You are on page 1of 9

Biochemical Pharmacology 181 (2020) 114149

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

Preclinical studies of a novel snake venom-derived recombinant disintegrin T


with antitumor activity: A review

Axel H. Schönthala, Stephen D. Swensonb,c, Thomas C. Chenb, Francis S. Marklandc,
a
Department of Molecular Microbiology and Immunology, Keck School of Medicine (KSOM), University of Southern California (USC), Los Angeles, CA 90089, USA
b
Department of Neurological Surgery, KSOM, USC, Los Angeles, CA 90089, USA
c
Department of Biochemistry and Molecular Medicine, KSOM, USC, Los Angeles, CA 90089, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Snake venoms consist of a complex mixture of many bioactive molecules. Among them are disintegrins, which
Brachytherapy are peptides without enzymatic activity, but with high binding affinity for integrins, transmembrane receptors
Contortrostatin that function to connect cells with components of the extracellular matrix. Integrin-mediated cell attachment is
Disintegrins critical for cell migration and dissemination, as well as for signal transduction pathways involved in cell growth.
Glioblastoma
During tumor development, integrins play key roles by supporting cancer cell proliferation, angiogenesis, and
Integrins
metastasis. The recognition that snake venom disintegrins can block integrin functions has spawned a number of
Liposomes
Ovarian cancer studies to explore their cancer therapeutic potential. While dozens of different disintegrins have been isolated,
Prostate cancer none of them as yet has undergone clinical evaluation in cancer patients. Among the best-characterized and
Snake venom preclinically most advanced disintegrins is vicrostatin (VCN), a recombinant disintegrin that was rationally
Vicrostatin designed by fusing 62 N-terminal amino acids derived from the disintegrin contortrostatin with 6 C-terminal
amino acids from echistatin, the disintegrins from another snake species. Bacterially produced VCN was shown
to target multiple tumor-associated integrins, achieving potent anti-tumor and anti-angiogenic effects in in vitro
and in vivo models in the absence of noticeable toxicity. This review will introduce the field of snake venom
disintegrins as potential anticancer agents and illustrate the translational development and cancer-therapeutic
potential of VCN as an example.

1. Introduction activity of disintegrins results from interference with the final step of
platelet aggregation, where disintegrins bind to integrin receptors on
Snake venoms consist of a highly complex mixture of biologically platelets and as a result prevent fibrinogen binding. This potent antic-
active proteins and peptides that serve the immobilization and diges- oagulant activity of disintegrins generated medical interest toward their
tion of prey, as well as the snake’s self-defense and survival. After in- use as potent anticoagulants, and eventually resulted in the develop-
jection into prey, this mixture exerts effects on bleeding, pain and in- ment and marketing of two therapeutic agents, eptifibatide and tir-
flammation, and generates cytotoxic, neurotoxic, and cardiotoxic ofiban [5]. Eptifibatide (Integrilin) is a cyclic heptapeptide [6,7] that
sequelae. Many of these venom constituents harbor enzymatic activity, represents a synthetically modified version of barbourin, a disintegrin
whereas others do not. Examples of the most abundant components isolated from the venom of the pygmy rattlesnake (Sistrurus miliarius
include members of the family of phospholipase A2, metalloprotei- barbouri) [8] (Fig. 1). Tirofiban (Aggrastat) is a non-peptide peptido-
nases, serine proteinases, hydrolases, lyases, endonucleases, alpha- mimetic [7,9] that was designed based on the snake venom disintegrin
neurotoxins, dendrotoxins, cardiotoxins, protease inhibitors and disin- echistatin, isolated from the venom of the saw-scaled viper (Echis car-
tegrins [1–4]. inatus) [10]. Both eptifibatide and tirofiban are widely used in the clinic
Disintegrins represent a large group of low molecular weight, non- as antiplatelet agents during coronary interventions, including acute
enzymatic polypeptides that specifically bind to cell surface integrins coronary syndrome [11,12].
present on platelets and other cells. Their biological function serves to Nearly three decades have passed since eptifibatide and tirofiban
counter blood clotting, thereby promoting the further spread of the first demonstrated the compelling clinical benefit of snake venom-based
snake’s venom throughout the circulation of its prey. The anticoagulant disintegrins [5]. Since then, many more venom derived disintegrins


Corresponding author.
E-mail address: markland@usc.edu (F.S. Markland).

https://doi.org/10.1016/j.bcp.2020.114149
Received 5 May 2020; Received in revised form 9 July 2020; Accepted 9 July 2020
Available online 12 July 2020
0006-2952/ © 2020 Elsevier Inc. All rights reserved.
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

Fig. 1. Examples of venomous snakes used for ob-


taining disintegrins A. The dusky pygmy rattlesnake
(Sistrurus miliarius barbouri) is endemic throughout
the southeastern U.S. and is the most commonly
encountered venomous snake in urbanized areas. Its
venom contains the disintegrin barbourin, which is
notable for its KGD motif, rather than the more
common RGD motif in many other integrins. B. The
saw-scaled viper (Echis carinatus) can be found in
parts of Central Asia and the Middle East. Its venom
contains the disintegrin echistatin. Although its
venom is lethal in < 10 percent of untreated vic-
tims, this snake is believed to cause more human
deaths than all other snake species combined, pri-
marily due to its aggressive nature and the difficul-
ties of many patients to reach hospitals with avail-
able anti-venom. C. The common green pit (or
bamboo pit) viper (Trimeresurus gramineus) is en-
demic to the southern part of India. Its venom con-
tains the disintegrin trigramin. D. The southern
copperhead viper (Agkistrodon contortrix contortrix)
is one of five copperhead subspecies that are com-
monly found in the southern U.S. Its venom contains
the disintegrin contortrostatin. Copperheads bite
more people than any other U.S. species of snake.
Although the bite is extremely painful, the venom is
not very potent and almost never fatal.

have been identified and their potential use explored for a variety of functional differences have not yet been completely established. Such
medical needs beyond their role as platelet aggregation inhibitors. analysis is complicated by cell type-specific integrin expression profiles
Specifically, a number of disintegrins are thought to hold significant that are dependent on the microenvironment within a spaciotemporal
potential for cancer therapy in view of their ability to impede cancer context. Moreover, integrins are known to interact with other receptor
cell motility, invasion, metastasis and tumor angiogenesis. Although so proteins as part of focal adhesion multiprotein networks [26] and their
far no disintegrin-based drug has been produced for cancer therapeutic expression levels can vary significantly between normal cells and tumor
purposes, many preclinical studies have demonstrated the great pro- cells [27–29].
mise of this field and have provided the rationale for further explora- Integrins exhibit structural diversity and undergo conformational
tion of this fertile area of study [13,14]. In the following, we will changes that are central to the regulation of their function. They exist in
summarize the current status of snake venom-based disintegrin re- three major conformational states: an inactive or low-affinity state, a
search with regard to its objective of developing effective cancer ther- primed or activated high-affinity state, and a ligand-bound or occupied
apeutic agents. In particular, we will discuss native contortrostatin and state [30,31]. This distinction adds to their complexity and further in-
recombinant vicrostatin as exemplary representatives from among the creases the challenge to target this class of receptors for therapeutic
group of snake venom-based disintegrins. purposes. However, the discovery of the RGD-binding ability of in-
tegrins exposed a promising target that is sought to be exploited by
2. Integrins as disintegrin targets interventions that harbor the RGD motif as the matching counterpart.
Among the latter are the snake venom-derived RGD-containing disin-
Integrins are transmembrane receptors that enable the interaction of tegrins.
cells with the extracellular matrix (ECM). They function to attach the
cell cytoskeleton to specific components of the ECM, such as fi- 3. Snake venom disintegrins
bronectin, vitronectin, laminin, and collagen, and in reverse initiate
signal transduction into the cell interior upon ECM binding. Integrin- Disintegrins are a class of disulfide-rich, non-enzymatic peptides,
mediated cell attachment is critical for cell spreading and migration, originally isolated from snake venom. While many of them contain an
whereas integrin-mediated signal transduction pathways are involved RGD motif, a number of disintegrins have been identified that harbor
in cell cycle regulation, survival, and secretion of proteases. Not sur- variations of this core tripeptide domain, such as VGD, MGD, WGD, and
prisingly, integrins play key roles in the process of carcinogenesis by KGD (with preservation of glycine and aspartic acid), MLD and ECD
impacting cancer cell proliferation and survival, angiogenesis, and (with preservation of aspartic acid), and R/KTS [32–35]. As would be
metastasis [15–18]. expected, the specific composition of this domain (in concert with other
In their active conformation, integrins consist of heterodimeric elements of the disintegrin peptide) impacts their high-affinity/high-
complexes of one alpha- and one beta-subunit. In mammalian cells, 18 specificity interaction with select integrin targets, and thus creates a
alpha- and 8 beta-subunits with the ability to form 24 different het- diversity of interventional potential for different biological and patho-
erodimers have been recognized. Their ability to interact with the ECM logical processes [32,36].
or with membrane-associated proteins on other cells frequently de- Overall, RGD-containing disintegrins represent the largest and best-
pends on a conserved tripeptide motif consisting of arginine, glycine studied family of disintegrins. While most of them are monomeric, a
and aspartate, which is commonly referred to as the Arg-Gly-Asp (RGD) few act as dimers [37]. NMR studies and crystal structures of disin-
motif [19–21]. It was originally discovered within the amino acid se- tegrins established that their binding activity further depends on the
quence of fibronectin [22] and since then has been confirmed in other appropriate pairing of several cysteine residues responsible for the
ECM proteins as well [23,24]. Eight of the integrin dimers are capable disintegrin fold, as well as on a mobile 11-amino acid loop protruding
of recognizing the RGD motif in natural ligands (namely: αIIbβ3, αvβ1, from the polypeptide core, which displays the tripeptide RGD motif
αvβ3, αvβ5, αvβ6, αvβ8, α5β1, and α8β1) [24,25], but details of their [2,13,33,38]. In addition, binding is fine-tuned by residues flanking the

