ArticlePDF Available

Prevention of in vivo lung tumor growth by prolonged local delivery of hydroxycamptothecin using poly(ester-carbonate)-collagen composites

Authors:

Abstract and Figures

Local tumor recurrence has a major impact on long-term patient survival following the surgical treatment of most cancers, and this is especially true with lung cancer. Consequently, methods to deliver chemotherapeutics locally at a lung tumor resection margin would be beneficial since: 1) systemic treatment approaches are ineffective or highly toxic; 2) the incidence of local recurrence does not warrant universal treatment of all patients with a highly morbid systemic therapy; and 3) surgical resection of recurrent disease is not an option and alternative rescue therapies are generally unsuccessful. To begin to meet this clinical need, we have prepared poly(glycerol monostearate-co-epsilon-caprolactone) films as a controlled, prolonged, and low dose delivery matrix for the potent anticancer agent 10-hydroxycamptothecin (HCPT). These drug-loaded films were applied to a collagen-based scaffold clinically indicated for the mechanical buttressing of lung tissue following surgical resection, resulting in a flexible composite that can be secured to the tissue that releases HCPT over seven weeks and thereby prevents the local growth and establishment of Lewis lung carcinoma tumors in vivo (a freedom of local tumor growth of 86%). In comparison, all animals treated with a larger intravenous dose of HCPT or unloaded composites developed rapid local tumors.
Content may be subject to copyright.
Prevention of in vivo lung tumor growth by prolonged local delivery of
hydroxycamptothecin using poly(ester-carbonate)-collagen composites
Jesse B. Wolinsky
a,b
, Rong Liu
c
, Joe Walpole
c
, Lucian R. Chirieac
d
, Yolonda L. Colson
c
, Mark W. Grinstaff
a,b,
a
Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
b
Department of Chemistry, Boston University, Boston, MA 02215, United States
c
Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, United States
d
Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, United States
abstractarticle info
Article history:
Received 7 January 2010
Accepted 15 February 2010
Available online 22 February 2010
Keywords:
Prevention
Lung tumors
Recurrence
Films
Local delivery
Local tumorrecurrence has a major impacton long-term patient survival followingthe surgical treatmentof most
cancers, and this is especiallytrue with lung cancer. Consequently, methods to deliver chemotherapeutics locally
at a lung tumor resection margin would be benecial since: 1) systemic treatment approaches are ineffective or
highly toxic;2) the incidence of localrecurrence does not warrant universaltreatment of all patients with a highly
morbid systemic therapy; and 3) surgical resection of recurrent disease is not an option and alternative rescue
therapies are generally unsuccessful. To begin to meet this clinical need, we have prepared poly(glycerol
monostearate-co-ε-caprolactone) lms as a controlled, prolonged, and low dose delivery matrix for the potent
anticancer agent 10-hydroxycamptothecin (HCPT). These drug-loaded lms were applied to a collagen-based
scaffold clinically indicated for the mechanical buttressing of lung tissue following surgicalresection, resulting in
aexible composite that can be secured to the tissue that releases HCPT over seven weeks and thereby prevents
the local growth and establishment of Lewis lung carcinoma tumors in vivo (a freedom of local tumor growth of
86%). In comparison, all animals treated with a larger intravenous dose of HCPT or unloaded composites
developed rapid local tumors.
© 2010 Elsevier B.V. All rights reserved.
1. Introduction
Polymers have enabled increasingly sophisticated therapies for the
diagnoses and treatment of cancer over the last two decades. Drugs,
contrast agents, and targeting moieties have been covalently-bound or
entrapped by a polymer in the form of prodrugs, micelles, particles, or
bulk material in an attempt to both increase solubility and localize
delivery to tumors via systemic targeting or local delivery [13].The
treatment of choice for localized solid tumors is surgery, where the
feasibility of removing all of the cancerous tissue is balanced against the
resultant morbidity to the patient. Local recurrence is of signicant
concern following primary treatment for many cancers including head
and neck [4],breast[5], lung [6],colon[7],rectal[8], and pancreatic [9]
malignancies. Locally recurrent tumors are initiated by residual cancer
cells remaining at or near the resection margins or site of initial
treatment. Given that microscopic disease can remain,surgical resection
is often used in conjunction with radiation and/or chemotherapy to
improve local control. While radiation and chemotherapy are common-
ly utilized as adjuvant therapies for more advanced primary cancers,
preventative therapy is administered only in selected cases to decrease
recurrence risk, due to theoften severe side effectsassociated with these
treatments and the inability to accurately predict in which patients the
benets of treatment would warrant the additional morbidity.
Consequently, a drug delivery platform that locally delivers thera-
peutic doses of drug directly to the site at highest risk for recurrent
disease, prevents recurrence at resection margins, and diminishes
signicantly the systemic toxicity associated with intravenous chemo-
therapy and external radiation, would offer signicant advantages over
other approaches. Loco-regional delivery is particularly benecial in
situations where: 1) therapeutic levels of chemotherapy are not
achievable due to poor aqueous solubility, non-ideal pharmacokinetics
or biodistribution; 2) systemic treatment approaches are ineffective or
highly toxic; 3) the incidence of local recurrence does not warrant
universal treatment of all patients with a highly morbid systemic
therapy; or 4) surgical resectionof recurrent disease is not an option and
alternative rescue therapies are generally unsuccessful. All of these
examples are characteristic of the clinical practice of lung cancer surgery.
Our interest is to utilize a tunable drug-eluting polymeric delivery
system to prevent recurrence of malignant disease in lung cancer
patients, as limited pulmonary reserve makes this patient population
particularly susceptible to local tumor recurrence. To this aim, we
have developed a tunable polymeric drug delivery platform. Herein
we describe poly(glycerol monostearate-co-ε-caprolactone) lms
loaded with the potent anticancer agent 10-hydroxycamptothecin
Journal of Controlled Release 144 (2010) 280287
Corresponding author. Department of Biomedical Engineering, Boston University,
Boston, MA 02215, United States.
E-mail address: mgrin@bu.edu (M.W. Grinstaff).
0168-3659/$ see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.jconrel.2010.02.022
Contents lists available at ScienceDirect
Journal of Controlled Release
journal homepage: www.elsevier.com/locate/jconrel
(HCPT), an analogue of irinotecan which has been clinically utilized in
the treatment of various lung cancer subtypes [10,11]. These lms can
be applied to a collagen-based scaffold clinically indicated for the
mechanical buttressing of lung tissue following surgical resection,
resulting in a exible composite that can be secured to the tissue along
the resection margins. We demonstrate that these lms are capable of
providing effective sustained release of HCPT over seven weeks and
thereby prevent the local growth and establishment of malignant cells
in an in vivo model of microscopic disease.
2. Materials and methods
2.1. Materials
All solvents were dried and freshly distilled before use or were
purchased from Sigma (Toluene). CH
2
Cl
2
was distilled under N
2
from
calcium hydride. Stannous 2-ethylhexanoate, ε-caprolactone, stearic
acid, N,N-dicyclohexylcarbodiimide, and 4-(dimethylamino)pyridine
were purchased from Aldrich. Palladium on carbon was purchased
from Acros. 10-hydroxycamptothecin was purchased from Sigma.
5-benzyloxy-1,3-dioxan-2-one was prepared as previously reported
[12]. All reactions were performed under nitrogen atmosphere unless
otherwise noted. NMR spectra were recorded on a Varian INOVA spec-
trometer (
1
H at 400 MHz). Chemical shifts were referenced to residual
solvent peaks (CHCl
3
peak at 7.24 ppm). DCM= dichloromethane,
DCC = N,N-dicyc lohexylcarbodiimid e, DM AP =4 -(dimethylamino)
pyridine, HCPT= 10-hydroxycamptothecin, PGC-OH= poly(glycerol-
co-ε-caprolactone), PGC-C18 = poly(glycerol monostearate-co-ε-
caprolactone), Pd/C= 10% palladium on activated carbon, and PBS=
phosphate buffered solution.
2.2. Synthesis of poly(glycerol-co-ε-caprolactone)
Poly(glycerol-co-ε-caprolactone), or PGC-OH, was prepared as
reported previously [12].Briey, the ε-caprolactone and 5-benzyloxy-
1,3-dioxan-2-one monomers were mixed at a 4:1 molar ratio in a
schlenk ask and subsequently evacuated and ushed with N
2
three
times. Sn(oct)
2
was used (M/I=500) to catalyze the ring-opening
polymerization of the co-monomers at 140 °C for 48 h, and the resulting
copolymer was isolated by precipitation in cold methanol (92% yield).
The benzyl-protecting groups were removed via palladium-catalyzed
hydrogenolysis (99% yield).
