepatocellular carcinoma. Cancer Chemother Pharmacol 68:539–545
103. McKeage MJ, Gu Y, Wilson WR, Hill A, Amies K, Melink TJ, Jameson MB (2011) A phase I
trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid
tumour patients. BMC Cancer 11:432
104. McKeage MJ, Jameson MB, Ramanathan RK, Rajendran J, Gu Y, Wilson WR, Melink TJ,
Tchekmedyian NS (2012) PR-104 a bioreductive pre-prodrug combined with gemcitabine
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 377

or docetaxel in a phase Ib study of patients with advanced solid tumours. BMC Cancer
12:496
105. Jamieson SM, Gu Y, Manesh DM, El-Hoss J, Jing D, Mackenzie KL, Guise CP,
Foehrenbacher A, Pullen SM, Benito J et al (2014) A novel fluorometric assay for aldo-keto
reductase 1C3 predicts metabolic activation of the nitrogen mustard prodrug PR-104A in
human leukaemia cells. Biochem Pharmacol 88:36–45
106. Roig AI, Eskiocak U, Hight SK, Kim SB, Delgado O, Souza RF, Spechler SJ, Wright WE,
Shay JW (2010) Immortalized epithelial cells derived from human colon biopsies express stem
cell markers and differentiate in vitro. Gastroenterology 138(1012–1021):e1011–e1015
107. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C
(2007) Distinct populations of cancer stem cells determine tumor growth and metastatic
activity in human pancreatic cancer. Cell Stem Cell 1:313–323
108. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM
(2007) Identification of pancreatic cancer stem cells. Cancer Res 67:1030–1037
109. Kallifatidis G, Rausch V, Baumann B, Apel A, Beckermann BM, Groth A, Mattern J, Li Z,
Kolb A, Moldenhauer G et al (2009) Sulforaphane targets pancreatic tumour-initiating cells by
NF-kappaB-induced antiapoptotic signalling. Gut 58:949–963
110. Rausch V, Liu L, Kallifatidis G, Baumann B, Mattern J, Gladkich J, Wirth T, Schemmer P,
Buchler MW, Zoller M et al (2010) Synergistic activity of sorafenib and sulforaphane
abolishes pancreatic cancer stem cell characteristics. Cancer Res 70:5004–5013
111. Li Y, Zhang T, Schwartz SJ, Sun D (2011) Sulforaphane potentiates the efficacy of
17-allylamino 17-demethoxygeldanamycin against pancreatic cancer through enhanced abro-
gation of Hsp90 chaperone function. Nutr Cancer 63:1151–1159
112. Kamal A, Boehm MF, Burrows FJ (2004) Therapeutic and diagnostic implications of Hsp90
activation. Trends Mol Med 10:283–290
113. Whitesell L, Lindquist SL (2005) HSP90 and the chaperoning of cancer. Nature reviews.
Cancer 5:761–772
114. Dai X, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi
G (2015) Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural
products as a potential therapeutic approach for cancer therapy. Exp Biol Med 240:760–773
115. Maksimovic-Ivanic D, Stosic-Grujicic S, Nicoletti F, Mijatovic S (2012) Resistance to TRAIL
and how to surmount it. Immunol Res 52:157–168
116. Wang F, Lin J, Xu R (2014) The molecular mechanisms of TRAIL resistance in cancer cells:
help in designing new drugs. Curr Pharm Des 20:6714–6722
117. Lim B, Allen JE, Prabhu VV, Talekar MK, Finnberg NK, El-Deiry WS (2015) Targeting
TRAIL in the treatment of cancer: new developments. Expert Opin Ther Targets
19:1171–1185
118. Trivedi R, Mishra DP (2015) Trailing TRAIL resistance: novel targets for TRAIL sensitization
in cancer cells. Front Oncol 5:69
119. Zhang L, Fang B (2005) Mechanisms of resistance to TRAIL-induced apoptosis in cancer.
Cancer Gene Ther 12:228–237
120. Matsui TA, Sowa Y, Yoshida T, Murata H, Horinaka M, Wakada M, Nakanishi R, Sakabe T,
Kubo T, Sakai T (2006) Sulforaphane enhances TRAIL-induced apoptosis through the induc-
tion of DR5 expression in human osteosarcoma cells. Carcinogenesis 27:1768–1777
121. Lowe SW, Ruley HE, Jacks T, Housman DE (1993) p53-dependent apoptosis modulates the
cytotoxicity of anticancer agents. Cell 74:957–967
122. Wang LH, Okaichi K, Ihara M, Okumura Y (1998) Sensitivity of anticancer drugs in Saos-2
cells transfected with mutant p53 varied with mutation point. Anticancer Res 18:321–325
123. Kandasamy K, Srivastava RK (2002) Role of the phosphatidylinositol 30 -kinase/PTEN/Akt
kinase pathway in tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis
in non-small cell lung cancer cells. Cancer Res 62:4929–4937
124. Jin CY, Moon DO, Lee JD, Heo MS, Choi YH, Lee CM, Park YM, Kim GY (2007)
Sulforaphane sensitizes tumor necrosis factor-related apoptosis-inducing ligand-mediated
378 S. Banerjee and S.B. Paruthy