2
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

integrin-recognition peptide and supported further by C-terminal tail provided adequate material for further functional studies.
sequences, which are thought to trigger conformational changes in the Using native CN, it could be established that this disintegrin re-
bound integrin receptor, thereby completing the selective inhibition of cognizes αIIbβ3, α5β1, αvβ3, and αvβ5 integrins [42,45,46]. The
integrin activity [33,34,39]. Intriguingly, disintegrins preferentially physiological binding partners for these integrins consist of fibrinogen,
bind to the activated conformation of integrins, which is present on fibronectin, vitronectin, collagen, von Willebrand factor (vWF),
motile cells, such as cancer cells and angiogenic endothelial cells but thrombospondin 1 (TSP-1), and fibroblast growth factor 2 (FGF-2), and
not generally on quiescent cells, thus making them attractive vehicles to therefore blocking their activities by exposure to CN was expected to
specifically target cancerous tissues [28]. In all, members of the RGD- have a variety of complex effects [32]. Consequently, beyond antic-
containing family of disintegrins should be able to inhibit the physio- oagulation, CN exhibited inhibitory impact on tumor cell attachment to
logical functions of α5β1, α8β1, αvβ1, αvβ3, αvβ5, αvβ6, αvβ8 and matrix, cell migration and invasion, and angiogenesis, in both in vitro
αIIbβ3 integrins, the so-called RGD receptors [32]. and in vivo studies [41,47–50] (see additional details below).
Since the 1987 report on the isolation of the first disintegrin, tri- Recombinant technology approaches have been applied to many
gramin, from the venom of the common green pit viper (Trimeresurus other snake venom disintegrins and helped to advance this area of re-
gramineus) [40] (Fig. 1), about 100 additional snake venom disintegrins search (for a list of recombinant disintegrins, see [34]). Purification of
have been reported (see listing in ref. [32]). Many of them were char- native peptides from snake venom not only is laborious, but the initial
acterized structurally and functionally and were confirmed to interact milking of snakes is not without risks to the health of the worker.
with and inhibit specific integrins, generating further efforts to study Furthermore, yields are generally small and can represent mixtures of
their potential anticancer activities. In our own studies, the Markland different disintegrins, as some snake venoms may contain more than
lab discovered contortrostatin (CN) in 1994 [41,42] a homodimeric one type of disintegrin [4,33]. Cloning of disintegrins supports synth-
disintegrin from the venom of Agkistrodon contortrix contortrix, the esis of greater yields of high-quality protein product and enables de-
Southern copperhead snake (Fig. 1). CN has become one of the most- tailed studies of structure–function relationships, along with NMR
studied members of this group of snake venom disintegrins. Its devel- analysis and determination of crystal structures [2,34]. Another useful
opment toward potential therapeutic applications was further sup- approach is the production of chimeric peptides to create novel pro-
ported by its incorporation into a newly-designed, chimeric, re- ducts with potentially enhanced or altered biological activities [39,51].
combinant disintegrin, where it was genetically fused with the C- For example, domain swap experiments were performed [39] with two
terminal tail of the viperid disintegrin echistatin, resulting in the bac- snake venom disintegrins, echistatin from the venom of the saw-scaled
terially produced disintegrin vicrostatin (VCN) [43]. viper Echis carinatus [10] (Fig. 1) and eristostatin from the leaf-nosed
viper Eristocophis macmahoni [52]. While the former displayed high
binding activity to αvβ3 and α5β1 integrins, the latter exhibited much
3.1. Contortrostatin and vicrostatin lower binding to these integrins. Intriguingly, transfer of the C-terminal
HKGPAT amino acid sequence from echistatin to eristostatin resulted in
CN was purified from snake venom using a multi-step chromato- the latter’s greatly increased recognition and inhibition of both αvβ3
graphic approach with yields of 1–2 mg per gram of lyophilized crude and α5β1 integrins [39]. Such results not only demonstrated that in-
venom [42]. Its initial characterization in platelet aggregation inhibi- herent specificity and activity of disintegrins can be genetically ma-
tion assays showed an IC50 of 49 nM for human platelet-rich plasma, nipulated and altered, but they also contributed to the recognition that,
and binding assays revealed its binding to αIIbβ3 integrin on platelets, beside the central RGD motif, C-terminal tail epitopes represent critical
thereby preventing fibrinogen binding and exerting anticoagulant ac- components of disintegrin-integrin interaction. Because αvβ3 integrin
tivity. Molecular characterization presented its composition as a in particular has been recognized as being frequently overexpressed in
homodimer with molecular mass of 13.5 kDa and 6750 Da for the in- cancer cells and plays a prominent role in tumor metastasis [53,54],
dividual subunits [42,44]. A cDNA library was made from the venom specific targeting of this integrin may prove especially beneficial for
gland of A. contortrix contortrix, which enabled cloning of the full-length cancer therapeutic purposes.
cDNA containing the CN disintegrin domain of 195 base pairs encoding Similar to the above domain swap experiments, CN was purpose-
65 amino acids [45] (Fig. 2). Fusion of the active domain to thioredoxin fully genetically modified by replacing three amino acids at its C-
supported expression in a bacterial system and it was possible to con- terminal end with the six amino acids mentioned above (HKGPAT) from
firm that integrin binding activity was retained. Due to the limited the C-terminus of echistatin (Fig. 2). The resulting peptide represented
amounts of disintegrins that are usually obtained from their natural a sequence-engineered disintegrin of 69 amino acids and 7146 Da,
snake venom source, functional expression of a recombinant version

Fig. 2. Sequence alignment of contortrostatin (CN),


vicrostatin (VCN), and echistatin Native CN is a
dimer with two identical chains oriented in an anti-
parallel fashion (which is not represented by the
configuration shown in the Figure), held together by
two interchain disulfide bonds. Additional sec-
ondary structures are created by intrachain disulfide
bonds. VCN is a monomer that was created by re-
combinantly modifying the primary CN sequence by
swapping the C-terminal tail (3 aa underlined with
dotted line in CN) for 6 aa (underlined in blue font
and italics) derived from the C-terminus of echis-
tatin (underlined in blue font and italics). The ad-
ditional glycine at the N-terminus (underlined in
green font) is acquired during cleavage of the
thioredoxin-VCN hybrid protein with a specific
protease, tobacco etch virus (TEV) protease, during
the synthesis and purification process. In all se-
quences the RGD tripeptide motif is depicted in red
bold font.