1
H NMR (CDCl
3
)1.301.40 (m, 2H, CH
2
),
1.571.68 (m, 4H, CH
2
CH
2
), 2.252.28 (m, 2H, CH
2
), 3.82 (broad p, 1H,
OCH), 4.034.06 (t, 2H, OCH
2
), and 4.104.21 (m, 4H, OCH
2
CH).
2.3. Synthesis of poly(glycerol stearic acid-co-ε-caprolactone)
Stearic acid (0.78 mmol), poly(glycerol-co-ε-caprolactone) (2.6 mmol),
dicyclohexylcarbodiimide (0.63 mmol), and 4-(dimethylamino)pyridine
(0.26 mmol) were dissolved in CH
2
Cl
2
. The solution was stirred at
room temperature for 18 h. The DCU was ltered and the solvent
evaporated. The product, PGC-C18, was dissolved in CH
2
Cl
2
and
precipitated in cold methanol. The solvent was decanted and subse-
quently dried by evaporation (83% yield). Addition of the stearic acid
side chain was determined by the presence of the methylene group at
the end of the alkyl chain, with a corresponding peak in the
1
HNMR
spectrum at 0.82 (m, 3H, CH
3
).
2.4. Preparation of lms on glass
Films (5 mg polymer; 100 μg HCPT) were cast from polymer
solutions onto glass cover-slips using a microsyringe. First, 10-
hydroxycamptothecin (HCPT) was added to CH
2
Cl
2
(0.1% w/v) in a
glass vial and homogenized for 60 min in a sonication bath to break
apart aggregates. Polymer (5% w/v) was then dissolvedinto the solution
by vortexing for 1 min. The solution was slowly added to the glass
surface and left to evaporate for 24 h, followed by further drying under
reduced pressure for another 24 h.
2.5. Preparation of lms on scaffolds
Polymer lms (3 mg polymer; 60 μg HCPT) were adhered to
collagen-based scaffolds for in vivo implantation studies. Peri-Strips
Dry® with Veritas® Collagen Matrix (Synovis LT) collagen strips were
chosen as a viable scaffolding material based on their wide use as
surgical staple-line reinforcement materials and demonstrated bio-
compatibility in human patients. HCPT-loaded lms were adhered to
the collagen scaffolds using a modied method as was used for the
glass cover-slips. Following the application and complete drying of a
base layer of unloaded polymer, an additional layer of HCPT-loaded
polymer was added, evaporated, and dried under reduced pressure.
2.6. Size exclusion chromatography
Molecularweight determinations were performed via size exclusion
chromatography using THF as the eluent on a Polymer Laboratories
PLgel 3 µm MIXED-E column (3 µm bead size) and a Rainin HPLC
system (temp= 25 °C; ow rate=1.0 mL/min). Polystyrene standards
(Polysciences, Inc.) were used for calibration. These data conrm that
the post-modication of PGC-OH with stearic acid did not result in
polymer degradation (PGC-OH: M
n
=8600, M
w
/M
n
=1.96; PGC-C18:
M
n
=12,300,M
w
/M
n
=1.85).
2.7. Scanning electron microscopy
Samples for scanning electron microscopy were prepared by
mounting the lms on an aluminum sample stub and then sputter-
coating with a 5 nm layer of goldpalladium alloy. Samples were then
imaged on a Zeiss SUPRA 40VP eld emission scanning electron
microscope using an accelerating voltage of 2 kV. Cross-sectional images
were obtained from lms that were fractured after submersion in liquid
nitrogen and subsequently clamped perpendicular to the base.
2.8. HCPT release kinetics from lms
The release kinetics from HCPT-loaded lms (n=4) were assessed
in PBS buffer at 37 °C. Specically, lms comprised of 5 mg polymer
(PGC-OH or PGC-C18) and 100 µg HCPT were cast onto glass cover-slips
and submerged in 50 mL of PBS. At specic time points, an aliquot of
release media was removed and the concentration of HCPT was
measured by uorescence spectroscopy (λ
Ex
=382 nm and
λ
Em
=550 nm, Photon Technology International QM-4/2005 spectro-
uorimeter). The release medium was changed at regular intervals to
maintain the pH and ensure sink conditions. A calibration curve was
constructed from nine samples of known HCPT concentrations with
linearity observed from 10 nM to 5 µM (R
2
=0.999) from which the
drug concentration of each aliquot was determined. After the
completion of release, the HCPT remaining in each lm was quantied.
Films were dissolved in CH
2
Cl
2
to release encapsulated drug, the solvent
evaporated, and PBS (150 mL) was added under rigorous stirring. The
uorescence spectrum was recorded and remaining drug measured.
2.9. Stability of HCPT
The ratio of active lactone to inactive carboxylate HCPT was
quantied by collecting aliquots of release media before signicant
conversion to the carboxylate form could occur. Samples were analyzed
by HPLC using an adapted method reported previously [13]. AnHP 1090
HPLC system was used with a uorescence detector (λ
ex
=380 nm,
λ
em
=540 nm), with a Phenomenex Prodigy 5 ODS reverse-phase
column (150× 4.6 mm, 5 μm). The mobile phase was composed of 25%
acetonitrile and 75% ammonium acetate buffer adjusted to a pH of 6.4
281J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
and delivered at 0.8 mL/min. Calibration curves were constructed for
both lactone (rt=3.7 min) and carboxylate (rt= 2.8 min) forms with
sensitivities of 0.5 ng/mL and 2 ng/mL respectively. At designated time
points, drug-loaded lms were transferred to fresh PBS (pH=6.4,
adjusted with acetic acid) for 60 min at 37 °C and aliquots were
removed andimmediately analyzed. HCPT converted from 100% lactone
form to 87% lactoneat pH = 6.4 over60 min for the control experiments.
The stability study was performed until the amount of drug released
became lower than our limit of detection. Long-term release kinetics
were also monitored for 58 days and conrmed the release prole
observed from the uorescence experiment. Cumulative release was
measured by removing aliquots from the release media and replaced
with fresh media at regular intervals to maintain sink conditions.
2.10. In vitro cytotoxicity assays
In vitro experiments were performed with Lewis lung carcinoma
(LLC) cells received as a kind gift from Dr. Judah Folkman's laboratory
at Children's Hospital, Boston and cultured with medium containing
10% FBS and 1% PS. Cells were plated at a density of 30,000 cells/well
in 24-well Transwell plates (Corning Inc.) and grown overnight. Films
were sterilized overnight under UV light and placed on tissue culture
treated polyester membrane inserts (0.4 μm pore size) before being
incubated with cultured cells and adding serum-containing culture
media. Films were co-incubated with the cells for 5-day intervals
before being transferred to new wells with freshly-plated cells to
assess continued drug release and cytotoxicity in the setting of new
media and freshly-plated tumor cells. To avoid reaching the HCPT
solubility limit (and thereby to decrease the rate and duration of drug
release), shorter intervals were used over the rst 5 days to simulate
the anticipated drug gradient present in living tissues. Cell viability
was quantied using a standard MTT cell proliferation assay.
2.11. Animal and tumor models
All animal studies were approved by the Institutional Animal Care
and Use Committee of Dana-Farber Cancer Institute, and all animal
care was performed humanely and in accordance with the designated
and approved standards. Female C57BL/6J mice at six to eight weeks
of age were obtained from Jackson Laboratories (Bar Harbor, Maine).
Mice were anesthetized with intraperitoneal injection of ketamine
(120 mg/kg) and xylazine (8 mg/kg). A 0.8 cm wide subcutaneous
pocket was prepared by blunt dissection under sterile conditions
between the shoulders on the dorsum of the mice. Ultraviolet-
sterilized polymer lms dried on pericardial strips (1.0 × 0.8 cm) with
or without HCPT (60 µg per lm) were inserted into the subcutaneous
pockets. The incision was closed with 50 polypropylene sutures.
Mice receiving sham surgery without lm implantation served as
control for wound healing studies (n= 3). Mice receiving an
intravenous injection (i.v.) of 200 µg HCPT served as a second control
group for administration of HCPT (dissolved in DMSO and diluted
with PBS (1:5 v/v)). The HCPT solution consisted of 100% lactone form
as measured by HPLC and was injected immediately upon formula-
tion. The body weights of mice were measured with a digital balance
biweekly. White blood cell counts from tail vein blood were
performed at day 10 after surgery.
Wound healing, including the integrity of skin and any signs of
wound infection, was assessed daily without knowledge of the type of
the lms implanted. Implanted lms and the surrounding tissues
were sampled for histological evaluation 10 days after surgery and
xed in 10% formalin. Formalin-xed, parafn-embedded pathology
specimens from each mouse were cut into 5-μm thick serial sections
and hemotoxylin and eosin (H&E) staining was performed. Micro-
scopic examination of the H&E sections was assessed independently
by a pathologist (L. Chirieac), without knowledge of the type of
implant or drug delivery regimen. Each specimen was evaluated for
tissue integrity, stage of infection, the magnitude and type of local
inammatory changes, reactive brosis, scar and granulation tissue
formation.