apoptosis through downregulation of ERK and Akt in lung adenocarcinoma A549 cells.
Carcinogenesis 28:1058–1066
125. Sorensen PH, Lynch JC, Qualman SJ, Tirabosco R, Lim JF, Maurer HM, Bridge JA, Crist
WM, Triche TJ, Barr FG (2002) PAX3-FKHR and PAX7-FKHR gene fusions are prognostic
indicators in alveolar rhabdomyosarcoma: a report from the children’s oncology group. J Clin
Oncol 20:2672–2679
126. Bergantin E, Quarta C, Nanni C, Fanti S, Pession A, Cantelli-Forti G, Tonelli R, Hrelia P
(2014) Sulforaphane induces apoptosis in rhabdomyosarcoma and restores TRAIL-sensitivity
in the aggressive alveolar subtype leading to tumor elimination in mice. Cancer Biol Ther
15:1219–1225
127. Yamanaka T, Shiraki K, Sugimoto K, Ito T, Fujikawa K, Ito M, Takase K, Moriyama M,
Nakano T, Suzuki A (2000) Chemotherapeutic agents augment TRAIL-induced apoptosis in
human hepatocellular carcinoma cell lines. Hepatology 32:482–490
128. Shankar S, Srivastava RK (2004) Enhancement of therapeutic potential of TRAIL by cancer
chemotherapy and irradiation: mechanisms and clinical implications. Drug Resist Updat
7:139–156
129. Kim H, Kim EH, Eom YW, Kim WH, Kwon TK, Lee SJ, Choi KS (2006) Sulforaphane
sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-resistant hepatoma
cells to TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of
DR5. Cancer Res 66:1740–1750
130. Shankar S, Ganapathy S, Srivastava RK (2008) Sulforaphane enhances the therapeutic
potential of TRAIL in prostate cancer orthotopic model through regulation of apoptosis,
metastasis, and angiogenesis. Clin Cancer Res 14:6855–6866
131. Sharpe B, Beresford M, Bowen R, Mitchard J, Chalmers AD (2013) Searching for prostate
cancer stem cells: markers and methods. Stem Cell Rev 9:721–730
132. Labsch S, Liu L, Bauer N, Zhang Y, Aleksandrowicz E, Gladkich J, Schonsiegel F, Herr I
(2014) Sulforaphane and TRAIL induce a synergistic elimination of advanced prostate cancer
stem-like cells. Int J Oncol 44:1470–1480
133. Shen G, Khor TO, Hu R, Yu S, Nair S, Ho CT, Reddy BS, Huang MT, Newmark HL, Kong
AN (2007) Chemoprevention of familial adenomatous polyposis by natural dietary com-
pounds sulforaphane and dibenzoylmethane alone and in combination in ApcMin/+ mouse.
Cancer Res 67:9937–9944
134. Shen G, Xu C, Chen C, Hebbar V, Kong AN (2006) p53-independent G1 cell cycle arrest of
human colon carcinoma cells HT-29 by sulforaphane is associated with induction of p21CIP1
and inhibition of expression of cyclin D1. Cancer Chemother Pharmacol 57:317–327
135. Chen H, Landen CN, Li Y, Alvarez RD, Tollefsbol TO (2013) Epigallocatechin gallate and
sulforaphane combination treatment induce apoptosis in paclitaxel-resistant ovarian cancer
cells through hTERT and Bcl-2 down-regulation. Exp Cell Res 319:697–706
136. Nair S, Hebbar V, Shen G, Gopalakrishnan A, Khor TO, Yu S, Xu C, Kong AN (2008)
Synergistic effects of a combination of dietary factors sulforaphane and (-) epigallocatechin-3-
gallate in HT-29 AP-1 human colon carcinoma cells. Pharm Res 25:387–399
137. Nair S, Barve A, Khor TO, Shen GX, Lin W, Chan JY, Cai L, Kong AN (2010) Regulation of
Nrf2- and AP-1-mediated gene expression by epigallocatechin-3-gallate and sulforaphane in
prostate of Nrf2-knockout or C57BL/6J mice and PC-3 AP-1 human prostate cancer cells.
Acta Pharmacol Sin 31:1223–1240
138. Zhang M, Binns CW, Lee AH (2002) Tea consumption and ovarian cancer risk: a case-control
study in China. Cancer Epid Biomark Prev 11:713–718
139. Lee AH, Su D, Pasalich M, Binns CW (2013) Tea consumption reduces ovarian cancer risk.
Cancer Epidemiol 37:54–59
140. Hu J, Hu Y, Hu Y, Zheng S (2015) Intake of cruciferous vegetables is associated with reduced
risk of ovarian cancer: a meta-analysis. Asia Pac J Clin Nutr 24:101–109
141. Nagle CM, Olsen CM, Bain CJ, Whiteman DC, Green AC, Webb PM (2010) Tea consumption
and risk of ovarian cancer. Cancer Causes Control 21:1485–1491
12 Therapeutic Paradigm Underscoring Glucosinolate Sulforaphane in Chemo- and. . . 379