3
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

which was called vicrostatin (VCN) [43,55]. It faithfully retained CN’s effects on tumor angiogenesis [49]. Combined with other studies that
targeting of αIIbβ3, α5β1, αvβ3, and αvβ5 integrins, but in addition have reported potent anti-angiogenic activity of CN and VCN
displayed greater than 10-fold higher affinity for α5β1 integrin, as was [48,55,66,67] (and of other disintegrins [68–71]), these results in-
expected based on the above described studies with echistatin and dicated two main features of CN and VCN: indirect inhibition of pri-
eristostatin [39]. In comparison to the homodimeric structure of CN, mary tumor growth through prevention of tumor angiogenesis, and
the novel chimeric disintegrin, VCN, was active as a monomer, thus direct blockage of metastasis via interruption of tumor cell migration
simplifying its production in larger quantities than those of several and invasion.
other disintegrins that have been cloned and expressed [34]. Using the Several subsequent studies validated the anti-breast cancer potency
Origami B Escherichia coli expression system, it was possible to reliably of CN and several other disintegrins and established their anti-meta-
produce 200 mg/L of purified VCN from the bacterial cell lysate. In static potential [45,72–74]. It was also demonstrated that inhibitory
contrast to the notoriously difficult-to-amass native disintegrins, VCN effects on adhesion and migration were specific to breast cancer cell
can be robustly produced in large quantity and without high-risk lines, but did not impact normal breast cell lines, such as MCF-10A
milking of snakes. Like CN, VCN was shown to inhibit tumor cell ad- [75]. The basis for this differential could be found in the overexpression
hesion, endothelial and tumor cell invasion, and angiogenesis of cancer-specific integrins, primarily integrin αvβ3, which represents
[43,55,56] (see additional details below). the prime target for CN and several other disintegrins to exert their
Given the complexity of the cancer microenvironment, in particular antimetastatic function [45,71–74].
in view of the propensity of cancer cells to change their integrin re- The above mentioned method of daily intratumor injection of CN,
pertoire in response to drug treatment, it would be advantageous to which was performed in a mouse breast cancer model to establish proof
target multiple integrins for optimized therapeutic impact [57]. In this of principle of in vivo activity [49], would not be practical for clinical
regard, the promiscuity of CN and VCN, i.e., their ability to target translation. Therefore, various modifications of disintegrin delivery are
multiple integrins simultaneously, is expected to provide a distinct being developed in order to support future clinical applications. In the
therapeutic benefit. A number of preclinical studies in various tumor context of CN development for breast cancer, a liposomal formulation
models have provided compelling evidence that these disintegrins in- (called LCN) was designed for intravenous delivery and shown to retain
deed harbor highly promising anticancer activity, and some of these active protein with full retention of biological activity [67]. Char-
studies will be presented below. acterizing a variety of parameters of twice-weekly LCN administration
in an orthotopic mouse breast cancer model, the authors emphasized
4. CN and VCN in breast cancer four advantages of this system: (i) prolonged circulatory half-life of LCN
over native CN, (ii) passive accumulation of LCN in tumor tissue, (iii)
Breast cancer is the leading type of cancer in women and accounted no platelet reactivity of LCN, and (iv) no immunogenicity of LCN; along
for 627,000 deaths worldwide in 2018 [58]. It is a molecularly het- with potent antiangiogenic effects and tumor growth inhibition [67]. In
erogeneous disease that is divided into different subtypes, based on all, these studies established evidence that proper formulation of CN
hormone receptor status, overexpression of human epidermal growth allowed delivery via a clinically acceptable delivery method.
factor receptor 2 (EGFR2/HER2/Neu), presence of mutations in breast In yet another approach, multimeric disintegrin protein polymer
cancer genes 1 or 2 (BRCA1/2), and an increasing number of ad- fusions targeting the tumor vasculature of murine breast cancer models
ditionally recognized molecular markers [59,60]. While early-stage were studied [56]. In these studies, the VCN sequence was genetically
disease is curable in the majority of patients, prognosis for advanced added to a cDNA encoding elastin-like polypeptide (A192) of 73.6 kDa,
breast cancer becomes worse, and morbidity and mortality from breast resulting in a fusion protein that spontaneously self-assembled into
cancer is largely associated with metastatic disease [61]. The man- higher order multimers, quite likely via intermolecular bonds between
agement of breast cancer depends on a variety of factors, including the many cysteine residues present on the VCN sequence (Fig. 2). Un-
subtype, stage, and the patient’s age. It generally involves surgery, like monomeric VCN, which is prone to efficient renal clearance due to
followed by radiation, hormone-blocking therapy in cases of hormone its small size of only 7.1 kDa, these high-molecular weight fusion
receptor-positive disease, and chemotherapy for advanced stages. De- complexes were above the renal filtration cutoff and persisted much
pending on the presence of overexpressed tyrosine kinase growth fac- longer in the circulation of mice, thereby allowing more time to accu-
tors, targeted antibodies (trastuzumab, cetuximab, etc.) or small-mo- mulate within the tumor tissue. The fusion protein retained the integrin
lecule inhibitors (lapatinib, olaparib, etc.) may be used. Newer targeting selectivity of its parental monomeric VCN molecule, as well as
experimental regimens are exploring the benefit of immune checkpoint its therapeutic activity when tested in an orthotopic mouse breast
inhibitory antibodies, antibody-drug conjugates, poly(ADP-ribose) cancer model in vivo [56]. As shown before with native CN and
polymerase (PARP) inhibitors, cyclin-dependent kinase 4/6 inhibitors, monomeric VCN, the anticancer effect of fused A192-VCN resulted from
inhibitors targeting the phosphoinositide 3-kinase (PI3K)/protein ki- a combination of inhibitory effects on tumor cells as well as on tumor-
nase B (AKT)/mammalian target of rapamycin (mTOR) pathway, and associated endothelial cells.
other modalities [62–64]. Yet, despite considerable and continuous
progress in this field, 42,000 women in the U.S. are expected to die 5. VCN in prostate cancer
from breast cancer in 2020 [65].
Several disintegrins, including CN and VCN, have been investigated Prostate cancer (PC) is the most prevalent and the second most le-
for their breast cancer therapeutic potential in various preclinical thal cancer among American men [76]. Duration of benefit from an-
models. A study from the Markland lab characterized the effects of CN drogen-deprivation therapy, the mainstay for treatment of metastatic
on adhesion, invasion, and proliferation of MDA-MB-435 human breast disease, is highly variable and difficult to predict. Metastatic PC in-
carcinoma cells in vitro, as well as on these cells’ growth and metastasis variably progresses to a castrate-resistant state in spite of androgen-
in an orthotopic nude mouse model in vivo [49]. It was reported that CN signaling networks remaining vitally active. Several potent agents tar-
blocked the attachment of cultured cells to ECM proteins fibronectin geting this network are associated with improved survival, including
and vitronectin and prevented cell invasion through an artificial Ma- androgen receptor antagonists such as enzalutamide, and agents tar-
trigel basement membrane, while at the same time it did not exert geting androgen synthesis via the CYP17 pathway, such as abiraterone
cytotoxic effects on these cells and did not inhibit proliferation in vitro. [77,78]. The addition of multimodal chemotherapy also has generated
In vivo, daily local injections of CN into the orthotopic tumor mass re- some limited benefit [79].
sulted in reduced tumor growth, along with greatly reduced numbers of Bone metastases are common in advanced PC and are an important
pulmonary macro-metastases and micro-metastases, and inhibitory cause of morbidity. Radium-223 is an alpha-emitting calcium mimetic

4
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

designed to decrease collateral damage to the bone marrow and can be anti-angiogenic potential of LVCN in vivo. Nude mice carrying xeno-
safely administered repeatedly [80]. Anti-angiogenic therapy is an al- grafted PC-3 cells were treated with LVCN or vehicle as described
ternative strategy for long-term control of PC progression, consistent above. After 5 weeks of treatment, tumor tissues were collected and
with the association between elevated tumor vascularity and poor subjected to immunostaining with anti-CD31/PECAM-1 antibodies, re-
prognosis [81]. Over time, however, all patients develop resistance to cognizing platelet endothelial cell adhesion molecule (PECAM-1) as a
these therapies [82,83]. marker of endothelial cells [97]. Computer-assisted quantitation of the
Although ionizing radiation has a primary anti-angiogenic effect in stained vessel areas was used to establish microvessel density (MVD) as
cancer, it also has a pro-angiogenic effect because of an effort by the a surrogate marker for tumor angiogenesis [98]. Using this method, it
tumor to protect its own vasculature from damage due to radiation could be established that treatment with LVCN resulted in a pro-
exposure [84]. Interestingly, it was reported that endothelial cell sen- nounced decrease in tumor associated MVD. Additional in vitro ex-
sitivity to external beam radiotherapy is enhanced by the simultaneous periments further confirmed VCN’s potent anti-angiogenic potency. For
administration of an integrin antagonist in a PC animal model [85]. As example, using human umbilical cord vascular endothelial cells
a consequence, advanced PC, irrespective of its hormonal dependence (HUVEC) in a tubulogenesis assay [99], VCN was shown to inhibit tu-
and chemotherapy resistance status, should be susceptible to anti-in- bule formation by these endothelial cells, along with pronounced dis-
tegrin therapy. This is true due to the fundamentally different mole- ruption of their actin cytoskeleton, at low nanomolar concentrations
cular mechanisms of action between anti-integrin therapy and the [55]. In all, these experiments conclusively established the anti-angio-
hormonal and chemotherapy agents presently utilized. genic potential of our recombinant disintegrin.
Activated integrins are not usually present in quiescent tissues, but
some, such as αvβ3 [86,87], play an important role in various neo- 6. VCN in ovarian cancer
plastic processes, which—in the case of prostate cancer—include bone
metastasis in particular [88–90]. The role of αvβ3 in enhancing skeletal Advanced Ovarian Cancer (OC) is a devastating and invariably fatal
invasion by prostate cancer cells underscores its potential as a target for disease. High-grade serous ovarian cancer (HGSOC) is the prevalent
therapeutic strategies aimed at inhibiting pathways involved in prostate subtype and accounts for 70–80% of OC deaths. The overall 5-year
cancer progression [91–93]. Bone metastases are present in ~70–80% survival in OC is only 30%. The standard treatment is aggressive sur-
of patients with prostate cancer. Preclinical studies provided convincing gery followed by adjuvant chemotherapy, but no current therapeutic
evidence that increased expression of αvβ3 leads to metastasis and bone solution can effectively prevent disease recurrence [100–102].
invasion [93]. This integrin mediates cell adhesion to the bone extra- Consistent molecular findings in HGSOC include the activation of
cellular matrix through recognition of conserved RGD motifs in various multiple components of the cellular invasion apparatus (i.e., invado-
ligands, including osteopontin, vitronectin, and fibronectin. It has also some-associated integrins and their downstream kinases [103]) and the
been reported that αvβ3 promotes binding to several extracellular presence of cancer stem cell-associated markers, which collectively
matrix proteins in the bone microenvironment that are needed both for correlate with invasiveness, chemoresistance, and progression
tumor cell colonization and tumor progression [93]. As well, metastasis [104–106]. The mode of OC dissemination is characteristic, but not
to remote sites, primarily to the bone, involves dissemination via the unique among solid tumors. Exfoliated OC cells from primary tumors
bloodstream, entailing adhesion of cancer cells within the vasculature, and already established peritoneal lesions assemble themselves into
which depends on activated integrins. Integrin αvβ3 has been shown to free-floating multicellular aggregates (cancer spheroids) which are
support PC cell attachment under blood flow conditions in an activa- further circulated by the peritoneal fluid to secondary sites of im-
tion-dependent manner [94–96], underscoring its important role in the plantation, where they attach, invade into the submesothelial con-
development of bone metastases and presenting it as a therapeutic nective tissue, and form new metastases [107]. The spheroid cells are
target in advanced prostate cancer. significantly more invasive and resistant to chemotherapy [108]. OC
In an effort to investigate the therapeutic potential of VCN in pre- micrometastases usually evade detection at the time of surgery, and
clinical prostate cancer models, the Markland lab applied a liposomal failure to eradicate them drives disease recurrence.
formulation of this disintegrin, called LVCN [55]. In general, liposomal Intravenously administered chemotherapy penetrates only poorly
encapsulation can provide a variety of advantages, including prolonged into the peritoneal compartment where OC is actively spreading. As a
half-life and enhanced drug delivery due to increased tumor entrap- result, significantly higher drug concentrations would have to be ad-
ment. Based on some favorable structural attributes inherent in disin- ministered, but would reach levels that exceed maximum tolerated
tegrin polypeptides, such as hydrophilicity and stability in solution at doses (MTDs) of chemotherapy. One solution to this problem consists of
low pH, in organic solvents, and at a range of temperatures, these the administration of chemotherapy directly into the intraperitoneal
molecules can be readily encapsulated with high efficiency, while re- (IP) space, and in advanced OC a 22% decrease in the risk of death was
taining full biological activity. In the case of VCN encapsulation to shown in patients who received combined IV/IP therapy versus IV
create LVCN, efficiency was 70% or greater [55]. therapy alone [109]. In 2006, the NCI issued the statement: “On the
LVCN was studied in two different mouse models of prostate cancer basis of the results of these randomized phase III clinical trials, a
[55]. In the first, human PC-3 prostate cancer cells were subcutaneously combination of IV and IP administration of chemotherapy conveys a
injected into the flank of nude mice. Different groups of mice were then significant survival benefit among women with optimally debulked
treated with LVCN, VCN, liposomes only, or with vehicle. LVCN exerted ovarian cancer, compared to IV administration alone” [110]. However,
profound inhibitory effects on tumor growth in this xenograft model. In when translating this recommendation into clinical practice over the
comparison, unencapsulated VCN displayed no quantifiable anti-tumor years that followed, the issue of tolerability became readily apparent
activity, which may in part be explained by its short circulatory half- [110,111]. Due to the altered peritoneal fluid dynamics following de-
life. In a second approach, it was investigated whether LVCN could treat bulking surgery, drug clearance from the peritoneal cavity becomes not
established prostate cancer bone metastases. In these studies, human only impeded but also more unpredictable than in normal subjects.
CWR22Rv1 prostate cancer cells were implanted into the tibia of mice, While it may result in prolonged exposure of residual tumors to much
followed by treatment as above. In this model as well, treatment of higher drug concentrations, it comes at the expense of additional off-
animals with LVCN, but not with VCN, resulted in strong inhibition of target toxicities [112,113]. Therefore, although efficacious, the taxane-
tumor growth. While these studies emphasized the antitumor activity of and platinum-based IP regimens are in general poorly tolerated, which
LVCN, they also showed that these treatments were very well tolerated is the main reason why a significant number of women opt out of IP
by the animals and did not cause obvious toxicities [55]. treatment despite its potential benefits [114]. Wright et al. [115] re-
The PC-3 prostate cancer mouse model was also used to evaluate the ported that although the combined IP/IV chemotherapy use increased