In the tumor establishment study, incisions were allowed to heal
for two days before LLC cell injection to avoid leakage of the cells. LLC
cells (750,000), suspended in 0.1 mL of PBS, were injected subcuta-
neously on top of the implanted lm via a 27-gauge needle. Tumor
growth and its local relationship to lms were monitored by palpation
and a digital caliper biweekly. Mice were euthanized if tumors
reached 2 cm in size. Rapid tumor growth in control mice and
morbidity resulting from distal metastases required sacrice of
multiple animals 2 weeks following LLC-injection, and thus equiva-
lent comparison of tumor growth was performed within 14 days of
tumor inoculation for all mice in the study.
2.12. Statistical analyses
Analysis of variation (ANOVA) was used to compare body weights
and percent change of body weight between experimental and
control animals; the post-hoc Tucky's multiple comparison test was
performed if the overall difference was signicant (Pb0.05). The WBC
counts were compared by Student's t-test. The incidence of tumor
growth overtopping lms in mice that received either HCPT-loaded
or unloaded lms was compared with Fisher's exact test. All sta-
tistics were performed by GraphPad Prism 4.0 software (La Jolla, CA).
Pb0.05 is considered statistically signicant. All data are presented as
mean±s.e.m.
3. Results
3.1. Film formation, characterization, and drug release studies
HCPT-loaded poly(glycerol-co-ε-caprolactone) (PGC-OH) and
poly(glycerol stearic acid-co-ε-caprolactone) (PGC-C18) lms were
prepared and evaluated. PGC-OH was synthesized as reported
previously[12]. PGC-C18 was synthesized by conjugating stearic acid
to PCG-OH to yield a signicantly more hydrophobic copolymer
(contact angles of PGC-OH and PGC-C18 lms were 87° and 125°,
respectively). The drug was pre-mixed in the solvent and homoge-
nized in an ultra-sonication bath to yield a colloidal solution of drug
particulates, followed by the addition of polymer and lm casting. In
this procedure, small insolubilized drug aggregates were embedded
into the polymer matrix as the volatile solvent evaporated. The PGC-
OH lms fractured over the rst few days when exposed to PBS at
37 °C. Conversely, the PGC-C18 formed uniform lms with improved
mechanical integrity. The scanning electron micrograph of a repre-
sentative PGC-C18 lm is shown in Fig. 1. The lm has a uniform
thickness (40 µm) with smooth surfaces and a homogenous non-
porous microstructure is evident via a cross-sectional view.
Drug-loaded PGC-C18 lms demonstrated gradual and sustained
release kinetics with no signicant initial burst of HCPT as compared to
unmodiedPGC-OH (Fig. 2) or other well known polymers such as PLGA
[14]. Specically, PGC-OH released 25% of its load over two days and
continued to elute drug for approximately three weeks. In contrast,
HCPT-loaded PGC-C18 lms released in a much more controlled
manner, initially releasing about 2% loading per day for the rst two
weeks, decreasing to approximately 1% per day for the following three
weeks, before diminishing to 0.5% release per day, with continued
release detected for at least 7 weeks. The lms were dissolved following
release and 100% mass balance was observed for HCPT in both groups.
The lms were also cast onto pericardial substrates for mechanical
support; pericardial scaffolds are currently used as reinforcement
materials for several types of surgeries including bariatric and
cardiothoracic procedures. The release kinetics of loaded PGC-C18
lms from pericardial substrates were similar to lms cast on glass
supports (Fig. 3). The lactonecarboxylate conversion of HCPT was
282 J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
investigated for loaded PGC-C18 lms. The drug was loaded into the
polymer in its 100% lactone (active) form and at regular intervals, the
lms were transferred to fresh release media (pH =6.4) for 60 min, the
media collected for stability analysis, and the lms transferred to fresh
media forlong-term release.In this manner,the lactone stabilityof HCPT
remaining in the lms over time can be determined. The conversion
from 100% lactone to carboxylate in these conditions over 60 min
yielded approximately 87% lactone form in the control studies. The
conversion to the carboxylate form was greatly reduced usingour lms,
stabilizing to about 84% lactone form over 49 days of release (Fig. 4). For
comparison, the t
1/2
of HCPT in PBS at 37 °C is reported as 22 min with
80% conversion of the lactone to the carboxylate occurring at
equilibrium [15].
3.2. Cytotoxicity studies
To determine the anti-proliferative efcacy and duration of
effective drug release for this polymeric lm on a lung cancer cell
line, HCPT-loaded PGC-C18 (2% drug w/w) lms were exposed to
cultured Lewis Lung Carcinoma (LLC) cells for 5-day intervals up to
50 days. Based on the release kinetic experiments described above, a
minimum HCPT concentration of 1 µM accumulates at the end of each
5-day interval, well-above the measured HCPT IC
50
of LLC cells
(b100 ng/mL). The lms exhibited signicant cytotoxicity for each
5-day interval exposure during the 50 day experiment (Fig. 5),
demonstrating that the lms are capable of releasing a sustained
dose of therapeutic drug for at least 7 weeks. In contrast, unloaded
PGC-C18 lms were not cytotoxic for the duration of the experiment.
3.3. Evaluation of composites in a lung cancer animal model
Based on the positive results from the in vitro study, a similar
HCPT-loading was used for these in vivo tests. Poly(glycerol stearic
acid-co-ε-caprolactone) was selected over PGC-OH as a delivery
matrix due to its preferable lm stability and release properties.
HCPT-loaded (2% wt/wt; 60 μg of HCPT) PGC-C18 lms adhered to
pericardial substrates were implanted subcutaneously into C57BL/6
mice. Two days were allowed for the healing of the incision before
Lewis lung carcinoma cells were injected on top of the lms to
approximate microscopic malignant disease. Local tumor growth was
dened as the development of subcutaneous tumor nodules overtop
the implanted lms. The experimental mice that received the polymer
lms loaded with HCPT (0.4 μg/mm
2
) exhibited a freedom of local
tumor growth of 86% (6/7 mice). In contrast, all but one mouse from
the animal group treated with unloaded polymer lms developed
signicant tumor growth within 14 days, for a freedom of local tumor
Fig. 1. Top. Chemical structures of poly(glycerol-co-ε-caprolactone) (PGC-OH) and poly(glycerol monostearate-co-ε-caprolactone) (PGC-C18). Bottom. Scanning electron micrographs of
10-hydroxycamptothecin-loaded poly(glycerol monostearate-co-ε-caprolactone) lms (left: cross-section; right:surface).
Fig. 2. Release proles of 10-hydroxycamptothecin from poly(glycerol-co-ε-caprolactone)
lms (n=3). PGC-OH: poly(glycerol-co-ε-caprolactone), PGC-C18: poly(glycerol mono-
stearate-co-ε-caprolactone).
283J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
growth of less than 12% (1/9 mice) (Fig. 6). Additionally, all of the
mice treated with a clinically-relevant intravenous dose of HCPT
(200 µg, approximately 10 mg/kg) developed rapid local tumor
growth. For several mice, tumor cells gained access to more distant
regional areas as a consequence of the tumor cell injection procedure
resulting in the delayed development of small regional tumors outside
the periphery of the HCPT-loaded polymer lms, also highlighting the
local nature of HCPT release from the lms.
Notably, this result indicates that this delivery strategy is local, and
does spare healthy surrounding tissues from high levels of drug,
thereby minimizing chemotherapy-induced toxicity. Consistent with
local delivery, there was no clinical evidence of impaired healing in
mice treated with unloaded or HCPT-loaded lms. The minimal
inammatory reaction and brosis was consistent with normal
wound healing and was indistinguishable between mice receiving
HCPT-loaded and unloaded lms, and mice undergoing sham surgery
(Fig. 7). There was no evidence of persistent local inammation,
infection, or subcutaneous uid accumulation upon inspection in any
group. Histological analysis of tissues at the surgical site demonstrat-
ed only mild inammatory changes, brosis and granulation tissue
formation consistent with the acute phase of normal, post-surgical
wound healing process. Mice receiving either unloaded or HCPT-
loaded lms did not experience any signicant decrease in body
weight over time. White blood cell counts were comparable between
mice implanted with unloaded or HCPT-loaded lms (9.53 ±
1.26×10
6
/mL, 7.00±0.23 × 10
6
/mL, respectively; PN0.05; Fig. 8).
4. Discussion
Although the eld of polymeric delivery devices for cancer
therapies is undergoing rapid growth, there has been a limited focus
on polymer implants for prevention of local tumor recurrence.