142. Jiang H, Shang X, Wu H, Huang G, Wang Y, Al-Holou S, Gautam SC, Chopp M (2010)
Combination treatment with resveratrol and sulforaphane induces apoptosis in human U251
glioma cells. Neurochem Res 35:152–161
143. Hussain A, Priyani A, Sadrieh L, Brahmbhatt K, Ahmed M, Sharma C (2012) Concurrent
sulforaphane and eugenol induces differential effects on human cervical cancer cells. Integr
Cancer Ther 11:154–165
144. Pappa G, Strathmann J, Lowinger M, Bartsch H, Gerhauser C (2007) Quantitative combina-
tion effects between sulforaphane and 3,30 -diindolylmethane on proliferation of human colon
cancer cells in vitro. Carcinogenesis 28:1471–1477
145. Kristal AR, Lampe JW (2002) Brassica vegetables and prostate cancer risk: a review of the
epidemiological evidence. Nutr Cancer 42:1–9
146. Liu B, Mao Q, Cao M, Xie L (2012) Cruciferous vegetables intake and risk of prostate cancer:
a meta-analysis. Int J Urol 19:134–141
147. Steinbrecher A, Nimptsch K, Husing A, Rohrmann S, Linseisen J (2009) Dietary glucosinolate
intake and risk of prostate cancer in the EPIC-Heidelberg cohort study. Int J Cancer
125:2179–2186
148. Frydoonfar HR, McGrath DR, Spigelman AD (2003) The effect of indole-3-carbinol and
sulforaphane on a prostate cancer cell line. ANZ J Surg 73:154–156
149. Larsson SC, Hakansson N, Naslund I, Bergkvist L, Wolk A (2006) Fruit and vegetable
consumption in relation to pancreatic cancer risk: a prospective study. Cancer Epid, Biomark
Prev 15:301–305
150. Hutzen B, Willis W, Jones S, Cen L, Deangelis S, Fuh B, Lin J (2009) Dietary agent, benzyl
isothiocyanate inhibits signal transducer and activator of transcription 3 phosphorylation and
collaborates with sulforaphane in the growth suppression of PANC-1 cancer cells. Cancer Cell
Int 9:24
151. Kuroiwa Y, Nishikawa A, Kitamura Y, Kanki K, Ishii Y, Umemura T, Hirose M (2006)
Protective effects of benzyl isothiocyanate and sulforaphane but not resveratrol against
initiation of pancreatic carcinogenesis in hamsters. Cancer Lett 241:275–280
152. Thakkar A, Sutaria D, Grandhi BK, Wang J, Prabhu S (2013) The molecular mechanism of
action of aspirin, curcumin and sulforaphane combinations in the chemoprevention of pan-
creatic cancer. Oncol Rep 29:1671–1677
153. Sutaria D, Grandhi BK, Thakkar A, Wang J, Prabhu S (2012) Chemoprevention of pancreatic
cancer using solid-lipid nanoparticulate delivery of a novel aspirin, curcumin and sulforaphane
drug combination regimen. Int J Oncol 41:2260–2268
154. Appari M, Babu KR, Kaczorowski A, Gross W, Herr I (2014) Sulforaphane, quercetin and
catechins complement each other in elimination of advanced pancreatic cancer by miR-let-7
induction and K-ras inhibition. Int J Oncol 45:1391–1400
155. Srivastava RK, Tang SN, Zhu W, Meeker D, Shankar S (2011) Sulforaphane synergizes with
quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Front Biosci
3:515–528
156. Shapiro TA, Fahey JW, Dinkova-Kostova AT, Holtzclaw WD, Stephenson KK, Wade KL,
Ye L, Talalay P (2006) Safety, tolerance, and metabolism of broccoli sprout glucosinolates and
isothiocyanates: a clinical phase I study. Nutr Cancer 55:53–62
Part III
Analytical and Processing Methods
Changing Trends in the Methodologies
of Extraction and Analysis of Hydrolytic 13
Products of Glucosinolates: A Review