5
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

significantly at many National Comprehensive Cancer Network centers latest therapy to be approved is the alternating electric fields generator
between 2003 and 2012, < 50% of eligible patients completed the IP NovoTTF/Optune [130], which may extend median overall survival by
chemotherapy course. about 5 months [131]. Regardless of the treatment regimen, however,
The Markland lab studied potential benefits of VCN in preclinical GB patients continue to have dismal prognosis and novel treatments are
OC mouse models. For these studies, VCN was formulated using Oxiplex urgently needed.
as a delivery vehicle. Oxiplex is a hydrogel consisting of polyethylene In mouse models of GB, the utility of VCN as a brachytherapy agent
oxide and carboxymethyl cellulose stabilized by calcium chloride was investigated. It was shown that the sole tyrosine residue at position
[116]. In clinical applications, it is used as a barrier gel by placing it at 51 of the VCN peptide was amenable to radiolabeling with full reten-
sites of tissue injury post-operatively, where it serves as a temporary tion of disintegrin biological activity [132]. Consequently, the Mark-
mechanical barrier separating opposing tissue surfaces to reduce the land lab created 131I-VCN for evaluation of its potential use as a bra-
extent of postoperative adhesions [116,117]. Adhesion prevention is chytherapy agent. VCN was directly radiolabeled with radioiodine
particularly relevant after gynecological surgery, which suggested the (131I) using a modification of the chloramine T method [133] and
option of formulating an anti-adhesive gel with VCN incorporated confirmation by mass spectrometry. The ensuing product was shown to
uniformly throughout the Oxiplex gel for local delivery immediately retain its ability to inhibit platelet aggregation with identical activity to
following OC surgery. that of non-iodinated parental VCN [132]. Furthermore, fluorescence-
This idea was investigated by using nude mice implanted into their activated cell sorting (FACS) demonstrated avid binding of 131I-VCN to
peritoneum with multicellular spheroids of highly aggressive human glioblastoma cell lines overexpressing the expected target integrins, in
OC cells. The use of such spheroids, rather than individual cells, is particular αvβ3 and αvβ5 integrins. After intravenous delivery of 131I-
considered a physiologically more relevant model in the case of OC, in VCN to nude mice with or without human GB cells implanted into their
particular because these three-dimensional clusters, which are also brains, a significant portion of the injected material was found in the
present in patient-derived effusions [118], display increased radio- and brains of tumor-bearing mice, whereas radioactivity was below the
chemoresistance [119–121] and tend to harbor a more aggressive, in- detectable threshold in the brains of mice without tumors.
vasive phenotype [107]. Therefore, to investigate the impact of VCN in Therapeutic activity of 131I-VCN was investigated in different in vivo
a more clinically relevant mouse model, the tumor cells were grown as mouse GB models [132]. In one model, the brains of nude mice were
spheroids in vitro before injection into the peritoneum [122]. As treat- implanted with human U87 GB cells. Treatment with vehicle only, 131I
ment, the animals received VCN alone, VCN incorporated into Oxiplex only, or 131I-VCN, established that only 131I-VCN treatment resulted in
gel, or Oxiplex alone. VCN exerted pronounced anticancer activity in significant (p < 0.04) survival benefit, whereas 131I alone did not
this tumor model. Intriguingly, both formulations of VCN, either in extend survival over that observed with vehicle treatment alone. This
saline or mixed with Oxiplex, were highly effective, and quantitation of encouraging result was extended in a nude mouse model with in-
bioluminescent imaging showed greater than 95% inhibition of tumor tracranial human U251 GB cells. In this study, 131I-VCN confirmed its
spread. Upon dissection, control animals (without any VCN treatment) therapeutic impact (p < 0.003 over untreated), and extended median
revealed extensive and widespread macroscopic carcinomatosis survival even further when combined with TMZ, the chemotherapeutic
throughout the peritoneal cavity, while the VCN-treated groups showed standard of care for GB patients [134]. In an immunocompetent mouse
a significant decrease in the total number of macroscopic foci. In fact, model, mouse brains were implanted with syngeneic murine GL216
several of the VCN-treated animals were devoid of visible macroscopic glioma cells that had been transfected with a plasmid encoding the gene
tumor foci by gross examination. for O6-methylguanine DNA methyltransferase (MGMT). MGMT is a
In vivo studies investigating VCN’s therapeutic activity were com- DNA repair enzyme that specifically corrects the cytotoxic O6-methyl-
plemented by toxicity studies in mice and rats. Both single-dose and guanine lesions produced by the alkylating activity of TMZ. As a result,
multi-dose applications were performed, and toxicity was evaluated by MGMT-overexpressing cells are profoundly resistant to killing by TMZ,
a number of parameters, including signs of overt toxicity, weight loss, and this mechanism of resistance represents a well-recognized clinical
or changes in physical activity of animals, microscopic and histo- problem [135]. It was therefore of great interest to determine whether
131
pathological analysis of organs, hematological parameters, and blood I-VCN would be able to exert therapeutic activity in such a clinically
chemistry panels [123]. None of these various assays yielded signs of relevant resistance model. Mice harboring MGMT-overexpressing
toxicological effects of VCN, even at repeat dosing as high as 500 mg/ glioma cells were treated with 131I-VCN or with 131I-VCN in combina-
kg. Therefore, as consistently observed throughout other in vivo ex- tion with TMZ and were further compared to the clinical standard of
periments [43,55], VCN appeared to be very well tolerated, setting a care, i.e., radiation combined with TMZ. All three regimens yielded
promising stage for advancement to Phase I clinical trials. significant (p < 0.05) survival benefit over untreated control animals.
Intriguingly, the 131I-VCN plus TMZ combination performed the best
131
7. VCN in Glioblastoma: I-VCN and was slightly better than radiation combined with TMZ. In ag-
gregate, these in vivo studies established very encouraging proof-of-
Malignant glioma, particularly glioblastoma (GB, WHO grade IV principle that VCN can be successfully used as a novel delivery me-
astrocytic glioma), accounts for over half of all gliomas, which in turn chanism to target a radioactive payload to tumor tissue.
account for 80% of all malignant brain and CNS tumors [124]. The
great majority of patients with GB are faced with a dismal prognosis. 8. Conclusions
Advances in neurosurgery, radiation, and chemotherapy during the past
decades have provided only small improvements in clinical outcome, The discovery of snake venom disintegrins has spawned a number of
with 5-year survival remaining at about 10% [125,126]. The first-line studies to investigate their potential benefit for cancer therapeutic
treatment of glioblastoma is usually surgery, both to confirm the di- purposes. Among the best-characterized peptides is VCN, a single chain
agnosis and to remove as much of the tumor as possible. Concurrent peptide genetically engineered based on the structure of native CN with
temozolomide (TMZ) with radiotherapy, followed by adjuvant TMZ, several modifications, in particular the replacement of the three COOH-
has produced a median survival of about 15 months, and this regimen is terminal amino acids of CN with the six amino acid sequence at the
now the standard of care for GB, as well as for grade III anaplastic COOH-terminus of echistatin to optimize VCN’s anticancer activity. The
glioma [125,126]. An approved treatment for the recurrent setting is complexity of the cancer microenvironment dictates that for optimal
the angiogenesis inhibitor bevacizumab [127]. Although it improves efficacy an anti-integrin therapeutic must target at least two members
outcome for most GB patients, the duration of benefits is limited and of the RGD-binding integrin class and preferably more given the ability
survival after initial bevacizumab progression is poor [128,129]. The of cancer cells to change their integrin repertoire in response to drug

