Breast cancer brain metastasis, which occurs in more than one third of metastatic patients with HER2+ tumors, is extremely difficult to treat with survival rates of <1 year. Even though aberrant HER2 receptor overexpression in breast cancer is highly correlated with brain metastasis, molecularly targeted anti-HER2 therapy such as trastuzumab and lapatinib cannot cross the blood brain barrier (BBB) to reach the metastatic site at therapeutic concentrations. This proposal addresses overcoming a critical barrier to delivering the 'molecularly targeted' anti-HER2 drug lapatinib to a specific metastatic site - the brain. Our approach exploits the unique properties of brain homing peptide Angiopep-2 to improve penetration of lapatinib across the BBB to treat HER2+ brain metastasis. The 19-amino acid peptide Angiopep-2 binds to the low-density lipoprotein receptor-related protein (LRP-1) receptor, which is transcytosed into the brain with a much higher BBB transcytosis efficiency than transferrin. The goal of this research is to develop brain permeable lapatinib Angiopep-2 conjugates that can enhance therapeutic drug levels in the brain to treat HER2+ breast cancer brain metastasis. We hypothesize that lapatinib-Angiopep-2 conjugates will cross the BBB more efficiently than lapatinib itself and lead to higher lapatinib accumulation in the brain metastasis consequently, therapeutic benefits will be obtained in targeting and destroying HER2+ breast cancer brain metastasis. We plan to test the hypothesis through three specific aims: (1) optimize lapatinib-Angiopep conjugates with improved solubility, serum stability, and ability to achieve localized lapatinib release; (2) demonstrate the effectiveness of brain penetration, cellular uptake and release, and cytotoxic potency of the optimized conjugates; (3) validate enhanced brain accumulation and therapeutic efficacy of optimized conjugates in HER2-overexpressing breast cancer brain metastasis models in immunodeficient mice. We will optimize synthetic protocols to chemically attach the lapatinib to the peptide backbone of Angiopep-2 via different cross-linkers. We will study the stability of conjugates in mouse and human serum and cytotoxicity in the HER2-overexpressing breast cancer cells. We will determine the BBB transport rates of the conjugates using in situ mouse brain perfusion technique. We will test the efficacy of conjugates in our established breast cancer brain metastasis mouse models by intracardiac and intracranial injection of brain-seeking HER2 overexpressing human breast cancer cells into nude mice, and monitoring tumor growth and/or regression after conjugate treatment. Successful completion of this project will identify a lapatinib delivery system that is capable of crossing the BBB and is active against breast cancer brain metastases animal models. If successful, the research can be translatable into the clinic and will potentially lead t a major step forward in extending the lives of breast cancer patients with HER2 positive brain metastases. This work will have a broader impact upon the delivery of other molecular targeted tyrosine kinase inhibitors such as gefitinib and erlotinib, structural analogs of lapatinib, for th treatment of CNS metastases originating from other cancers.

Public Health Relevance

Women with HER2-positive breast cancer are more likely to develop brain metastases; a devastating clinical problem occurring in approximately one third (25-34%) of women with metastatic breast cancer. Unfortunately; no FDA approved therapeutic regimen is available. This proposal addresses overcoming a critical barrier to delivering the ''molecularly targeted'' anti-HER2 drug lapatinib to a specific metastatic site - the brain. Our approach seeks to exploit the unique properties of the brain homing peptide Angiopep-2 to improve penetration of lapatinib across the BBB to brain cancer metastasis. The proposed work is directly relevant to the NIH's mission to enhance health; lengthen life; and reduce the burdens of illness and disability. The proposed research will fill the existing gap of organ specific delivry issues of molecularly targeted anti-HER2 therapy. If successful; this work can be readily translatable into the clinic and we have the potential to introduce to clinicians a novel therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Academic Research Enhancement Awards (AREA) (R15)
Project #
7R15CA182769-02
Application #
9380207
Study Section
Special Emphasis Panel (ZRG1-OTC-N (80)A)
Program Officer
Forry, Suzanne L
Project Start
2014-12-16
Project End
2017-11-30
Budget Start
2016-09-01
Budget End
2017-11-30
Support Year
2
Fiscal Year
2015
Total Cost
$370,110
Indirect Cost
$124,190
Name
University of Houston
Department
Pharmacology
Type
Schools of Pharmacy
DUNS #
036837920
City
Houston
State
TX
Country
United States
Zip Code
77204
Liu, Xinli (2016) Bone site-specific delivery of siRNA. J Biomed Res 30:264-71