Rohit Arora, Sakshi Bhushan, and Saroj Arora

Abstract
The increasing resistance among various pests and pathogens to the available
synthetic medicines had lead to the compulsion for exploring new and improved
alternatives. The phytochemicals have arisen as effective and safer alternate.
Among the many plant secondary metabolites, glucosinolates surpass in biolog-
ical activity and hence have been exhaustively extracted and explored for their
activity against these dreaded diseases. This augmented demand for exploring the
biological properties of all the available glucosinolates, especially their hydrolytic
products, has lead to the development of new methods for extraction and analysis
of these metabolites. The extraction methods are designed to match the volatility
of the compound without degrading its quality. These methods have been
improved to choose the best extraction conditions including the extracting sol-
vents. The extracted glucosinolate hydrolytic products are further analyzed using
an array of analytical methods ranging from as simple as paper chromatography
to as complex as microchip analysis. These methods are designed and developed
to match the needs of accuracy, reliability, and repeatability in addition to the cost
effectiveness. This review thus is a highlight and a milestone for the scientists and
budding researchers working on the crucial task of extracting, analyzing, and
exploring the biological properties of glucosinolate hydrolytic products.

Keywords
Glucosinolate hydrolytic products • Biosynthesis • Hydrodistillation • GC-MS •
HPLC • Microchip analysis

R. Arora (*) • S. Bhushan (*) • S. Arora (*)


Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar,
Punjab, India
e-mail: rtarora23@gmail.com; bhushan.sakshi8@gmail.com; sarojarora.gndu@gmail.com

# Springer International Publishing Switzerland 2017 383


J.-M. Mérillon, K.G. Ramawat (eds.), Glucosinolates, Reference Series in
Phytochemistry, DOI 10.1007/978-3-319-25462-3_13
384 R. Arora et al.

Abbreviations
AITC Allyl isothiocyanate
DCM Methylene chloride
ELISA Enzyme linked immunosorbent assay
FID Flame ionization detector
GC Gas chromatography
GHPs Glucosinolate hydrolytic products
GSLs Glucosinolates
HILIC Hydrophilic interaction liquid chromatography
HPLC High performance liquid chromatography
HS Head space
HSCCC High speed counter-current chromatography
ITCs Isothiocyanates
MS Mass spectrometry
NIRS Near infrared reflectance spectroscopy
NMR Nuclear magnetic resonance
PAPS 3-phosphoadenosine-50 -phosphosufate
PC Paper chromatography
S-GT: UDPG Thiohydroximateglucosyl transferase
SIXCPC Strong ion-exchange centrifugal partition chromatography
SIXCPE Strong ion-exchange centrifugal partition extraction
TLC Thin layer chromatography
UHPLC Ultra high performance liquid chromatography

Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385
2 Glucosinolates Biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 386
3 Bioactivities of Glucosinolates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387
3.1 Bactericidal/Antibacterial Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388
3.2 Antifungal Activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 389
3.3 Insecticidal Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 389
3.4 Antimutagenic Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390
3.5 Antiproliferative Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390
4 Glucosinolate Hydrolytic Products Extraction Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390
4.1 Cold Extraction Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 391
4.2 Hot Extraction Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 391
4.3 Other Extraction Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 393
5 Glucosinolate Hydrolytic Products Analysis Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 395
5.1 Chromatography Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 395
5.2 Non-Chromatographic Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 398
6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 401
7 Cross-References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 385

1 Introduction

Glucosinolates (GSLs) are class of plant secondary metabolites that contain


“sulfur” and are present in a number of plant families [1]. These plant
metabolites are associated with the presence of enzyme myrosinase also
known as thioglucosidase. Myrosinase belong to class of ß-thioglucosidases
present in myrosin cells also known as idioblasts that are scattered throughout
the tissue of GSL containing plant families. Both GSLs and myrosinase are
present together in the different parts of the plant, viz. root, shoot, stem,
seeds, etc. [2]. The plant injury results in the activation of
“GSL –MYROSINASE” system and results in the formation of “mustard oil
bomb.” Such response of plant tissue leads to the generation of unstable
thiohydroximate-O-sulfate moieties. The nonenzymatic removal of the sulfate
group followed by rearrangement of the left out core structure results in
variety of bioactive products also known as hydrolytic products. GSLs show
abundant structural diversity due to the presence of a vast variety of side chain
structures [3]. These structures are usually associated with protein amino acids
such as alanine, valine, leucine, isoleucine, phenylalanine, tyrosine, trypto-
phan, and methionine (Fig. 1). GSLs are mainly known due to the immense
biological properties of their hydrolytic products.
Glucosinolates are broken down into different hydrolytic products by the
action of myrosinase (Scheme 1). The hydrolytic products include
isothiocyanates (ITCs), nitriles, epithionitriles that depends upon the plant
species, side chain substitutions, pH, and ion concentration [4]. These hydro-
lytic products are responsible for various biological properties of this metabo-
lite. The breakdown products of GSL are not only responsible for the flavor and
aroma of plant families but also possess biological activities/properties includ-
ing action against different organisms (insect, fungi, and bacteria) along with
cancer chemoprevention [5]. Among the different hydrolytic products, ITCs are
known to possess anticarcinogenic properties in vitro as well as in vivo
systems.