6
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

treatment [57]. VCN, which targets multiple integrin members in- (1999) 227–231.
cluding (but not limited to) αvβ3, αvβ5, α5β1 and αIIbβ3, admirably [10] Z.R. Gan, R.J. Gould, J.W. Jacobs, P.A. Friedman, M.A. Polokoff, Echistatin. A
potent platelet aggregation inhibitor from the venom of the viper, Echis carinatus,
meets this criterion. VCN has demonstrated robust cancer-therapeutic J. Biol. Chem. 263 (36) (1988) 19827–19832.
activity in a great variety of preclinical animal models, along with [11] X. Zhou, X. Wu, H. Sun, J. Li, Efficacy and safety of eptifibatide versus tirofiban in
undetectable toxicity, pointing to a fairly large therapeutic window. It acute coronary syndrome patients: a systematic review and meta-analysis, J. Evid.
Based Med. 10 (2) (2017) 136–144.
can be encapsulated in liposomes for enhanced tumor delivery, mixed [12] C.Y. Koh, R.M. Kini, From snake venom toxins to therapeutics–cardiovascular
with surgical anti-adhesive gels, or chemically modified with radio- examples, Toxicon 59 (4) (2012) 497–506.
isotopes for tumor-selective delivery of a toxic payload, all of which [13] Y.S. Chan, R.C.F. Cheung, L. Xia, J.H. Wong, T.B. Ng, W.Y. Chan, Snake venom
toxins: toxicity and medicinal applications, Appl. Microbiol. Biotechnol. 100 (14)
present additional options for translational development and inclusion (2016) 6165–6181.
in cancer therapeutic regimens. Among the key steps in the near future [14] H. Waheed, S.F. Moin, M.I. Choudhary, Snake venom: from deadly toxins to life-
is the submission of an investigational new drug (IND) application, to saving therapeutics, Curr. Med. Chem. 24 (17) (2017) 1874–1891.
[15] J. Cooper, F.G. Giancotti, Integrin signaling in cancer: mechanotransduction,
enable phase I safety studies in cancer patients. The successful clinical
stemness, epithelial plasticity, and therapeutic resistance, Cancer Cell 35 (3)
introduction and long-term market experience of two other disintegrin (2019) 347–367.
derived agents, the antiplatelet compounds eptifibatide and tirofiban, [16] H. Hamidi, J. Ivaska, Every step of the way: integrins in cancer progression and
may serve as encouraging examples in this regard. It therefore appears metastasis, Nat. Rev. Cancer 18 (9) (2018) 533–548.
[17] S. Raab-Westphal, J.F. Marshall, S.L. Goodman, Integrins as therapeutic targets:
promising for VCN, and potentially other disintegrins currently un- successes and cancers, Cancers (Basel) 9 (9) (2017) 110.
dergoing development, to advance to clinical testing in the near future. [18] G. Sokeland, U. Schumacher, The functional role of integrins during intra- and
extravasation within the metastatic cascade, Mol. Cancer 18 (1) (2019) 12.
[19] X. Lu, D. Lu, M.F. Scully, V.V. Kakkar, Integrins in drug targeting-RGD templates
CRediT authorship contribution statement in toxins, Curr. Pharm. Des. 12 (22) (2006) 2749–2769.
[20] D.A. Cheresh, Structural and biologic properties of integrin-mediated cell adhe-
Axel H. Schonthal: Conceptualization, Writing - original draft, sion, Clin. Lab. Med. 12 (2) (1992) 217–236.
[21] E. Ruoslahti, M.D. Pierschbacher, New perspectives in cell adhesion: RGD and
Writing - review & editing. Stephen D. Swenson: Data curation, integrins, Science 238 (4826) (1987) 491–497.
Investigation, Methodology. Thomas C. Chen: Funding acquisition. [22] M.D. Pierschbacher, E. Ruoslahti, Cell attachment activity of fibronectin can be
Francis S. Markland: Funding acquisition, Conceptualization, Project duplicated by small synthetic fragments of the molecule, Nature 309 (5963)
(1984) 30–33.
administration, Supervision, Writing - original draft, Writing - review & [23] Y.A. Kadry, D.A. Calderwood, Chapter 22: structural and signaling functions of
editing. integrins, Biochim. Biophys. Acta (BBA) – Biomembr. 1862 (5) (2020) 183206.
[24] M. Nieberler, U. Reuning, F. Reichart, J. Notni, H.J. Wester, M. Schwaiger,
M. Weinmuller, A. Rader, K. Steiger, H. Kessler, Exploring the role of RGD-re-
Declaration of Competing Interest
cognizing integrins in cancer, Cancers (Basel) 9 (9) (2017) 116.
[25] R.O. Hynes, Integrins: bidirectional, allosteric signaling machines, Cell 110 (6)
The authors declare that they have no known competing financial (2002) 673–687.
[26] B. Geiger, J.P. Spatz, A.D. Bershadsky, Environmental sensing through focal ad-
interests or personal relationships that could have appeared to influ- hesions, Nat. Rev. Mol. Cell Biol. 10 (1) (2009) 21–33.
ence the work reported in this paper. [27] S.M. Albelda, Role of integrins and other cell adhesion molecules in tumor pro-
gression and metastasis, Lab. Invest. 68 (1) (1993) 4–17.
[28] J.S. Desgrosellier, D.A. Cheresh, Integrins in cancer: biological implications and
Acknowledgements therapeutic opportunities, Nat. Rev. Cancer 10 (1) (2010) 9–22.
[29] M.M. Zutter, S.A. Santoro, W.D. Staatz, Y.L. Tsung, Re-expression of the alpha 2
We thank all current and former members of the Markland lab for beta 1 integrin abrogates the malignant phenotype of breast carcinoma cells, Proc.
Natl. Acad. Sci. U.S.A. 92 (16) (1995) 7411–7415.
their valuable contributions to disintegrin research. Funding to support [30] M. Bachmann, S. Kukkurainen, V.P. Hytonen, B. Wehrle-Haller, Cell adhesion by
research in the Markland lab has been provided by the National Cancer integrins, Physiol. Rev. 99 (4) (2019) 1655–1699.
Institute of the National Institutes of Health, USA, Grants R41 [31] W. Wang, B.H. Luo, Structural basis of integrin transmembrane activation, J. Cell.
Biochem. 109 (3) (2010) 447–452.
CA126001, R41 CA168228 and R41 CA165626, the Department of [32] J.K. Arruda Macedo, J.W. Fox, M. de Souza Castro, Disintegrins from snake ve-
Defense, US Army, USA, Grants PC030704 and OC060278, and the noms and their applications in cancer research and therapy, Curr. Protein Pept.
Southern California Clinical and Translational Science Institute at the Sci. 16 (6) (2015) 532–548.
[33] J.J. Calvete, C. Marcinkiewicz, D. Monleon, V. Esteve, B. Celda, P. Juarez, L. Sanz,
University of Southern California, USA. The funding sources had no
Snake venom disintegrins: evolution of structure and function, Toxicon 45 (8)
influence on the assembly and writing of this manuscript. Animal re- (2005) 1063–1074.
search performed in the Markland lab was in accordance with all ap- [34] V. David, B.B. Succar, J.A. de Moraes, R.F.G. Saldanha-Gama, C. Barja-Fidalgo,
plicable ethical standards and was reviewed and approved by the R.B. Zingali, Recombinant and chimeric disintegrins in preclinical research, Toxins
(Basel) 10 (8) (2018) 321.
Institutional Animal Care and Use Committee (IACUC) at the University [35] E.M. Walsh, C. Marcinkiewicz, Non-RGD-containing snake venom disintegrins,
of Southern California. functional and structural relations, Toxicon 58 (4) (2011) 355–362.
[36] J.J. Calvete, J.W. Fox, A. Agelan, S. Niewiarowski, C. Marcinkiewicz, The presence
of the WGD motif in CC8 heterodimeric disintegrin increases its inhibitory effect
References on alphaII(b)beta3, alpha(v)beta3, and alpha5beta1 integrins, Biochemistry 41 (6)
(2002) 2014–2021.
[1] R.J. McCleary, R.M. Kini, Non-enzymatic proteins from snake venoms: a gold mine [37] J.J. Calvete, M.P. Moreno-Murciano, R.D. Theakston, D.G. Kisiel,
of pharmacological tools and drug leads, Toxicon 62 (2013) 56–74. C. Marcinkiewicz, Snake venom disintegrins: novel dimeric disintegrins and
[2] A. Munawar, S.A. Ali, A. Akrem, C. Betzel, Snake venom peptides: tools of bio- structural diversification by disulphide bond engineering, Biochem. J. 372 (Pt 3)
discovery, Toxins (Basel) 10 (11) (2018) 474. (2003) 725–734.
[3] P.G. Ojeda, D. Ramirez, J. Alzate-Morales, J. Caballero, Q. Kaas, W. Gonzalez, [38] J.P. Xiong, T. Stehle, R. Zhang, A. Joachimiak, M. Frech, S.L. Goodman,
Computational studies of snake venom toxins, Toxins (Basel) 10 (1) (2017) 8. M.A. Arnaout, Crystal structure of the extracellular segment of integrin alpha
[4] T. Tasoulis, G.K. Isbister, A review and database of snake venom proteomes, Vbeta3 in complex with an Arg-Gly-Asp ligand, Science 296 (5565) (2002)
Toxins (Basel) 9 (9) (2017) 290. 151–155.
[5] J.J. Calvete, The continuing saga of snake venom disintegrins, Toxicon 62 (2013) [39] I. Wierzbicka-Patynowski, S. Niewiarowski, C. Marcinkiewicz, J.J. Calvete,
40–49. M.M. Marcinkiewicz, M.A. McLane, Structural requirements of echistatin for the
[6] K. Schror, A.A. Weber, Comparative pharmacology of GP IIb/IIIa antagonists, J. recognition of alpha(v)beta(3) and alpha(5)beta(1) integrins, J. Biol. Chem. 274
Thromb. Thrombolysis 15 (2) (2003) 71–80. (53) (1999) 37809–37814.
[7] M. Hashemzadeh, M. Furukawa, S. Goldsberry, M.R. Movahed, Chemical struc- [40] T.F. Huang, J.C. Holt, H. Lukasiewicz, S. Niewiarowski, Trigramin. A low mole-
tures and mode of action of intravenous glycoprotein IIb/IIIa receptor blockers: a cular weight peptide inhibiting fibrinogen interaction with platelet receptors ex-
review, Exp. Clin. Cardiol. 13 (4) (2008) 192–197. pressed on glycoprotein IIb-IIIa complex, J. Biol. Chem. 262 (33) (1987)
[8] R.M. Scarborough, J.W. Rose, M.A. Hsu, D.R. Phillips, V.A. Fried, A.M. Campbell, 16157–16163.
L. Nannizzi, I.F. Charo, Barbourin. A GPIIb-IIIa-specific integrin antagonist from [41] M. Trikha, Y.A. De Clerck, F.S. Markland, Contortrostatin, a snake venom disin-
the venom of Sistrurus m. barbouri, J. Biol. Chem. 266 (15) (1991) 9359–9362. tegrin, inhibits beta 1 integrin-mediated human metastatic melanoma cell adhe-
[9] E.J. Topol, T.V. Byzova, E.F. Plow, Platelet GPIIb-IIIa blockers, Lancet 353 (9148) sion and blocks experimental metastasis, Cancer Res. 54 (18) (1994) 4993–4998.