Our approach utilized a novel copolymer composed of glycerol and
ε-caprolactone biocompatible building blocks whose properties can
be tuned via functionalizable side chain group modication to yield a
delivery system with prolonged release kinetics that can be applied to
pericardial strips already approved for suture line reinforcement in
lung surgery [12]. The resulting composites are thin and exible (see
Fig. 3), and easily administered along the surface or resection margins
of soft tissue via the application of a standard surgical stapler
commonly utilized in the resection of localized tumors. Specically,
poly(glycerol-co-ε-caprolactone) (PGC-OH) was chosen as a delivery
matrix because the copolymer can be easily modied to affect a range
of properties (i.e., crystallinity, permeability, hydrophobicity, etc.)
known to alter drug release from bulk polymer [12,16]. A second
Fig. 3. Left. Release of HCPT-loaded poly(glycerol monostearate-co-ε-caprolactone) lms off glass or pericardial substrates (n= 3). Right. Photograph of a exible PGC-C18 lm
adhered to pericardium while submerged in phosphate buffered solution after 1 day.
Fig. 4. Stability of the lactone form of 10-hydroxycamptothecin remaining in poly
(glycerol monostearate-co-ε-caprolactone) over time (n= 4). Media collected after
60 min of release at pH = 6.4, dashed line represents conversion of control HCPT in
same conditions.
Fig. 5. Proliferation of LLC cells after exposure to HCPT-loaded poly(glycerol
monostearate-co-ε-caprolactone) lms (n= 3). Fresh cells were exposed to lms for
multi-day intervals over the release life of the lms. For example, after 45 days of
release in vitro, drug-loaded lms were exposed to a new plate of fresh cells for ve
days demonstrated signicant cytotoxicity compared to unloaded lms. Percent
proliferation is normalized to positive serum control.
284 J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
polymer comprised of poly(glycerol monostearate-co-ε-caprolactone)
(PGC-C18), a stearic acid-modied analogue of PGC-OH, was also
synthesized for these studies (Fig. 1). By doing so, we were able to
investigate two copolymer analogues with identical back-bone struc-
tures yet featuring disparate side chains, ultimately leading to
substantially different physical properties. Fatty acid side chains were
incorporated into this polymer in order to increase the hydrophobicity
of the polymer matrix and allow the comparison of drug release
kinetics from polymer analogues with markedly divergent hydropho-
bicities. An in-depth evaluation of a variety of other lipophilic PGC-OH
analogues in addition to PGC-C18 was performed, and the results of
these experiments will be reported elsewhere.
10-Hydroxycamptothecin (HCPT) was selected as a prototype
drug because it is a known chemotherapeutic agent that has
demonstrated strong anti-tumor activity but whose practical use
has been limited due to poor aqueous solubility and instability in
aqueous solution. Furthermore, like the FDA-approved camptothecin
Fig. 6. Freedom from tumor growth overtop lms implanted subcutaneously is sig-
nicantly higher for HCPT-loaded lms compared to unloaded (P= 0.0022), HCPTi.v.
(P=0.0005) and sham surgery (P=0.0005) (symbols used for clarity). For the sham
surgery and i.v. control, growth occurred at the site of tumor cell injection. Some mice
were euthanized before day 14 due to excessive tumor mass. *Mice with loaded lms
eventually develop tumor distant from the lm, conrming that drug release is local.
HCPT content: loaded lm (60 µg), i.v. injection (200 µg).
Fig. 7. Normal wound healing after polymer lm implantation. A. Sham surgery (× 40); B. unloaded lms (×40); C. loaded lm (× 40); D. sham surgery (× 200); E. unloaded lms
(×200); F. loaded lm (× 200).
Fig. 8. White blood cell count of mice implanted with unloaded or HCPT-loaded lms
ten days following treatment (PN0.05).
285J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
analogues irinotecan and topotecan, systemic administration of HCPT
is associated with signicant morbidity [17]. Thus, the entrapment of
HCPT in the copolymer lms was expected to circumvent the
solubility obstacle and reduce side effects by locally delivering the
anticancer agent directly to the site of microscopic malignant disease.
Additionally, the camptothecins have a pH-sensitive lactone ring that
opens to become inactive at a physiological pH (t
1/2
=22 min) [18].In
agreement with other polymer systems, the polymer-embedded drug
was mostly preserved in its active lactone form [18,19]. As PGC-18
does not degrade appreciably over at least six months, the
preservation of the lactone form of HCPT was likely due to its
protection from the aqueous environment inside the polymer matrix.
10-Hydroxycamptothecin has been formulated into polymer
nanoparticles [2527], microparticles [28],andmicellarstruc-
tures [29,30] and demonstrated some inhibition of primary tumor
growth following systemic injection but efcacy against local tumor
recurrence has yet to be demonstrated with these controlled release
formulations. PEGylated niosomes were modied with transferrin for
receptor-mediated delivery of HCPT and shown to inhibit tumor
growth (79% inhibition) signicantly more than non-transferrin
modied niosomes (46%) and intravenous HCPT injection (29%) in a
sarcoma murine model as reported by Hong et al. [31]. Similarly,
Zhang et al. described HCPT-loaded poly(caprolactone-co-lactide)-b-
PEG-b-poly(caprolactone-co-lactide) nanoparticles that demonstrat-
ed inhibition of tumor growth rate by 80% in a sarcoma murine model
compared to that of the systemic injection of HCPT (67%) [25].
Perhaps more importantly, polymeric delivery of camptothecin drugs
has been shown to stabilize camptothecin molecules in their active
lactone form while the drug is embedded in its polymer delivery
vehicle. Shenderova et al. reported that encapsulation of HCPT within
poly(lactide-co-glycolide) microspheres stabilizes the camptothecin
in its lactone form over at least two months of drug release in vitro,
attributed most likely to the acidic environment present within the
slowly-degrading microsphere [28]. Hateet al. observed a similar
result with biodegradable injectable ε-caprolactone oligomers over a
release period of 16 weeks. The minimal conversion to the carboxyl-
ate form was attributed to the impedance of water into the
hydrophobic oligomer, delaying polymer degradation and subsequent
creation of an acidic microenvironment [19].
For our application, lung cancer recurrence represents a signicant
clinical challenge in patients who commonly have limited pulmonary
reserve. Surgery for lung cancer patients with poor lung function due
to emphysema or other underlying lung diseases is often limited to
non-anatomic wedge resections of the involved parenchyma rather
than the traditional lobectomy which involves removal of signicantly
more lung tissue. Unfortunately, when compared to lobectomy,
smaller wedge resections are associated with a signicantly higher
incidence of microscopically positive surgical resection margins and
double the local recurrence rate to 16% [20]. Given the limited
treatment options available for recurrent disease, and, the fact that
the majority of these patients will die as a result of their disease,
interventions to prevent recurrence are critically important to this
patient population [21,22]. Therefore, a lung cancer model was chosen
to establish proof of concept for the described HCPT-loaded PGC-C18
lm delivery system in vivo.
Today, there are only a handful of delivery devices reported for the
prevention of local growth of early or residual disease and only one of
these technologies has been successfully translated towards clinical
use. Paclitaxel-loaded thermosensitive chitosan-based hydrogels,
developed by Leroux et al., were implanted four days after tumor
cell inoculation and demonstrated a signicant decrease, but not
complete inhibition, of tumor growth. Matsuda et al. created a
polyurethane-based pouch which was sutured subcutaneously in
tumor-bearing mice and loaded with gemcitabine three days after
tumor inoculation [23]. Four of six mice supporting loaded devices
had no observable tumor mass during the 30 day observation period,
but the remaining two mice developed tumors at a rate comparable to
the control mice. Cross-linked chitosan hydrogels have also been
loaded with a radioisotope to deliver localized radiotherapy for
prevention of tumor recurrence in breast cancer [24]. Implants loaded
with
131
I-norcholesterol were co-implanted with 4T1 metastatic
mammary mouse tumor cells to simulate microscopic residual disease
and tumor growth was prevented in 69% of the mice. The most
established local delivery device to date is used in the treatment of
malignant glioma, an aggressive brain cancer that often recurs near
the resection margins of the primary tumor. Commercially manufac-
tured by MGI Pharma (now Eisai) under the brand name Gliadel®, the
device delivers the chemotherapeutic carmustine over several days
from a rigid biodegradable polyanhydride wafer placed near the
resection margins, that degrades within three weeks of implantation.
Patients receiving these wafers, which are placed in the resection
cavity after tumor debulking, exhibited an increase in mean survival
from 20 weeks to 28 weeks. Although the wafer had a modest impact
on the survival of treated patients, patients reported a markedly
higher quality of life compared to those treated by conventional
systemic chemotherapy [3234]. These previous results inspire the
development of additional technologies directed towards improving
clinical efcacy and expanding local treatment to other types of
tissues.