Fig. 1 Structure of
glucosinolate
386 R. Arora et al.

Scheme 1 Formation of
glucosinolate hydrolytic
products

2 Glucosinolates Biosynthesis

Biosynthesis of glucosinolates has been widely studied for exploring the


anticarcinogenic properties of this compound. The biosynthesis of GSLs is classified
on the basis of precursor amino acid. GSL synthesis is three step process, firstly
amino acid chain elongation, secondly synthesis of glucon from the amino acid, and
lastly chain modification (glucon addition) [6].

1. Side chain elongation


GSLs are synthesized from chains elongated forms of phenylalanine, valine, and
methionine. The synthesis of GSLs is similar to the synthesis of leucine from
amino acid valine. The transamination of amino acid results in the production of
α-keto acid, followed by addition/condensation of acetyl CoA, and finally by
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 387

process of isomerization involved in the shifting of the hydroxyl group and


oxidation to ultimately attain elongated keto acid which is further used to obtain
amino acid.
2. Formation of glucone moiety
The first step in the formation of glucone moiety is the conversion of amino acid
oxime. Several studies have supported that various enzyme are involved in the
conversion of different amino acids that are responsible for biosynthesis of GSLs.
The above studies are basically carried out on the microsomal systems. The
enzymes involved in conversion of tyrosine and phenylalanine to their
corresponding oxime are the best example of such conversion involving GSL
synthesis. These enzymes are known to be responsible for the formation of
oximes that are further required for glycoside biosynthesis. The enzymatic system
involved in the chain elongation of methionine and phenylalanine is flavin
monooxygenase and that of tryptophan to indole acetaldoxime is peroxidase.
Above facts support the involvement of homologous enzyme system in such
conversions. The intermediate moieties between the oxime and thiohydroximate
have not been observed. It has been speculated that oxime is oxidized to nitro
compound which conjugates with cysteine and functions as a thiol source. The
above reaction is catalyzed by glutathione-S-transferase resulting in the formation
of S-alkylthiohydroximate. In the final step of glucone formation, the
thiohydroximate is S-glucosylated by S-GT (UDPG: thiohydroximateglucosyl
transferase) to form a desulfoglucosinolate. The above formed product is further
sulfated by 3-phosphoadenosine-50 -phosphosulfate (PAPS). Several studies have
shown the role of S-GT in sulfonation.
3. Side chain modifications
The side chain modifications of the elongated chains occur following glucone
formation. The aliphatic side chains that are attached to methionine are especially
modified. The pathway basically includes oxidation to methylsulfinylalkyl
followed by the removal of methyl sulfinyl group and desaturation. The above
step results in the formation of alkyl glucosinolates which on hydroxylation
results in hydroxyl alkenyl glucosinolates. The phenomenon responsible for
these modifications has been studied in case of 3-butenyl GSL that is catalyzed
by cytochrome P450 hydroxylase. The benzoyloxyalkyl are formed due to the
conjugation of hydroxyl alkyl GSL with benzoic acid. Interestingly, the side
chains of GSLs from branched amino acid and phenylalanine can be hydroxyl-
ated. In addition to this, tryptophan amino acid chains usually adds methoxy
group to result in its modification (Scheme 2).

3 Bioactivities of Glucosinolates

The formation of biologically important hydrolytic products such as isothiocyanates,


nitriles, epithionitriles, oxazolidine-2-thione, and thiocyanates along with other
products from glucosinolates makes this secondary metabolite an important choice
for preventing and curing diseases in plants and animals.
388 R. Arora et al.

Scheme 2 Process of biosynthesis of glucosinolates

3.1 Bactericidal/Antibacterial Activity

Glucosinolate hydrolytic products (GHPs) are known for their bactericidal activ-
ities against a wide range of bacterial strains. Among these, ITCs are effective
against Helicobactor pylori and its related strains [7]. Allyl ITC (AITC) is often
used as a preservative in industries. The aryl and alkyl GHPs have shown cytotoxic
effects against Salmonella typhimurium [8]. Benzyl ITC has been tested for
treatment against respiratory and urinary tracts [9]. The level of toxicity and
activity against pathogens varies with the type of strain. It has been shown that
GHPs present in Brassica napus have the potential to inhibit the growth of
Aphomyces euteiches along with propionibacterium [10]. Glucoraphanine contains
sulforaphane that has effective bactericidal action against H. pylori and related
strains along with strains resistant to antibiotics [11]. Hydrolytic products like
AITC, 4-hydroxybenzyl ITC, methyl ITC, 4-(Methylsulfinyl) butyl ITC, phenyl
ITC, and oxazolidienethiones showed bactericidal effects against different strains
of bacteria such as Bacillus cereus IFO-13494, Bacillus subtilis IFO-13722,
Salmonella enteritidis JCM-1891, Staphylococcus aureus IFO-12732, Escherichia
coli 0157:H7, Helicobacter pylori, and nitrifying bacteria [12–14]. Literature
survey reveals antibacterial activities of most of the GHPs, but their exact mech-
anism of action is not known. However, the action can be attributed to the tendency
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 389

of GHPs to inactivate various intracellular enzymes, thus resulting in oxidative


breakdown of sulfur bridges [15].