7
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

[42] M. Trikha, W.E. Rote, P.J. Manley, B.R. Lucchesi, F.S. Markland, Purification and from Macrovipera venom, inhibits integrin-mediated cell adhesion, migration and
characterization of platelet aggregation inhibitors from snake venoms, Thromb. angiogenesis, Lab. Invest. 85 (12) (2005) 1507–1516.
Res. 73 (1) (1994) 39–52. [71] O.H. Ramos, A. Kauskot, M.R. Cominetti, I. Bechyne, C.L. Salla Pontes,
[43] R. Minea, C. Helchowski, B. Rubino, K. Brodmann, S. Swenson, F. Markland Jr., F. Chareyre, J. Manent, R. Vassy, M. Giovannini, C. Legrand, H.S. Selistre-de-
Development of a chimeric recombinant disintegrin as a cost-effective anti-cancer Araujo, M. Crepin, A. Bonnefoy, A novel alpha(v)beta (3)-blocking disintegrin
agent with promising translational potential, Toxicon 59 (4) (2012) 472–486. containing the RGD motive, DisBa-01, inhibits bFGF-induced angiogenesis and
[44] B. Mercer, F. Markland, C. Minkin, Contortrostatin, a homodimeric snake venom melanoma metastasis, Clin. Exp. Metastasis 25 (1) (2008) 53–64.
disintegrin, is a potent inhibitor of osteoclast attachment, J. Bone Miner. Res. 13 [72] Z. Deng, J. Chai, Q. Zeng, B. Zhang, T. Ye, X. Chen, X. Xu, The anticancer prop-
(3) (1998) 409–414. erties and mechanism of action of tablysin-15, the RGD-containing disintegrin, in
[45] Q. Zhou, P. Hu, M.R. Ritter, S.D. Swenson, S. Argounova, A.L. Epstein, breast cancer cells, Int. J. Biol. Macromol. 129 (2019) 1155–1167.
F.S. Markland, Molecular cloning and functional expression of contortrostatin, a [73] R.S. Yang, C.H. Tang, W.J. Chuang, T.H. Huang, H.C. Peng, T.F. Huang, W.M. Fu,
homodimeric disintegrin from southern copperhead snake venom, Arch. Biochem. Inhibition of tumor formation by snake venom disintegrin, Toxicon 45 (5) (2005)
Biophys. 375 (2) (2000) 278–288. 661–669.
[46] Q. Zhou, M.T. Nakada, P.C. Brooks, S.D. Swenson, M.R. Ritter, S. Argounova, [74] S.E. Lucena, Y. Jia, J.G. Soto, J. Parral, E. Cantu, J. Brannon, K. Lardner,
C. Arnold, F.S. Markland, Contortrostatin, a homodimeric disintegrin, binds to C.J. Ramos, A.I. Seoane, E.E. Sanchez, Anti-invasive and anti-adhesive activities of
integrin alphavbeta5, Biochem. Biophys. Res. Commun. 267 (1) (2000) 350–355. a recombinant disintegrin, r-viridistatin 2, derived from the Prairie rattlesnake
[47] V. Golubkov, D. Hawes, F.S. Markland, Anti-angiogenic activity of contortrostatin, (Crotalus viridis viridis), Toxicon 60 (1) (2012) 31–39.
a disintegrin from Agkistrodon contortrix contortrix snake venom, Angiogenesis 6 [75] L. Montealegre-Sanchez, S.N.C. Gimenes, D.S. Lopes, S.C. Teixeira, L. Solano-
(3) (2003) 213–224. Redondo, V. de Melo Rodrigues, E. Jimenez-Charris, Antitumoral potential of
[48] Q. Zhou, M.T. Nakada, C. Arnold, K.Y. Shieh, F.S. Markland Jr., Contortrostatin, a lansbermin-i, a novel disintegrin from porthidium lansbergii lansbergii venom on
dimeric disintegrin from Agkistrodon contortrix contortrix, inhibits angiogenesis, breast cancer cells, Curr. Top. Med. Chem. 19 (22) (2019) 2069–2078.
Angiogenesis 3 (3) (1999) 259–269. [76] T.R. Rebbeck, G.P. Haas, Temporal trends and racial disparities in global prostate
[49] Q. Zhou, R.P. Sherwin, C. Parrish, V. Richters, S.G. Groshen, D. Tsao-Wei, cancer prevalence, Can. J. Urol. 21 (5) (2014) 7496–7506.
F.S. Markland, Contortrostatin, a dimeric disintegrin from Agkistrodon contortrix [77] K. Mori, N. Miura, H. Mostafaei, F. Quhal, R. Sari Motlagh, B. Pradere, S. Kimura,
contortrix, inhibits breast cancer progression, Breast Cancer Res. Treat. 61 (3) T. Kimura, S. Egawa, A. Briganti, P.I. Karakiewicz, S.F. Shariat, Sequential therapy
(2000) 249–260. of abiraterone and enzalutamide in castration-resistant prostate cancer: a sys-
[50] S. Swenson, S. Ramu, F.S. Markland, Anti-angiogenesis and RGD-containing snake tematic review and meta-analysis, Prostate Cancer Prostatic Dis. (2020) March 9
venom disintegrins, Curr. Pharm. Des. 13 (28) (2007) 2860–2871. [online ahead of print].
[51] G. Mezo, F. Hudecz, Synthesis of linear, branched, and cyclic peptide chimera, [78] X. Zheng, X. Zhao, H. Xu, X. Han, H. Xu, X. Dong, R. Peng, L. Yang, Q. Wei, J. Ai,
Methods Mol. Biol. 298 (2005) 63–76. Efficacy and safety of abiraterone and enzalutamide for castration-resistant pros-
[52] M.A. McLane, M.A. Kuchar, C. Brando, D. Santoli, C.A. Paquette-Straub, tate cancer: a systematic review and meta-analysis of randomized controlled trials,
M.E. Miele, New insights on disintegrin-receptor interactions: eristostatin and Medicine 98 (44) (2019) e17748.
melanoma cells, Haemostasis 31 (3–6) (2001) 177–182. [79] M.J. Ferris, Y. Liu, J. Ao, J. Zhong, M. Abugideiri, T.W. Gillespie, B.C. Carthon,
[53] J.A. Eble, J. Haier, Integrins in cancer treatment, Curr. Cancer Drug Targets 6 (2) M.A. Bilen, O. Kucuk, A.B. Jani, The addition of chemotherapy in the definitive
(2006) 89–105. management of high risk prostate cancer, Urol. Oncol. 36 (11) (2018) 475–487.
[54] S. Katsamakas, T. Chatzisideri, S. Thysiadis, V. Sarli, RGD-mediated delivery of [80] M.C. Cursano, M. Iuliani, C. Casadei, M. Stellato, G. Tonini, G. Paganelli,
small-molecule drugs, Future Med. Chem. 9 (6) (2017) 579–604. D. Santini, U. De Giorgi, Combination radium-223 therapies in patients with bone
[55] R.O. Minea, C.M. Helchowski, S.J. Zidovetzki, F.K. Costa, S.D. Swenson, metastases from castration-resistant prostate cancer: a review, Crit. Rev. Oncol.
F.S. Markland Jr., Vicrostatin - an anti-invasive multi-integrin targeting chimeric Hematol. 146 (2020) 102864.
disintegrin with tumor anti-angiogenic and pro-apoptotic activities, PLoS ONE 5 [81] Z. Melegh, S. Oltean, Targeting angiogenesis in prostate cancer, Int. J. Mol. Sci. 20
(6) (2010) e10929. (11) (2019) 2676.
[56] S.M. Janib, J.A. Gustafson, R.O. Minea, S.D. Swenson, S. Liu, M.K. Pastuszka, [82] M. Carceles-Cordon, W.K. Kelly, L. Gomella, K.E. Knudsen, V. Rodriguez-Bravo,