An important design requirement for this application is the
sustained release of drug over several weeks where microscopic
disease is most likely to reside. Continuous administration of systemic
chemotherapy is precluded by dose-limiting toxicity, while post-
operative treatment is avoided due to concerns of wound healing
impairment and increased risk of infection. These complications are
circumvented given that prolonged local delivery requires a substan-
tially smaller amount of drug, thereby minimizing systemic complica-
tions. Moreover, recurrent solid tumors will develop at a positive
margin following resection with a much more rapid growth rate
compared to the initial primary tumor [35].As1015% of cells in a
solid tumor are expected to be in the DNA synthesis phase, only those
cells would experience increased sensitivity to anti-neoplastic agents
at a given time point [3638]. For these reasons, we expect that the
continuous exposure of a chemotherapeutic drug to tumor cells over
multiple cell cycles would be signicantly more effective than a bolus
or short term administration of drug.
The controlled and sustained drug delivery from poly(glycerol
monostearate-co-ε-caprolactone) lms described in this study offers
several unique advantages over other local delivery vehicles. First,
complete inhibition of local tumor establishment was achieved at the
surface of the HCPT-loaded implanted lms in all but one animal (a
dramatic result when compared to prior studies) whereas a standard
intravenous dose of HCPT three times greater than the dose delivered
by a lm did not affect tumor growth. Second, the copolymer
implemented here can be chemically modied for tunable drug
release kinetics, as exemplied by the unmodied (PGC-OH) and
lipophilic (PGC-C18) derivative lms compared in this study. For
example, the burst release from hydrophilic PGC-OH is likely due to
the dissolution of HCPT deposited at the surface of the PGC-OH lm
and this effect may be exacerbated as the lm fractures, thus
increasing the surface area of the lm and facilitating rapid release
over several days. The hydrophobic PGC-C18 demonstrated no such
burst. Third, highly localized delivery was observed, since local tumor
growth was prevented but the surrounding tissue was unaffected by
HCPT, displaying only mild local inammation associated with normal
wound healing. This observation indicates that only treated surfaces
receive signicant concentrations of chemotherapy, thus enabling
normal healing and preserving surrounding healthy tissue. Finally, the
HCPT-polymer platform is assembled via facile processing methods,
allowing for the preparation of exible lms with custom shapes and
surface areas. The results suggest that a polymeric lm-based delivery
system can be locally administered to prevent tumor cell proliferation
286 J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
and recurrence following limited surgical resections and other
primary therapies where surgical margins are more likely to exhibit
small foci of microscopic residual tumor. We are developing animal
models to assess the efcacy of these lms to prevent recurrent tumor
growth in vivo following surgical resection. The encouraging results
obtained in the current study warrant further exploration of drug-
loaded lm-based implants for the delivery of anticancer agents to
regions of potential microscopic disease with the ultimate goal of
preventing tumor recurrence following surgical resection.
Acknowledgements
This work was supported in part by a grant from the Wallace H.
Coulter Foundation (MWG & YLC) and the George H.A. Clowes, Jr., MD,
FACS Memorial Research Career Development Award (YLC) through
the American College of Surgeons. The authors wish to thank Boston
University and Brigham and Women's Hospital for their support, and
the Animal Resources Facility at Dana-Farber Cancer Institute for their
excellent animal care.
References
[1] W.M. Saltzman, L.K. Fung, Polymeric implants for cancer chemotherapy, Adv.
Drug Deliv. Rev. 26 (1997) 209230.
[2] T.M. Allen, P.R. Cullis, Drug delivery systems: entering the mainstream, Science
303 (2004) 18181822.
[3] M.A. Moses, H. Brem, R. Langer, Advancing the eld of drug delivery: taking aim at
cancer, Cancer Cell 4 (2003) 337341.
[4] S.J. Wong, M. Machtay, Y. Li, Locally recurrent, previously irradiated head and neck
cancer: concurrent re-irradiation and chemotherapy, or chemotherapy alone?
J. Clin. Oncol. 24 (2006) 26532658.
[5] M. Clemons, S. Danson, T. Hamilton, P. Goss, Locoregionally recurrent breast
cancer: incidence, risk factors and survival, Cancer. Treat. Rev. 27 (2001) 6782.
[6] A. El-Sherif, H.C. Fernando, R. Santos, B. Pettiford, J.D. Luketich, J.M. Close, R.J.
Landreneau, Margin and local recurrence after sublobar resection of non-small
cell lung cancer, Ann. Surg. Oncol. 14 (2007) 24002405.
[7] T.E.Read,M.G.Mutch,B.W.Chang,M.S.McNevin,J.W.Fleshman,E.H.Birnbaum,
R.D. Fry, P.F. Caushaj, I.J. Kodner, Locoregional recurrenceand survival after curative
resection of adenocarcinoma of the colon, J. Am. Coll. Surg. 195 (2002) 3340.
[8] Y. Moriya, Treatment strategy for locally recurrent rectal cancer, Jpn. J. Clin. Oncol.
36 (2006) 127131.
[9] E.A. Newman, D.M. Simeone, M.W. Mulholland, Adjuvant treatment strategies for
pancreatic cancer, J. Gastrointest. Surg. 10 (2006) 916926.
[10] F. Oshita, H. Saito, K. Yamada, K. Noda, Phase II study of paclitaxel and irinotecan
chemotherapy in patients with advanced nonsmall cell lung cancer, Am. J. Clin.
Oncol. 30 (2007) 358360.
[11] M. Kawahara, Irinotecan in the treatment of small cell lung cancer: a review of
patient safety considerations, Expert Opin. Drug Safety 5 (2006) 303312.
[12] J.B. Wolinsky, W.C. Ray III, Y.L. Colson, M.W. Grinstaff, Poly(carbonate-ester)s based
on units of 6-hydroxyhexanoic acid and glycerol, Macromolecules 40 (2007)
70657068.
[13] D.F. Chollet, L. Goumaz, A. Renard, G. Montay, L. Vernillet, V. Arnera, D.J. Mazzo,
Simultaneous determination of the lactone and carb oxylate forms of the
camptothecin derivative CPT-11 and its metabolite SN-38 in plasma by high-
performance liquid chromatography, J. Chromatogr., B, Biomed. Sci. Appl. 718
(1998) 163175.
[14] B. Ertl, P. Platzer, M. Wirth, F. Gabor, Poly(D,L-lactic-co-glycolic acid) microspheres
for sustaineddelivery and stabilizationof camptothecin,J. Control. Release 61 (1999)
305317.
[15] T.G. Burke, Z. Mi, E thyl sub stitu tion at the 7 posit ion ext ends th e half-l ife of
10-hydroxycamptothecin in the presence of human serum albumin, J. Med.
Chem. 36 (1993) 25802582.
[16] C.G. Pitt, M.M. Gratzl, A.R. Jeffcoat, R. Zweidinger, A. Schindler, Sustained drug
delivery systems II: factors affecting release rates from poly(epsilon-caprolactone)
and related biodegradable polyesters, J. Pharm. Sci. 68 (1979) 15341538.
[17] Y.H. Ping, H.C. Lee, J.Y. Lee, P.H. Wu, L.K. Ho, C.W. Chi, M.F. Lu, J.J. Wang, Anticancer
effects of low-dose 10-hydroxycamptothecin in human colon cancer, Oncol. Rep.
15 (2006) 12731279.
[18] A.Shenderova, T.G.Burke,S.P.Schwendeman,Stabilizationof 10-hydroxycamptothecin
in poly(lactide-co-glycolide) microsphere delivery vehicles, Pharm. Res. 14 (1997)
14061414.
[19] A. Hate, D. Knight, B. Amsden, A biodegradable injectable thermoplastic for
localized camptothecin delivery, J. Pharm. Sci. 93 (2004) 11951204.
[20] R.J. Landreneau, D.J. Sugarbaker, M.J. Mack, S.R. Hazelrigg, J.D. Luketich, L.
Fetterman, M.J. Liptay, S. Bartley, T.M. Boley, R.J. Keenan, P.F. Ferson, R.J. Weyant,
K.S. Naunheim, Wedge resection versus lobectomy for stage I (T1 N0 M0) non-
small-cell lung cancer, J. Thorac. Cardiovasc. Surg. 113 (1997) 691698 discussion
698700.
[21] F. Detterbeck, M.P. Rivera, M.A. Socinski, J. Rosenman, Diagnosis and Treatment of
Lung Cancer: An Evidence-Based Guide for the Pr acticing Physician, W. B.
Saunders Company, Philadelphia, 2001.
[22] N. Martini, M.S. Bains, M.E. Burt, M.F. Zakowski, P. McCormack, V.W. Rusch, R.J.
Ginsberg, Incidence of local recurrence and second primary tumors in resected
stage I lung cancer, J. Thorac. Cardiovasc. Surg. 109 (1995) 120129.