3.2 Antifungal Activities

The members of family Brassicaceae have the tendency to act against many fungi
that are phytopathogenic in nature [16]. Presence of ITCs such as AITC and
phenethyl ITC had resulted in antifungal activities of plant extracts belonging to
mustard family [17]. Many fungi such as Pernospora parasitica, Sclerotium rolfsii,
and Pythium ultimum were controlled using cabbage tissues due to the presence of
GHPs in them [18]. GHPs in addition to this were known to possess antifungal action
against Fusarium culmorum, Sclerotinia sclerotiorum, Pythium irregular, Rhizocto-
nia solani, and Diaporthe phaseolorum [19]. The fungi acting on wheat were also
shown to be inhibited by various GHPs [20]. It has also been observed that Brassica
napus c.v. inhibited fungal pathogens such as S. sclerotium [21]. GHPs have also
shown strong fungicidal activity against many fungal species such as Aspergillus
niger, Colletotrichum circinans, Peronospora parasitica, Fusarium graminearum,
Rhioctonia solani, Alternaria alternata, Penicillium glaucum, etc. [22]. The toxicity
and antifungal activity of GHPs depends on the chemical structure of ITCs [23]. The
mechanism of their action depends on their tendency to enhance and trigger the
defence mechanism of plants either by killing the pathogen or by providing protec-
tion by creating a plant barrier against respective fungi (pathogen) [24].

3.3 Insecticidal Activity

GHPs are bioactive agents responsible for their insecticidal properties against
different pests including nematodes [25]. The ability of these compounds to biode-
grade naturally makes it an excellent biofumigant [26]. Aromatic ITCs were found to
be effective insecticidal agents against Otiorhynchus sulcatus. GHPs have also been
reported to act against Naupactus leucoloma (weevil larvae) [27]. Inhibitory activity
of AITC, benzyl ITCs, methyl ITCs, phenyl ITCs, phenylethyl ITCs, and propenyl
ITC have been reported in a number of insects such as Cyclocephala, Musa
domestica, Rhyzopertha dominicia, Otiorhynchus sulcatus, Limonius influscatus,
Naupactus leucoloma, and Drosophila melanogaster [28–30].
Several reports have suggested nematode inhibitory effect of GHPs. It has been
observed that rotation of crops/plants belonging to Brassicaceae family resulted in
nematode control [31]. Various studies have demonstrated nematicidal activity
against Tylenchulus semipenetrans, Heterodera glycines, Meloidogyne chitwoodi,
M. incognita, M. javanica, and Globodera rostochiensis [32–35]. Plants belonging
to Brassicaceae family have also been known to possess herbicidal properties due to
the presence of various allelochemicals. Brown and Morra [36] reported the inhibi-
tion of growth of Lactuca sativa when grown along with B. napus. Such results have
suggested the ability of GHPs to play important role as a biofumigant [37].
390 R. Arora et al.

In addition to the above phenomenon, Brassica members have shown immense


potential to act as a green manure, thus limiting the use of artificial fertilizers
[38]. Vigna unguiculata when grown in the presence of Brassica showed reduction
in weeds [39]. Petersen et al. [40] have demonstrated the potential of various ITCs to
act against weeds. The Brassica species were able to suppress weeds such as
Echinochloa crus-galli (L.), Amaranthus hybridus L. (smooth pigweed), Sonchus
asper (L.) Hill (spiny sowthistle), Triticum aestivum L. (wheat), Echinochloa
crus-galli (L.) Beauv. (barnyard grass), Alopecurus myosuroides Huds. (black
grass), and Matricaria inodora L. (scentless mayweed). It has been proposed that
the mechanism of GHPs as a herbicide may be due to their interference and
inhibition in protein synthesis and other processes involved in plant growth [40].

3.4 Antimutagenic Activity

Literature survey has revealed that GHPs have strong antimutagenic effect. It was
observed that cauliflower extract acted as an effective antimutagenic agent against
quninoline induced mutagenicity [41]. It has been seen that broccoli extract admin-
istered to male BALB/C strain resulted in the reduction of mutagenicity thus
supporting antimutagenic effect of GHPs [42]. Similar results have also been
reported during studies involving metabolic activation involving S. typhimurium
TA 98. A study conducted by Murugun et al. [43] reported that broccoli extracts
prepared in ethanol resulted in suppression of mutagenicity caused by respective
mutagens on all the strains including TA 98, TA 102, and TA 1535.