L.L. Lock, H. Cui, F.S. Markland, P.S. Conti, Z. Li, J.A. MacKay, Multimeric dis- J. Domingo-Domenech, Cellular rewiring in lethal prostate cancer: the architect of
integrin protein polymer fusions that target tumor vasculature, drug resistance, Nat. Rev. Urol. 17 (5) (2020) 292–307.
Biomacromolecules 15 (7) (2014) 2347–2358. [83] A.K. Miyahira, R.B. Den, M.I. Carlo, R. de Leeuw, T.A. Hope, F. Karzai,
[57] H.M. Sheldrake, L.H. Patterson, Strategies to inhibit tumor associated integrin R.R. McKay, S.S. Salami, J.W. Simons, K.J. Pienta, H.R. Soule, Tumor cell het-
receptors: rationale for dual and multi-antagonists, J. Med. Chem. 57 (2014) erogeneity and resistance; report from the 2018 Coffey-Holden Prostate Cancer
6301–6315. Academy Meeting, Prostate 79 (3) (2019) 244–258.
[58] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global [84] P. Grabham, P. Sharma, The effects of radiation on angiogenesis, Vasc. Cell 5 (1)
cancer statistics 2018: GLOBOCAN estimates of incidence and mortality world- (2013) 19.
wide for 36 cancers in 185 countries, CA Cancer J. Clin. 68 (6) (2018) 394–424. [85] A. Abdollahi, D.W. Griggs, H. Zieher, A. Roth, K.E. Lipson, R. Saffrich, H.J. Grone,
[59] K. Barzaman, J. Karami, Z. Zarei, A. Hosseinzadeh, M.H. Kazemi, S. Moradi- D.E. Hallahan, R.A. Reisfeld, J. Debus, A.G. Niethammer, P.E. Huber, Inhibition of
Kalbolandi, E. Safari, L. Farahmand, Breast cancer: biology, biomarkers, and alpha(v)beta3 integrin survival signaling enhances antiangiogenic and antitumor
treatments, Int. Immunopharmacol. 84 (2020) 106535. effects of radiotherapy, Clin. Cancer Res. 11 (17) (2005) 6270–6279.
[60] U. Testa, G. Castelli, E. Pelosi, Breast cancer: a molecularly heterogenous disease [86] N.P. McCabe, S. De, A. Vasanji, J. Brainard, T.V. Byzova, Prostate cancer specific
needing subtype-specific treatments, Med Sci (Basel) 8 (1) (2020) 18. integrin alphavbeta3 modulates bone metastatic growth and tissue remodeling,
[61] S. Al-Mahmood, J. Sapiezynski, O.B. Garbuzenko, T. Minko, Metastatic and triple- Oncogene 26 (42) (2007) 6238–6243.
negative breast cancer: challenges and treatment options, Drug Deliv. Transl. Res. [87] S. Stucci, M. Tucci, A. Passarelli, F. Silvestris, Avbeta3 integrin: pathogenetic role
8 (5) (2018) 1483–1507. in osteotropic tumors, Crit. Rev. Oncol. Hematol. 96 (1) (2015) 183–193.
[62] J.J. Chan, T.J.Y. Tan, R.A. Dent, Novel therapeutic avenues in triple-negative [88] R. San Martin, R. Pathak, A. Jain, S.Y. Jung, S.G. Hilsenbeck, M.C. Pina-Barba,
breast cancer: PI3K/AKT inhibition, androgen receptor blockade, and beyond, A.G. Sikora, K.J. Pienta, D.R. Rowley, Tenascin-C and integrin alpha9 mediate
Ther. Adv. Med. Oncol. 11 (2019) 1758835919880429. interactions of prostate cancer with the bone microenvironment, Cancer Res. 77
[63] W. Janni, A. Schneeweiss, V. Muller, A. Wockel, M.P. Lux, A.D. Hartkopf, (21) (2017) 5977–5988.
N. Nabieva, F.A. Taran, H. Tesch, F. Overkamp, D. Luftner, E. Belleville, F. Schutz, [89] Z. Zhao, E. Li, L. Luo, S. Zhao, L. Liu, J. Wang, R. Kang, J. Luo, A PSCA/PGRN-NF-
P.A. Fasching, T.N. Fehm, H.C. Kolberg, J. Ettl, Update breast cancer 2019 Part 2 - kappaB-integrin-alpha4 axis promotes prostate cancer cell Adhesion to bone
implementation of novel diagnostics and therapeutics in advanced breast cancer marrow endothelium and enhances metastatic potential, Mol. Cancer Res. 18 (3)
patients in clinical practice, Geburtshilfe Frauenheilkd 79 (3) (2019) 268–280. (2020) 501–513.
[64] E. Mezni C. Vicier M. Guerin R. Sabatier F. Bertucci A. Goncalves New therapeutics [90] S. Ziaee, L.W. Chung, Induction of integrin alpha2 in a highly bone metastatic
in HER2-positive advanced breast cancer: towards a change in clinical practices? human prostate cancer cell line: roles of RANKL and AR under three-dimensional
Cancers (Basel) 12(6) (2020) E1573. suspension culture, Mol. Cancer 13 (2014) 208.
[65] American Cancer Society. Cancer Facts & Figures 2020, American Cancer Society [91] H.L. Goel, J. Li, S. Kogan, L.R. Languino, Integrins in prostate cancer progression,
(2020). Endocr. Relat. Cancer 15 (3) (2008) 657–664.
[66] F.S. Markland, K. Shieh, Q. Zhou, V. Golubkov, R.P. Sherwin, V. Richters, [92] M.C. Juan-Rivera, M. Martinez-Ferrer, Integrin inhibitors in prostate cancer,
R. Sposto, A novel snake venom disintegrin that inhibits human ovarian cancer Cancers (Basel) 10 (2) (2018) 44.
dissemination and angiogenesis in an orthotopic nude mouse model, Haemostasis [93] K.A. Kwakwa, J.A. Sterling, Integrin alphavbeta3 signaling in tumor-induced bone
31 (3–6) (2001) 183–191. disease, Cancers (Basel) 9 (7) (2017) 84.
[67] S. Swenson, F. Costa, R. Minea, R.P. Sherwin, W. Ernst, G. Fujii, D. Yang, [94] J.A. Nemeth, M.L. Cher, Z. Zhou, C. Mullins, S. Bhagat, M. Trikha, Inhibition of
F.S. Markland Jr., Intravenous liposomal delivery of the snake venom disintegrin alpha(v)beta3 integrin reduces angiogenesis, bone turnover, and tumor cell pro-
contortrostatin limits breast cancer progression, Mol. Cancer Ther. 3 (4) (2004) liferation in experimental prostate cancer bone metastases, Clin. Exp. Metastasis
499–511. 20 (5) (2003) 413–420.
[68] T.F. Huang, C.H. Yeh, W.B. Wu, Viper venom components affecting angiogenesis, [95] M. Trikha, E. Raso, Y. Cai, Z. Fazakas, S. Paku, A.T. Porter, J. Timar, K.V. Honn,
Haemostasis 31 (3–6) (2001) 192–206. Role of alphaII(b)beta3 integrin in prostate cancer metastasis, Prostate 35 (3)
[69] I.C. Kang, Y.D. Lee, D.S. Kim, A novel disintegrin salmosin inhibits tumor angio- (1998) 185–192.
genesis, Cancer Res. 59 (15) (1999) 3754–3760. [96] D.Q. Zheng, A.S. Woodard, M. Fornaro, G. Tallini, L.R. Languino, Prostatic car-
[70] K.Z. Olfa, L. Jose, D. Salma, B. Amine, S.A. Najet, A. Nicolas, L. Maxime, cinoma cell migration via alpha(v)beta3 integrin is modulated by a focal adhesion
Z. Raoudha, M. Kamel, M. Jacques, S. Jean-Marc, A.A.Lebestatin, a disintegrin kinase pathway, Cancer Res. 59 (7) (1999) 1655–1664.