[23] T. Manabe, H. Okino, R. Maeyama, K. Mizumoto, M. Tanaka, T. Matsuda, New
infusion device for trans-tissue, sustained local delivery of anticancer agent to
surgically resected tissue: potential use for suppression of local recurrence of
pancreatic cancer, J. Biomed. Mater. Res. B. Appl. Biomater. 73 (2005) 203207.
[24] A.K. Azab, J. Kleinstern, V. Doviner, B. Orkin, M. Srebnik, A. Nissan, A. Rubinstein,
Prevention of tumor recurrence and distant metastasis formation in a breast
cancer mouse model by biodegradable implant of 131I-norcholesterol, J. Control.
Release 123 (2007) 116122.
[25] L. Zhang, Y. Hu, X. Jiang, C. Yang, W. Lu, Y.H. Yang, Camptothecin derivative-loaded
poly(caprolactone-co-lactide)-b-PEG-b-poly(caprolactone-co-lactide) nanoparticles
and their biodistribution in mice, J. Control. Release 96 (2004) 135148.
[26] L. Zhang, M. Yang, Q. Wang, Y. Li, R. Guo, X. Jiang, C. Yang, B. Liu, 10-
Hydroxycamptothecin loaded nanoparticles: preparation and antitumor activity
in mice, J. Control. Release 119 (2007) 153162.
[27] J. Wang, R. Wang, L.B. Li, Preparation and properties of hydroxycamptothecin-
loaded nanoparticles made of amphiphilic copolymer and normal polymer,
J. Colloid Interface Sci. 336 (2009) 808813.
[28] A. Shenderova, T.G. Burke, S.P. Schwendeman, The acidic microclimate in poly
(lactide-co-glycolide) microspheres stabilizes camptothecins,Pharm. Res. 16 (1999)
241248.
[29] C. Zhang, Y. Ding, L.L. Yu, Q. Ping, Polymeric micelle systems of hydroxycamp-
tothecin based on amphiphilic N-alkyl-N-trimethyl chitosan derivatives, Colloids
Surf., B Biointerfaces 55 (2007) 192199.
[30] X. Yang, L. Li, Y. Wang, Y. Tan, Preparation, pharmacokinetics and tissue
distribution of micelles made of reverse thermo-responsive polymers, Int. J.
Pharm. 370 (2009) 210215.
[31]M.Hong,S.Zhu,Y.Jiang,G.Tang,Y.Pei,Efcient tumor targeting of
hydroxycamptothecin loaded PEGylated niosomes modied with transferrin,
J. Control. Release 133 (2009) 96102.
[32] H. Brem, M.S. Mahaley Jr., N.A. Vick, K.L. Black, S.C. Schold Jr., P.C. Burger, A.H.
Friedman,I.S. Ciric,T.W. Eller, J.W.Cozzens, etal., Interstitialchemotherapywith drug
polymer implants for the treatment of recurrent gliomas, J. Neurosurg. 74 (1991)
441446.
[33] H. Brem, S. Piantadosi, P.C. Burger, M. Walker, R. Selker,N.A. Vick, K. Black, M.Sisti, S.
Brem, G. Mohr, et al., Placebo-controlled trial of safety and efcacy of intraoperative
controlled delivery by biodegradable polymers of chemotherapy for recurrent
gliomas.The Polymer-brainTumor TreatmentGroup, Lancet345 (1995) 10081012.
[34] H. Brem, Polymers to treat brain tumours, Biomaterials 11 (1990) 699701.
[35] S.E. Shackney, G.W. McCormack, G.J. Cuchural Jr., Growth rate patterns of solid
tumors and their relation to responsiveness to therapy: an analytical review, Ann.
Intern. Med. 89 (1978) 107121.
[36] V.T. DeVita, T.S. Lawrence, S.A. Rosenberg, DeVita, Hellman, and Rosenberg's
Cancer: Principles and Practice of Oncology, Lippincott Williams & Wilkins, 2008.
[37] W.H. Wolberg, F.J. Anseld, The relation of thymidine labeling index in human
tumors in vitro to the effectiveness of 5-uorouracil chemotherapy, Cancer Res.
31 (1971) 448450.
[38] J.S. Meyer, Cell kinetic measurements of human tumors, Human Pathol. 13 (1982)
874877.
287J.B. Wolinsky et al. / Journal of Controlled Release 144 (2010) 280287
... An in vitro study to decrease local recurrences in a murine model of Lewis lung carcinoma using biocompatible poly(glycerol monostearate-co-e-caprolactone) polymer film-loaded paclitaxel (PTX) implants was conducted by Liu et al. [33]. Wolinsky et al. utilized a tunable drug-eluting polymeric delivery platform made up of poly(glycerol monostearate-co-εcaprolactone) films loaded with anticancer agent 10-hydroxycamptothecin (HCPT) [34]. The film formed a flexible composite when applied to a collagen-based scaffold clinically indicated for the mechanical reinforcement of lung tissue. ...
... Osmotic system-based implantable device Prostate cancer [30] β-lapachone-containing polymer implants (millirods) Prostate cancer [31] Magnetically controlled drug delivery MEMS device Prostate cancer [32] Biocompatible poly(glycerol monostearate-co-e-caprolactone) polymer film-loaded PTX Lewis lung carcinoma [33] Polymeric delivery platform made up of poly(glycerol monostearate-co-ε-caprolactone) films loaded with HCPT Lewis lung carcinoma [34] Polyester nanofibers of the PLGA-PLA-PCL nanofiber implant Brain gliomas [35] Smart hyperthermia nanofibrous scaffolds consisting of the N-isopropylacrylamide and N-hydroxymethylacrylamide polymers Skin cancer [38] PTX-paclitaxel; HCPT-10-hydroxycamptothecin. ...
Article
Full-text available
Breast cancer is one of the leading causes of death in the female population worldwide. Standard treatments such as chemotherapy show noticeable results. However, along with killing cancer cells, it causes systemic toxicity and apoptosis of the nearby healthy cells, therefore patients must endure side effects during the treatment process. Implantable drug delivery devices that enhance therapeutic efficacy by allowing localized therapy with programmed or controlled drug release can overcome the shortcomings of conventional treatments. An implantable device can be composed of biopolymer materials, nanocomposite materials, or a combination of both. This review summarizes the recent research and current state-of-the art in these types of implantable devices and gives perspective for future directions.
... Previous studies have found that hydroxycamptothecin is basically not absorbed by the bladder mucosa. It inhibits DNA replication, transcription, and mitosis by acting on DNA topoisomerase I and has an inhibitory effect on the development of tumors [27,28]. e adverse reactions are relatively small, and the patient's recurrence rate is low, which is better than the preventive effect of mitomycin and BCG. ...
Article
Full-text available
Objective: To observe the clinical effect of Xiaozheng Decoction combined with bladder perfusion with hydroxycamptothecin in the treatment of bladder cancer. Methods: A total of 92 bladder cancer patients admitted to our hospital from January to December 2018 were selected and divided into an observation group and a control group according to the random number table method, with 46 cases in each group. The observation group was given Xiaozheng Decoction combined with bladder perfusion with hydroxycamptothecin, and the control group was given hydroxycamptothecin. The levels of serum-related factors (intercellular adhesion molecule-1 (ICAM-1), E-cadherin, cell adhesion molecules (CAM), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF)), white blood cell (WBC) level, immune function indexes, short-term total response rate, and incidence of adverse reactions were compared between the two groups before and after treatment. Results: After 2 years of postoperative treatment, the levels of ICAM-1, E-cadherin, CAM-1, FGF, and VEGF (a, b, c) in both groups were improved compared with those before treatment and the observation group was better than the control group (p < 0.01). The number of WBCs was significantly higher than in the control group after Traditional Chinese Medicine (TCM) treatment. The observation group was better than the control group in increasing CD3+ and CD4+ levels and decreasing CD8+ level (p < 0.05), indicating that this prescription could improve the immune function of patients. The recurrence rate in the observation group was 6.52% after 2 years of treatment, lower than 17.39% in the control group. Color ultrasound parameters showed that there were no statistically significant differences in arrive time (AT) and time to peak (TTP) between patients with and without recurrence and peak intensity (PI) and washout time (WT) were higher in patients with recurrence than in patients without recurrence (p < 0.01). The incidence of adverse reactions was significantly lower than that of the control group (p < 0.01). Conclusion: The clinical effect of Xiaozheng Decoction combined with hydroxycamptothecin on the treatment of bladder cancer was clear and superior to that of hydroxycamptothecin, which could effectively improve the serological indicators of patients with a low incidence of adverse reactions and prolong the survival cycle of patients. Therefore, it is worthy of promotion and application.
