Download Cancer Graduate Category:

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Graduate
Category: Interdisciplinary Topics, Centers and Institutes
Degree Level: PhD
Abstract ID# 1361
PARP Inhibitor Nanotherapy for Cancer Treatment
Paige Baldwin1, Anders Ohman2, Anne van de Ven3, Daniela Dinulescu2, Srinivas Sridhar3
1. Bioengineering, Northeastern University, 2. Brigham and Women’s Hospital, 3. Nanomedicine Science and Technology Center, Northeastern University
Abstract
Poly(ADP-ribose) Polymerase (PARP) plays an important role in a number of DNA
repair pathways. PARP inhibitors, such as Olaparib exploit the concept of synthetic
lethality by selectively targeting cancer cells with defective DNA repair pathways,
while leaving healthy cells with multiple repair pathways unharmed. These drugs are
currently only available in oral form which results in limited bioavailability, poor
tumor accumulation, and systemic toxicity. Nanoparticle formulations of Olaparib
were developed to allow intravenous (IV) or intraperitoneal (IP) delivery, providing
greater bioavailability and tumor accumulation, while limiting systemic toxicities.
NanoOlaparib was synthesized and characterized before further testing in vitro and
in vivo. In vitro it has been tested in a panel of ovarian cancer cell lines to elucidate
sensitivity profiles. NanoOlaparib was also tested in an ovarian cancer IP spread
model. Animals were treated IP with NanoOlaparib alone, and in combination with
cisplatin. Bioluminescence imaging illustrated that NanoOlaparib administered IP
daily resulted in a greater inhibition of tumor growth than those treated with oral
Olaparib daily. Radiosensitization with NanoOlaparib was tested in a radiation
resistant prostate cancer cell line, FK01 and a xenograft model with the same cells to
mimic castration resistant prostate cancer. The FK01 xenografts are highly
radioresistant with little difference between untreated and radiation only animals.
NanoOlaparib delays tumor growth, while the combination of radiation and
NanoOlaparib shrinks tumors. These results show that NanoOlaparib amplifies the
therapeutic efficacy of PARP inhibition and imply a promising role for the
nanoformulation in a variety of cancers.
Characterization
(a)
(b)
Prostate Cancer
(c)
(a)
Figure 1. NanoOlaparib characterization via (a) transmission electron micrograph, (b)
nanoparticle tracking laser scattering size distribution, and (c) release profile.
Average diameter is 71.7 nm and formulation can be loaded up to 11.52 mM.
(a)
Ovarian Cancer
(b)
Background
Figure 4. Subcutaneous engraftment of FK01 cells, derived from Ptenpc-/-;Trp53pc-/mice. Animals were treated intravenously with 40 mg/kg NanoOlaparib twice
weekly. Animals received a single dose of 10 Gy radiation on day 10. Fold change in
tumor volume (a) and a Kaplan meier survival curve depicting the lifespan of each
group (b).
Conclusions
(c)
(d)
Objective
Develop and characterize nanoparticle formulations (NanoOlaparib) of the PARP
inhibitor Olaparib to enhance delivery to the tumor site. The nanoformulation allows
for intravenous delivery and 100% availability in the vasculature or intraperitoneal
delivery for cancers such as ovarian cancer in which the tumor is disseminated
throughout the IP cavity.
(b)
Figure 2. Dose response curves for (a) Olaparib and (b) NanoOlaparib and (c)
comparison of IC50 values. Chart (d) displays genomic profiles of tested cell lines.
In vivo
(a)
(b)
(c)
• PARP-1 is a DNA repair
protein which plays a role in:
• Base Excision Repair
• Homologous
Recombination
• Non-Homologous End
Joining
• Transcriptional
Regulation
• PA1 is most sensitive to NanoOlaparib which may be attributed to genetic
instability at 11/13 polymorphic loci, each containing (CA)¬n microsatellites
possibly causing mutations in critical genes with coding repeat sequences.
• The high sensitivity of 4412 and 4306 cell lines suggests that PTEN deletion
confers similar sensitivity to PARP inhibitors as BRCA2 deletion. The results
indicate that BRCA mutations and deletions are just as susceptible as PTEN
deletions while high genetic instability showing the greatest sensitivity.
• NanoOlaparib is highly potent, when delivered IP effectively treats the tumor,
leading to 50% reduction in tumor volume.
• Toxicity can be attributed to 100% of the dose being delivered directly to the
peritoneal cavity with a sustained release, therefore, overdosing the mice with
the daily administration, leading to toxicity (death).
• Dosing needs to be tailored to the pharmacokinetics of the nanoformulation
rather than the oral formulation, to determine the optimal dose regime.
• The FK01 xenografts are highly radioresistant with little difference between
untreated and radiation only animals.
• NanoOlaparib as a monotherapy delays tumor growth but does not shrink
tumors.
• The combination of Radiation and NanoOlaparib clearly shrinks tumor growth.
Some animals show very small tumors, while tumors seem to regrow in others.
Future Directions
• Determination of the optimal dosing regime for the nanoformulations.
• Characterization of a nanoformulation for a more potent PARP inhibitor,
Talazoparib.
• Development of a combination nanoplatform for delivery of a PARP inhibitor and
a DNA damaging agent such as cisplatin.
(d)
(e)
Oral Formulation Nanoformulation
Unprotected,
Protected
undergoes first pass nanocarrier, evades
metabolized
first pass metabolism
~10% bioavailability ~100% Bioavailability
Circulation half-life 6- Circulation half-life
7 hours
24-48 hours
Acknowledgements
Supported by IGERT grant NSF-DGE- 0965843, Army W81XWH-14-1-0092and Mazzone
Foundation.
References
Figure 3. IP spread model using 404 cells derived from BRCA2-/-, TP53-/-, PTEN-/GEMMs. Animals were treated with (a) empty nanoparticles (b) 50 mg/kg
NanoOlaparib IP or (c) 50 mg/kg oral Olaparib daily. Average fold change in
bioluminescence during treatment (d) and Kaplan meier survival curve (e).
1. Perets, R., Wyant, G. a, Muto, K., et al. (2013). Transformation of the fallopian tube
secretory epithelium leads to high-grade serous ovarian cancer in Brca;Tp53;Pten models.
Cancer Cell, 24(6), 751–65.
2. C.J. Lord and A. Ashworth (2013) Mechanisms of resistance to therapies targeting BRCAmutant cancers., Nature medicine. 19, 1381–8.
3. Nature Reviews Drug Discovery 2, 347-360 (May 2003) doi:10.1038/nrd1088
4. Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, et al. Crucial role of p53dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature.
2005;436:725-30.