8
A.H. Schönthal, et al. Biochemical Pharmacology 181 (2020) 114149

[97] H.M. DeLisser, P.J. Newman, S.M. Albelda, Platelet endothelial cell adhesion 1194–1216.
molecule (CD31), Curr. Top. Microbiol. Immunol. 184 (1993) 37–45. [118] H.J. Allen, C. Porter, M. Gamarra, M.S. Piver, E.A. Johnson, Isolation and mor-
[98] Y. Miyata, H. Sakai, Reconsideration of the clinical and histopathological sig- phologic characterization of human ovarian carcinoma cell clusters present in
nificance of angiogenesis in prostate cancer: usefulness and limitations of micro- effusions, Exp Cell Biol 55 (4) (1987) 194–208.
vessel density measurement, Int. J. Urol. 22 (9) (2015) 806–815. [119] M. Bardies, P. Thedrez, J.F. Gestin, B.M. Marcille, D. Guerreau, A. Faivre-Chauvet,
[99] K.L. DeCicco-Skinner, G.H. Henry, C. Cataisson, T. Tabib, J.C. Gwilliam, M. Mahe, C. Sai-Maurel, J.F. Chatal, Use of multi-cell spheroids of ovarian carci-
N.J. Watson, E.M. Bullwinkle, L. Falkenburg, R.C. O’Nneill, A. Morin, J.S. Wiest, noma as an intraperitoneal radio-immunotherapy model: uptake, retention ki-
Endothelial cell tube formation assay for the in vitro study of angiogenesis, J. Vis. netics and dosimetric evaluation, Int. J. Cancer 50 (6) (1992) 984–991.
Exp. 91 (2014) e51312. [120] J.L. Becker, T.L. Prewett, G.F. Spaulding, T.J. Goodwin, Three-dimensional growth
[100] D.D. Bowtell, S. Bohm, A.A. Ahmed, P.J. Aspuria, R.C. Bast Jr., V. Beral, J.S. Berek, and differentiation of ovarian tumor cell line in high aspect rotating-wall vessel:
M.J. Birrer, S. Blagden, M.A. Bookman, J.D. Brenton, K.B. Chiappinelli, morphologic and embryologic considerations, J. Cell. Biochem. 51 (3) (1993)
F.C. Martins, G. Coukos, R. Drapkin, R. Edmondson, C. Fotopoulou, H. Gabra, 283–289.
J. Galon, C. Gourley, V. Heong, D.G. Huntsman, M. Iwanicki, B.Y. Karlan, A. Kaye, [121] I.V. Filippovich, N.I. Sorokina, N. Robillard, J.F. Chatal, Radiation-induced
E. Lengyel, D.A. Levine, K.H. Lu, I.A. McNeish, U. Menon, S.A. Narod, B.H. Nelson, apoptosis in human ovarian carcinoma cells growing as a monolayer and as
K.P. Nephew, P. Pharoah, D.J. Powell Jr., P. Ramos, I.L. Romero, C.L. Scott, multicell spheroids, Int. J. Cancer 72 (5) (1997) 851–859.
A.K. Sood, E.A. Stronach, F.R. Balkwill, Rethinking ovarian cancer II: reducing [122] S.D. Swenson, C. Silva-Hirschberg, F.S. Markland, Methods for evaluation of a
mortality from high-grade serous ovarian cancer, Nat. Rev. Cancer 15 (11) (2015) snake venom-derived disintegrin in animal models of human cancer, Methods Mol.
668–679. Biol. 2068 (2020) 185–204.
[101] Y.L. Liu, Q. Zhou, A. Iasonos, V.N. Emengo, C. Friedman, J.A. Konner, [123] S. Swenson, R. Minea, S. Zidovetzki, C. Helchowski, F. Costa, F.S. Markland, Anti-
R.E. O'Cearbhaill, C. Aghajanian, D. Zamarin, Subsequent therapies and survival angiogenesis and disintegrins, in: R.M. Kini, K.J. Clemetson, F.S. Markland,
after immunotherapy in recurrent ovarian cancer, Gynecol. Oncol. 155 (1) (2019) M.A. McLane, T. Morita (Eds.), Toxins and Hemostasis, Springer, New York, 2010,
51–57. pp. 301–330.
[102] T. May, R. Comeau, P. Sun, J. Kotsopoulos, S.A. Narod, B. Rosen, P. Ghatage, A [124] Q.T. Ostrom, H. Gittleman, P. Farah, A. Ondracek, Y. Chen, Y. Wolinsky,
comparison of survival outcomes in advanced serous ovarian cancer patients N.E. Stroup, C. Kruchko, J.S. Barnholtz-Sloan, CBTRUS statistical report: primary
treated with primary debulking surgery versus neoadjuvant chemotherapy, Int. J. brain and central nervous system tumors diagnosed in the United States in
Gynecol. Cancer 27 (4) (2017) 668–674. 2006–2010, Neuro Oncol. 15 (suppl. 2) (2013) ii1-56.
[103] R. Pelaez, A. Pariente, A. Perez-Sala, I.M. Larrayoz, Integrins: moonlighting pro- [125] R. Stupp, M.E. Hegi, W.P. Mason, M.J. van den Bent, M.J. Taphoorn, R.C. Janzer,
teins in invadosome formation, Cancers (Basel) 11 (5) (2019) 615. S.K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A.A. Brandes,
[104] J. Hatina, M. Boesch, S. Sopper, M. Kripnerova, D. Wolf, D. Reimer, C. Marth, J. Gijtenbeek, C. Marosi, C.J. Vecht, K. Mokhtari, P. Wesseling, S. Villa,
A.G. Zeimet, Ovarian cancer stem cell heterogeneity, Adv. Exp. Med. Biol. 1139 E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J.G. Cairncross, R.O. Mirimanoff,
(2019) 201–221. Effects of radiotherapy with concomitant and adjuvant temozolomide versus
[105] F. Tomao, A. Papa, M. Strudel, L. Rossi, G. Lo Russo, P. Benedetti Panici, radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-
F.R. Ciabatta, S. Tomao, Investigating molecular profiles of ovarian cancer: an year analysis of the EORTC-NCIC trial, Lancet Oncol. 10 (5) (2009) 459–466.
update on cancer stem cells, J Cancer 5 (5) (2014) 301–310. [126] R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn,
[106] M. Varas-Godoy, G. Rice, S.E. Illanes, The crosstalk between ovarian cancer stem K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer,
cell niche and the tumor microenvironment, Stem Cells Int 2017 (2017) 5263974. S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer,
[107] K.L. Sodek, M.J. Ringuette, T.J. Brown, Compact spheroid formation by ovarian R.O. Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide for
cancer cells is associated with contractile behavior and an invasive phenotype, Int. glioblastoma, N. Engl. J. Med. 352 (10) (2005) 987–996.
J. Cancer 124 (9) (2009) 2060–2070. [127] Y. Narita, Bevacizumab for glioblastoma, Ther. Clin. Risk Manage. 11 (2015)
[108] K. Shield, M.L. Ackland, N. Ahmed, G.E. Rice, Multicellular spheroids in ovarian 1759–1765.
cancer metastases: biology and pathology, Gynecol. Oncol. 113 (1) (2009) [128] D.A. Abrams, J.A. Hanson, J.M. Brown, F.P. Hsu, J.B. Delashaw Jr., D.A. Bota,
143–148. Timing of surgery and bevacizumab therapy in neurosurgical patients with re-
[109] K. Jaaback, N. Johnson, T.A. Lawrie, Intraperitoneal chemotherapy for the initial current high grade glioma, J. Clin. Neurosci. 22 (1) (2015) 35–39.
management of primary epithelial ovarian cancer, Cochrane Database Syst. Rev. [129] D.A. Reardon, J.E. Herndon 2nd, K.B. Peters, A. Desjardins, A. Coan, E. Lou,
11 (2011) CD005340. A.L. Sumrall, S. Turner, E.S. Lipp, S. Sathornsumetee, J.N. Rich, J.H. Sampson,
[110] A.G. Zeimet, D. Reimer, A.C. Radl, A. Reinthaller, C. Schauer, E. Petru, N. Concin, A.H. Friedman, S.T. Boulton, D.D. Bigner, H.S. Friedman, J.J. Vredenburgh,
S. Braun, C. Marth, Pros and cons of intraperitoneal chemotherapy in the treat- Bevacizumab continuation beyond initial bevacizumab progression among re-
ment of epithelial ovarian cancer, Anticancer Res. 29 (7) (2009) 2803–2808. current glioblastoma patients, Br. J. Cancer 107 (9) (2012) 1481–1487.
[111] M. Kwa, S. Edwards, A. Downey, E. Reich, R. Wallach, J. Curtin, F. Muggia, [130] K.D. Swanson, E. Lok, E.T. Wong, An overview of alternating electric fields
Ovarian cancer in BRCA mutation carriers: improved outcome after in- therapy (NovoTTF therapy) for the treatment of malignant glioma, Curr. Neurol.
traperitoneal (IP) cisplatin, Ann. Surg. Oncol. 21 (5) (2014) 1468–1473. Neurosci. Rep. 16 (1) (2016) 8.
[112] M. Kwa, F. Muggia, Ovarian cancer: a brief historical overview of intraperitoneal [131] R. Stupp, S. Taillibert, A.A. Kanner, S. Kesari, D.M. Steinberg, S.A. Toms,
trials, Ann. Surg. Oncol. 21 (5) (2014) 1429–1434. L.P. Taylor, F. Lieberman, A. Silvani, K.L. Fink, G.H. Barnett, J.J. Zhu,
[113] C. Marchetti, F. De Felice, G. Perniola, I. Palaia, A. Musella, V. Di Donato, J.W. Henson, H.H. Engelhard, T.C. Chen, D.D. Tran, J. Sroubek, N.D. Tran,
G. Cascialli, L. Muzii, V. Tombolini, P. Benedetti, Panici, Role of intraperitoneal A.F. Hottinger, J. Landolfi, R. Desai, M. Caroli, Y. Kew, J. Honnorat, A. Idbaih,
chemotherapy in ovarian cancer in the platinum-taxane-based era: a meta-ana- E.D. Kirson, U. Weinberg, Y. Palti, M.E. Hegi, Z. Ram, Maintenance therapy with
lysis, Crit. Rev. Oncol. Hematol. 136 (2019) 64–69. tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma:
[114] M. Markman, J.L. Walker, Intraperitoneal chemotherapy of ovarian cancer: a re- a randomized clinical trial, JAMA 314 (23) (2015) 2535–2543.
view, with a focus on practical aspects of treatment, J. Clin. Oncol. 24 (6) (2006) [132] S. Swenson, R.O. Minea, C.D. Tuan, T.Z. Thein, T.C. Chen, F.S. Markland, A novel
988–994. venom-derived peptide for brachytherapy of glioblastoma: preclinical studies in
[115] A.A. Wright, A. Cronin, D.E. Milne, M.A. Bookman, R.A. Burger, D.E. Cohn, mice, Molecules 23 (11) (2018) 2918.
M.C. Cristea, J.J. Griggs, N.L. Keating, C.F. Levenback, G. Mantia-Smaldone, [133] F.C. Greenwood, W.M. Hunter, J.S. Glover, The preparation of I-131-labelled
U.A. Matulonis, L.A. Meyer, J.C. Niland, J.C. Weeks, D.M. O'Malley, Use and ef- human growth hormone of high specific radioactivity, Biochem. J. 89 (1963)
fectiveness of intraperitoneal chemotherapy for treatment of ovarian cancer, J. 114–123.
Clin. Oncol. 33 (26) (2015) 2841–2847. [134] R. Stupp, M.J. van den Bent, M.E. Hegi, Optimal role of temozolomide in the
[116] M. Metwally, Y. Cheong, T.C. Li, A review of techniques for adhesion prevention treatment of malignant gliomas, Curr. Neurol. Neurosci. Rep. 5 (3) (2005)
after gynaecological surgery, Curr. Opin. Obstet. Gynecol. 20 (4) (2008) 345–352. 198–206.
[117] S. Farag, P.F. Padilla, K.A. Smith, M.L. Sprague, S.E. Zimberg, Management, pre- [135] S.Y. Lee, Temozolomide resistance in glioblastoma multiforme, Genes Dis. 3 (3)
vention, and sequelae of adhesions in women undergoing laparoscopic gyneco- (2016) 198–210.
logic surgery: a systematic review, J. Minim. Invasive Gynecol. 25 (7) (2018)

You might also like