3.5 Antiproliferative Activity

GHPs have also been studied for their potential to act against carcinogens and
mutagens. These phytochemicals have shown antitumor ability in tissues such as
liver, colon, pancreas, bladder, etc. [44]. Hydrolytic products of methyl sulfinyl GSL
have been observed to play important role in phase II enzymes induction [45]. Several
studies using animal models have supported anticarcinogenic effects of cruciferous
vegetables [46, 47]. Toxicity induced by administration of carcinogens in animals
has also been observed to decrease following the administration of GHPs [48–51].

4 Glucosinolate Hydrolytic Products Extraction Methods

The GHPs as discussed earlier have an array of biological properties ranging from
antioxidant, herbicidal, to anticancer activities. Their immense importance in phar-
maceutical industries plays a key role in identifying new and improved extraction
methods, which will not only increase their yield but their composition as well. The
two major types of extraction methods used for this purpose are cold extraction and
hot extraction.
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 391

4.1 Cold Extraction Methods

The cold extraction of GHPs is based on the solvent extraction of these important
metabolites from different plant sources, viz. root, shoot, leaves, seeds, etc. [52,
53]. In this method, the plant material is washed to remove any dust particle if
present. The material is then crushed and immediately added into solvent to prevent
any loss of metabolite due to evaporation. The solvent depends on the type of
metabolite required, since GHPs can be found in both polar as well as nonpolar
phase of solvents [54]. In general, the crushed plant material is first added into
hexane for defattening of the material. The time required for defattening depends on
the laboratory developed method, but usually 4–24 h incubation in solvent is
sufficient for this purpose. For ensuring the proper defattening process, after the
initial incubation, the solvent is replaced with fresh hexane and the same is kept for
24 h. Following incubation, both the solvents are combined, filtered through 0.22 μm
filter, and evaporated at 30  C under vacuum using rotary evaporator. The defattened
solvent often contains a number of GHPs and hence is also stored for further
analysis. After the initial defattening, other solvents relevant to the experiment in
hand are used for the extraction of other GHPs. But usually, methylene chloride
(DCM) is the most commonly used solvent for the extraction of GHPs. The
concentrated extracts are stored in a vial at – 40  C until further use [55].
In another extraction method by Fahey et al. [56], triple solvents, viz., dimethyl-
sulfoxide, dimethylformamide, and acetonitrile were used. The method involved the
homogenization of the plant material followed by the addition of equal volume of the
three solvents. The temperature of this mixture was kept at a low temperature of
–50  C to prevent the escaping of GHPs. The triple solvent may also be replaced
with dual solvents or more according to the experimental requirements. The GHPs
with the specific polarity will be absorbed in the respective solvent and can be
separated using separating funnel and dried under vacuum at 30  C to obtain
the GHPs.
In another set of experiments by Arora et al. [55], the plant material was
homogenized using distilled water and the extracting solvent was added to it. The
extraction solvents were varied in different methods (methylene chloride, diethyl
ether, and ethyl acetate). The GHPs were extracted thrice with the extraction solvents
and the solvents were pooled and passed through anhydrous sodium sulfate. The
extraction solvents were dried using three different drying conditions, viz. air drying,
nitrogen gas drying, and rotary evaporator drying. The experiment showed that
dichloromethane was the most effective extraction solvent and rotary evaporation
as the most efficient drying condition.

4.2 Hot Extraction Methods

The hot extraction methods in contrast to the earlier discussed method requires a
higher temperature for the extraction of GHPs from different plant materials. Since
the GHPs are volatile in nature, therefore, hot extraction methods are much suitable
392 R. Arora et al.

Fig. 2 Soxhlet apparatus, where (a) Soxhlet apparatus and (b) heating mantle

for the extraction of these valuable secondary metabolites. There are two basic
methods for hot extraction of GHPs. The first being Soxhlet extraction in which
the crushed plant material (seeds, leaves, roots, etc.) is added in the Soxhlet
apparatus (Fig. 2). The desired solvent such as methylene chloride or 80% methanol
is added in the round bottom flask. The flask is fitted with Soxhlet apparatus and is
heated at temperature ranging from 30  C to 50  C using heating mantle for a time
period of 3–10 h. The saturated solvent containing GHPs thus obtained is collected
and concentrated under vacuum using rotary evaporator [57, 58].
In the other hot extraction method known as hydrodistillation, only one solvent
is used, i.e., distilled water. The crushed plant material is added in a round bottom
flask and is mixed with distilled water. The flask is fitted with a Clevenger’s
apparatus and the mixture is heated using heating mantle until boiling (Fig. 3).
The temperature is reduced to 60  C to continue the boiling process for 2–3 h. The
condensed volatile GHPs are collected in the outer pipe. Following the completion
of the protocol, the extract and water is removed and the GHPs are obtained using
either methylene chloride or diethyl ether as solvent [55]. The significance of this
method for isolation of GHPs leads to the development of a number of modifica-
tions for improving this method for obtaining higher yield in less time as discussed
below.
Microwave assisted hydrodistillation is a modification of traditional hydrodis-
tillation in which the heating equipment was changed from heating mantle to a
microwave. As per the method given by Wei et al. [59], a microwave was modified to
keep the setup including a flat bottom flask and a Clevenger’s apparatus inside it
(Fig. 4). The microwave was operated at a power of 577 W and for a time period of
37.5 min. The water present in the extract was removed by passing the mixture
through anhydrous sodium sulfate. Although this modification provided no improve-
ment in the yield or composition of the extract, but a significant decrease in the
extraction time was observed.
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 393