Article
Prodrug assembled nanoparticles integrate the merits of both prodrug and nanoparticle, which significantly improve pharmacokinetic parameters, enhance tumorous accumulation and decrease adverse effects, while they are challenged by disassembly upon dilution in blood, masking the superiority of nanoparticles (NPs). Herein, a reversibly "double locked" hydroxycamptothecin (HCPT) prodrug nanoparticle decorated with cyclic RGD peptide (cRGD) is developed for safe and efficient chemotherapy of orthotopic lung cancer in mice. HCPT prodrug is constructed from acetal (ace)-linked cRGD-PEG-ace-HCPT-ace-acrylate polymer, which is self-assembled into the nanoparticles with "the first lock" of HCPT. Then the nanoparticles undergo the in situ UV-crosslinking of the acrylate residues for constructing "the second lock" of HCPT. The obtained "double locked" nanoparticles (T-DLHN) with simple and well-defined construction are demonstrated to possess extremely high stability against 100-fold dilution and acid-triggered "unlock" including de-crosslinking and liberation of the pristine HCPT. In an orthotopic lung tumor of mouse model, T-DLHN reveals a prolonged circulation time of about 5.0 h, superb lung tumor-homing capacity with tumorous drug uptake of about 7.15%ID/g, resulting in significantly boosted anti-tumor activity and reduced adverse effects. Hence, these nanoparticles utilizing "double lock" and acid-triggered "unlock" strategies represent a unique and promising nanoplatform for safe and efficient drug delivery. STATEMENT OF SIGNIFICANCE: : Prodrug assembled nanoparticles have the unique properties of the well-defined structure, systemic stability, improved pharmacokinetics, passive targeting and decreased adverse effects. However, prodrug assembled NPs would disassemble against extensive dilution in the blood circulation when intravenously injected into the body. Herein, we have designed a cRGD-directed reversibly "double-locked" HCPT prodrug nanoparticle (T-DLHN) for safe and efficient chemotherapy of orthotopic A549 human lung tumor xenografts. Upon intravenous injection, T-DLHN can overcome the shortcoming of disassembly against extensive dilution, prolong the circulation time due to the "double locked" configuration and then mediate targeted drug delivery into the tumors. After uptaken into the cells, T-DLHN undergoes concurrent de-crosslinking and liberation of HCPT under acidic condition for enhanced chemotherapeutic efficacy with negligible adverse effects.
Article
Biomaterials are developed to aid a variety of regenerative medicine strategies, such as providing a framework for cell adhesion and proliferation or serving as carriers of bioactive factors, while stem cells are increasingly implanted in biomaterial scaffolds to improve therapeutic efficacy. Advanced biomaterials like metals, synthetic polymers, and ceramics are used in bone regeneration technology. The ultimate goal of biomaterial-directed SC (stem cells) culture is to replicate the physical and biochemical characteristics of the physiological SC niche. The primary structural component of tumour ECM (extracellular matrix) is collagen. Cancer initiation, EMT (epithelial-mesenchymal transition), drug resistance, and CSC (cancer stem cells) self-renewal have all been linked to collagen subtypes. The enhancement of liver CSCs has already been investigated using collagen I-based platforms. Alginate and chitosan are two naturally occurring polymers with biological macromolecules that are similar. Biomaterial-based therapies, on the whole, offer incredible versatility and tailorability in the fight against the disease. They could also be used as tissue-engineered scaffolds for immune cell replenishment, potentially making them a key weapon in the next generation of therapeutic approaches.
Article
Full-text available
Nanofibers and hydrogels are two forms of biomaterial scaffolds that have different properties and different applications. Polysaccharides have been used for the preparation of both scaffolds due to their biocompatibility and biodegradability. In this study, our main objective was to compare the tumor cell growth and tumor reduction potential of drug‐loaded scaffolds. This was achieved using different methods for cell proliferation, epithelial–mesenchymal Transition studies, and biomarker expression‐related assays. Anticancer drug doxorubicin was loaded to the scaffolds using adsorption and in vitro and in vivo tumor reduction was also performed. Results showed that both scaffolds supported the growth of cancer cells but did not promote epithelial‐mesenchymal transition. A significant difference in drug release was observed and this may have influenced the in vivo tumor reduction. A fast release of the drug and higher tumor‐reducing potential was observed for hydrogel. The studies confirmed that different types of scaffolds have different properties. Biomedical applications related to the structure can be dissimilar in different scenarios.
Article
Diseases related to the lungs are among the most prevalent medical problems threatening human life. The treatment options and therapeutics available for these diseases are hindered by inadequate drug concentrations at pathological sites, a dearth of cell-specific targeting and different biological barriers in the alveoli or conducting airways. Nanostructured delivery systems for lung drug delivery have been significant in addressing these issues. The strategies used include surface engineering by altering the material structure or incorporation of specific ligands to reach prespecified targets. The unique characteristics of nanoparticles, such as controlled size and distribution, surface functional groups and therapeutic release triggering capabilities, are tailored to specific requirements to overcome the major therapeutic barriers in pulmonary diseases. In the present review, the authors intend to deliver significant up-to-date research in nanostructured therapies in inflammatory lung diseases with an emphasis on biocomposite-based nanoparticles.
Article
Brain metastases are intracranial recurrence of extracranial malignant tumors with a high incidence and poor prognosis. Owing to the particularity of intracranial localization, clinical diagnosis (neuroimaging and biopsy) of brain metastases is associated with shortcomings such as delayed diagnosis and biopsy invasiveness. Clinical treatment modalities consist of local therapies (resection and radiotherapy) and systemic therapies (chemotherapy, targeted therapy and immunotherapy). However, the efficiency of clinical therapies is severely hindered by 1) neurological impairment, 2) relapse, 3) blood-brain barrier, and 4) therapy resistance. Nanomedicine possesses great potentials in combating these clinical issues. This article reviewed backgrounds and recent advances of applying nanomedicine to address these above issues in clinical diagnosis and treatment of brain metastases.
Article
Full-text available
In globally, cancer is a second leading disease next to cardiovascular diseases in non-communicable diseases, which affect the all ages, sex, social status, ethnicity and primary cause of illness related death. Traditionally, systemic delivery drug systems like chemotherapy via oral capsule, injections of nanoparticles/micro particles, immunotherapy and others, which can inhibit or halt the progression of tumors. The short half-life of drugs which cannot achieve the targeted dose level to the tumor site and not able to target desired cell and commonly produces the organ toxicity. Recently, researchers have been attempting to direct delivery agents for cancer therapy. One of the best methods is a local therapy system, which deliver the drug directly via implantable procedure and it’s achieved the maximum concentration of the desire drug at the tumor site, non-target systemic exposure and minimize the organ toxicity to the patients. Biomaterial implants are widely used in the local concurrent delivery of chemotherapy and anti-angiogenic agents, local delivery of poly-chemotherapy, gene therapy as an alternative to drug delivery, scaffolds for cancer immunotherapy and polymer-based composites of drug molecules. There are different types of polymers like poly anhydride poly [bis (p-carboxy-phenoxy) propane-sebacic acid] copolymer [p(CPP:SA)], fatty acid dimer-sebacic acid copolymer (FAD-SA), poly (lactic-co-glycolic acid) copolymer (PLGA), poly (ε-caprolactone) (PCL), poly (glycerol monostearate-co-caprolactone), alginate and silica, used in successively cancer therapy. In order to minimize the risk of unwanted side effect of different types of biomaterials implants, it’s biocompatible to reduce the ability to elicit the inflammatory effect to the implanted area or the site. Therefore, the key role of choosing the appropriate and biocompatible implants to particular therapy is an indispensable. This should be validated with respect to risk benefit ratio in case of cancers. Biomaterial based implant local delivery systems provide more versatile and tailorable approach to against treatment of different types of the cancer.
Article
Full-text available
The synthesis of biodegradable copolymers based on the units of 6-Hydroxyhexanoic acid and glycerol is presented. ε-Caprolactone was copolymerized with 5-benzyloxy-1,3- dioxan-2-one to form a copolymer that afforded a secondary hydroxyl pendant side groups with the capacity for subsequent functionalization after the removal of the benzyl side chains from the glycerol carbonate units via a mild hydrogenolysis reaction. Copolymers were synthesized from the varying mole ratios of the two monomers, ε-Caprolactone and 5-benzyloxy-1,3-dioxan-2-one, and molecular weight measurements were obtained using size exclusion chromatography and thermal data was recorded using differential scanning calorimetry. The relative mole fractions of the monomers in the resulting co-polymers were determined using 1H NMR analysis. The results show that the copolymers possess a great glass transition temperature and a mean particle diameter of 125 nm with a narrow polydispersity index.