Fig. 3 Clevenger’s apparatus


used in hydrodistillation
method

4.3 Other Extraction Methods

Apart from the above mentioned extraction methods involving the conventional hot
and cold extraction methods, a number of new alternatives have also been performed
to obtain the improved yield and composition. Among these, supercritical fluid
extraction is an advanced technique involving the use of supercritical fluids having
properties of both liquid as well as gas (Fig. 5). The most common supercritical fluid
used by the researchers is CO2, owing to its low cost, nontoxicity, odorless, and
colorless properties. An important aspect of using this fluid is that it can be modified
to gas or liquid as per extraction requirements. A comparative extraction of GHPs
using supercritical extraction by CO2 revealed an improvement in the composition
and amount [60].
The GHPs may also be extracted in an indirect way by an initial extraction of
intact GSLs. This method involves the pulverization of plant material followed by
the deactivation of myrosinase enzyme using liquid nitrogen or boiling water. The
intact GSLs thus obtained are extracted using different solvents, viz. methanol, ethyl
acetate, and methylene chloride. The intact GSLs thus obtained are desulfated in
394 R. Arora et al.

Fig. 4 Setup for microwave assisted hydrodistillation

Fig. 5 Equipment for


supercritical fluid extraction
13 Changing Trends in the Methodologies of Extraction and Analysis of. . . 395

Fig. 6 Indirect extraction of


glucosinolate hydrolytic
products using DEAE
sephadex column

DEAE sephadex column using an external supply of either artificial or natural


myrosinase enzyme (Fig. 6). An important advantage of this method is that the
type of GHPs required may be modified as per our requirement by changing the
column conditions (acidic, basic, or neutral pH) and temperature (low, room tem-
perature, or high) [61].

5 Glucosinolate Hydrolytic Products Analysis Methods

The GHPs extracted by the above mentioned methods can be utilized for a number of
biological activities such as antifungal, antibacterial, herbicidal, and anticancer
properties. Before utilizing it for any purpose, the analysis of its composition and
quantity of each compound is necessary to analyze the synergistic or antagonistic
effect of each compound. The analytical methods thus employed range from the
most common and traditional methods like paper and thin layer chromatography to
much advanced microchip analysis.

5.1 Chromatography Methods

The chromatographic methods involve the analysis of different components of the


extract on the basis of polarity and size.

5.1.1 Paper Chromatography


Paper chromatography (PC) is the oldest method during early 1900s for the analysis
of GHPs. The thiourea formed following the reaction amid ammonia and
isothiocyanates is detected and separated by paper. While the hydrophobic compo-
nents are analyzed using water saturated chloroform, the hydrophilic components on
the other hand involved the mixture of butanol, toluene, and water as mobile phase.
The low reproducibility of GHP analysis using this method makes it a difficult
choice for analysis [62].
396 R. Arora et al.

5.1.2 Thin Layer Chromatography


Thin layer chromatography (TLC) is much reliable and repeatable method for the
analysis of GHPs. The method uses silica or cellulose gel formed on glass plate as a
stationary phase, while different combination of solvents as per the polarity are used
as mobile phase. TLC can be used for both qualitative as well as quantitative analysis
of GHPs. The repeatability, reproducibility, and the ability to analyze a large number
of samples at the same time make this method highly useful for the initial analysis of
GHPs [63].

5.1.3 High Performance Liquid Chromatography


High performance liquid chromatography (HPLC) is the analytical technique mostly
preferred for the analysis of GHPs since late 1900s. The accurate results along with
an ease of reproducibility and repeatability makes it an idle analytical method till
date. The variation in the mobile phase, flow rate, and the column used helps in
analyzing almost all the combination of GHPs present in different extracts (Fig. 7).
The different compounds present in the extracts are analyzed using different detec-
tors such as photodiode array and the much important mass spectrometer
(MS) [64]. The MS attachment makes it easier for the identification of a compound
without the presence of any standard by detecting the molecular mass of compound.
New and improved versions of HPLC are regularly being formed having all the
advantages of the regular HPLC, while solving its limitations. Some of these
improved HPLC instruments are ultra high performance liquid chromatography
(UHPLC), hydrophilic interaction liquid chromatography (HILIC), and high speed
counter-current chromatography (HSCCC) [65, 66].

Fig. 7 High performance liquid chromatography equipment

You might also like