Book
DeVita, Hellman, and Rosenberg's Cancer: Principles & Practice of Oncology, 10th edition has garnered universal acclaim as the world's definitive, standard-setting oncology reference. More than 400 respected luminaries explore today's most effective strategies for managing every type of cancer by stage of presentation - discussing the role of all appropriate therapeutic modalities as well as combined-modality treatments. This multidisciplinary approach will help your cancer team collaboratively face the toughest clinical challenges and provide the best possible care for every cancer patient. Access the complete contents online or on your mobile device, with quarterly updates reflecting late-breaking developments in cancer care on LWW Health Library. Take full advantage of the latest advances with brand-new chapters on Hallmarks of Cancer, Molecular Methods in Cancer, Oncogenic Viruses, Cancer Screening, and new sections on Genetic testing and counseling for cancer, plus comprehensive updates throughout - including coverage of the newest biologic therapies. Make optimal, well-coordinated use of all appropriate therapies with balanced, multidisciplinary advice from a surgeon, a medical oncologist, and a radiation oncologist in each major treatment chapter. Review the latest molecular biology knowledge for each type of cancer and its implications for improved management. Make the best decisions on cancer screening and prevention, palliative care, supportive oncology, and quality-of-life issues. Access and receive quarterly content updates for Cancer: Principles and Practice, 10th Edition on the NEW and ENHANCED LWW Health Library while on the go. Additionally, for the first 6 months, Cancer: Principles and Practice of Oncology, 10th Edition purchasers will get access to the entire Oncology Collection on LWW Health Library - an incredible value at no additional charge!. © 2011 by Wolters Kluwer Health / Lippincott Williams & Wilkins. All rights reserved.
Article
Chemotherapy for brain tumours has been limited because of difficulty in achieving adequate exposure to the tumour without systemic toxicity. We have developed a method for local sustained release of chemotherapeutic agents by their incorporation into biodegradable polymers. Implantation of the drug-impregnated polymer at the tumour site allows prolonged local exposure with minimal systemic exposure. We conducted a randomised, placebo-controlled, prospective study to evaluate the effectiveness of biodegradable polymers impregnated with carmustine to treat recurrent malignant gliomas. In 27 medical centres, 222 patients with recurrent malignant brain tumours requiring re-operation were randomly assigned to receive surgically implanted biodegradable polymer discs with or without 3·85% carmustine. Randomisation balanced the treatment groups for all of the prognostic factors examined. Median survival of the 110 patients who received carmustine polymers was 31 weeks compared with 23 weeks for the 112 patients who received only placebo polymers (hazard ratio=0·67, p=0·006, after accounting for the effects of prognostic factors). Among patients with glioblastoma, 6-month survival in those treated with carmustine-polymer discs was 50% greater than in those treated with placebo (mortality=32 of 72 [44%] vs 47 of 73 [64%], p=0·02). There were no clinically important adverse reactions related to the carmustine polymer, either in the brain or systemically. Interstitial chemotherapy delivered with polymers directly to brain tumours at the time of surgery seems to be a safe and effective treatment for recurrent malignant gliomas
Article
The release rates of several steroids from films and capsules of homopolymers and copolymers of ε-caprolactone, DL-lactic acid, and glycolic acid were measured in vitro and in vivo for up to 200 days. Relatively constant release rates from capsules (reservoir devices) were observed only under certain conditions. Factors that influence the drug release kinetics were evaluated. Release from poly(ε-caprolactone) and poly(ε-caprolactone-co-DL-lactic acid) was diffusion controlled. Release from poly(DL-lactic acid-co-glycolic acid) was associated with polymer degradation. Release from poly(DL-lactic acid) was very slow when diffusion controlled.
Article
Cancer chemotherapy is not always effective. Difficulties in drug delivery to the tumor, drug toxicity to normal tissues, and drug stability in the body contribute to this problem. Polymeric materials provide an alternate means for delivering chemotherapeutic agents. When anticancer drugs are encapsulated in polymers, they can be protected from degradation. Implanted polymeric pellets or injected microspheres localize therapy to specific anatomic sites, providing a continuous sustained release of anticancer drugs while minimizing systemic exposure. In certain cases, polymeric microspheres delivered intravascularly can be targeted to specific organs or tumors. This article reviews the principles of chemotherapy using polymer implants and injectable microspheres, and summarizes recent preclinical and clinical studies of this new technology for treating cancer.
Article
10-Hydroxycamptothecin (HCPT) loaded nanoparticles made from poly(caprolactone-co-lactide)-b-PEG-b-poly(caprolactone-co-lactide) (PCLLA-PEG-PCLLA) block copolymer, were prepared by a novel two-step nanoprecipitation method using an interior-chemistry strategy. The satisfactory drug loading content (>13%) as well as high encapsulation efficiency (>85%) was achieved. Cytotoxicity test indicated that the HCPT-loaded nanoparticles had enhanced in vitro cytotoxicity compared to free drug. Progressively, in vivo antitumor activity and HCPT biodistribution in sarcoma-180 (S-180) bearing mice after intravenous injection of the HCPT-loaded nanoparticles show that HCPT-loaded nanoparticles exhibited superior in vivo antitumor effect and remarkably different biodistribution than the commercially available HCPT injection.
Article
Stabilized micelle structure nanoparticles were prepared using Pluronic F127 and poly(butylcyanoacrylate) (PBCA). To increase the drug loading of nanoparticles, D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) was additionally included into the nanoparticle composition. The poorly soluble anticancer drug 10-hydroxycamptothecin (HCPT) was used as a model drug and incorporated into nanoparticles. The results obtained from FT-IR and DSC confirmed that HCPT was molecularly dispersed in nanoparticles and no chemical reaction occurred. The size of the nanoparticles measured by DLS demonstrated that the size distribution was narrow and the average diameter was less than 200 nm. The morphology of the nanoparticles observed by TEM indicated that the nanoparticles exhibited a smooth surface and distinct spherical shape. In vitro release experiments indicated that the HCPT-loaded nanoparticles showed sustained release profiles. The results of a drug loading test revealed that adding TPGS could increase the drug loading. The drug loading of stabilized micelle structure nanoparticles with 70% of TPGS was about 0.0425+/-0.0011% w/w compared to 0.0254+/-0.0008% w/w found for the nanoparticles without TPGS. The results of CMC value tests showed that the CMC values of the stabilized nanoparticles were approximately 10-fold lower than those of the nonstabilized micelles (from 2.0x10(-5) to 2.5x10(-4) M). Cytotoxicity tests showed that the cytotoxicity of HCPT-loaded nanoparticles against cancer cells in vitro was remarkably higher than that of free drugs. 10-Hydroxycamptothecin-loaded nanoparticles may serve as a stable delivery system for poorly soluble HCPT.
Article
New reverse thermo-responsive polymers, poly(ethylene oxide)-poly(propylene oxide) multiblock copolymers (poly(ether-carbonate)s) were synthesized. The micelles made of new reverse thermo-responsive polymers were also prepared loaded with the poorly soluble anticancer drug, hydroxycamptothecin (HCPT). The structure characterization of poly(ether-carbonate)s was determined by (1)H NMR and FT-IR analysis. The critical micelle concentration (CMC), critical micelle temperature (CMT), size distribution and drug release in vitro were determined. The pharmacokinetics and tissue distribution in vivo for novel copolymer micelles were studied. The experimental results showed that the micelles was spherical in appearance and dispersed well. The process of HCPT release from micelles in vitro was composed of two steps, abrupt release and sustained release. After i.v. administration (2h), the drug concentration of poly(ether-carbonate) micelles group in liver in mice was 3.46microg/g, while that of HCPT injection group was 0.401microg/g. Compared with HCPT injection, the elimination half-life of poly(ether-carbonate) micelles group was prolonged remarkably from 1.3 to 12.5h. The poly(ether-carbonate) micelles showed a combination of liver targeting and sustained drug release in experiments on animals.
Article
The aim of the present report was to exploit the possibility of combination of the stealth action by polyethylene glycol cyanoacrylate-co-hexadecyl cyanoacrylate (PEG-PHDCA) modified niosomes and active targeting function of transferrin (Tf) by transferrin receptor-mediated endocytosis to promote drug delivery to solid tumor following intravenous administration with hydroxycamptothecin (HCPT) as model drug. HCPT-loaded PEG-niosomes (PEG-NS) were prepared by thin-film hydration and ultrasound method; the periodate-oxidated Tf was coupled to terminal amino group of PEG to produce the active targeting vesicles with average diameters of 116 nm. The uptake of Tf-PEG-NS into KB cells was concentration and time dependent, which could be inhibited by low temperature and free Tf, indicating that the endocytosis process was energy-driven and receptor specific. Compared with HCPT injection, non-stealth niosomes and PEG-NS, Tf-PEG-NS demonstrated the strongest cytotoxicity to three carcinomatous cell lines (KB, K562 and S180 cells), the greatest intracellular uptake especially in nuclei, the highest tumor concentration and largest area under the intratumoral hydroxycamptothecin concentration curve, as well as the most powerful anti-tumor activity with the inhibition rate of 71% against S180 tumor in mice. The results showed that the transferrin modified PEGylated niosomes could be one of the promising solutions to the delivery of anti-tumor drugs to tumor.