Neuroblastoma is the most common extra-cranial solid tumor of childhood with an appalling 30% cure rate in children with advanced disease. There is a clear need for new chemotherapeutics, as current drugs are only marginally effective at the high doses that result in toxic acute and grave long term side effects. The overall objective of this proposal is to employ modern techniques of protein engineering to develop a new generation of non-immunogenic and pharmacologically optimized enzymes for chemotherapy of neuroblastomas and other central nervous system (CNS) cancers through L-Methionine (L-Met) depletion. L-Met is required not only for protein synthesis but also as the precursor for methylation reductions and for the biosynthesis of polyamines. Tumors have a much greater requirement for L-Met than normal tissues and become apoptotic when its availability is restricted. i.v. administration of bacterial (Pseudomonas) methionine-g-lyase is able to mediated near complete depletion of L-Met in serum and has been shown to drastically inhibit tumor growth of neuroblastomas, glioblastomas and prostate carcinomas in mouse xeongrafts. Furthermore strong synergistic effects with microtubule depolymerization agents have been reported. Unfortunately, in clinical trials the bacterial enzyme was shown to have very poor pharmacological properties (t 1/2 in serum only 2 hrs) and was found to be highly immunogenic in primates eliciting severe adverse responses that resulted in anaphylactic shock and death. While the human genome does not encode any methionine lyase enzymes, in preliminary studies we deployed protein engineering strategies to generate potentially non-immunogenic variants of the human enzyme cystathionine-g-lyase that: (a) exhibit high L-Met degradation activity in vitro and in vivo, (b) display a lower IC50 for neuroblastoma cell lines than their bacterial counterparts and (c) are about 10-fold more stable in mice. Here we will employ structure guided mutagenesis and directed evolution strategies to: 1. Engineer catalytically optimized """"""""human L-methioninases"""""""" i.e. cystathionine-g-lyase enzymes with very high activity for L-Met degradation, even better stability in serum and high selectivity. 2. Develop optimized formats of the """"""""human L-methioninases"""""""" for prolonged persistence in circulation by either site-specific PEGylation or by fusion to long intrinsically disordered polypeptide sequences (XTEN) and determine their pharmacokinetic and pharmacodynamic properties. 3. Evaluate the efficacy of these enzymes in the mouse xenograft model of human neuroblastoma tumors formed using clinical cell lines established either in diagnosis or relapse. The utility of these enzymes will be investigated both as monotherapy and in combination therapy with vincristine.

Public Health Relevance

Neuroblastoma is the most common extra-cranial solid tumor of childhood with an appalling 30% cure rate in children with advanced disease. There is a clear need for new chemotherapeutics, as current drugs are only marginally effective at the high doses that result in toxic acute and grave long term side effects. We are developing a drug engineered from a human enzyme that can kill tumors as a single agent by attacking a key metabolic point of neuroblastoma cells and is showing fantastic promise when used in combination with low non-toxic doses of current chemotherapeutics.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA154754-02
Application #
8208991
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Welch, Anthony R
Project Start
2011-01-01
Project End
2014-12-31
Budget Start
2012-01-01
Budget End
2012-12-31
Support Year
2
Fiscal Year
2012
Total Cost
$430,276
Indirect Cost
$89,607
Name
University of Texas Austin
Department
Engineering (All Types)
Type
Schools of Engineering
DUNS #
170230239
City
Austin
State
TX
Country
United States
Zip Code
78712
Cramer, Shira L; Saha, Achinto; Liu, Jinyun et al. (2017) Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat Med 23:120-127
Yan, Wupeng; Stone, Everett; Zhang, Yan Jessie (2017) Structural Snapshots of an Engineered Cystathionine-?-lyase Reveal the Critical Role of Electrostatic Interactions in the Active Site. Biochemistry 56:876-885
Li, Wenzong; Irani, Seema; Crutchfield, Amanda et al. (2016) Intramolecular Cleavage of the hASRGL1 Homodimer Occurs in Two Stages. Biochemistry 55:960-9
Burrage, Lindsay C; Sun, Qin; Elsea, Sarah H et al. (2015) Human recombinant arginase enzyme reduces plasma arginine in mouse models of arginase deficiency. Hum Mol Genet 24:6417-27
Chrysostomou, Constantine; Quandt, Erik M; Marshall, Nicholas M et al. (2015) An alternate pathway of arsenate resistance in E. coli mediated by the glutathione S-transferase GstB. ACS Chem Biol 10:875-82
Ahmed, Mahiuddin; Cheung, Nai-Kong V (2014) Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy. FEBS Lett 588:288-97
Paley, Olga; Agnello, Giulia; Cantor, Jason et al. (2013) GFP reporter screens for the engineering of amino acid degrading enzymes from libraries expressed in bacteria. Methods Mol Biol 978:31-44
Cheung, Nai-Kong V; Dyer, Michael A (2013) Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer 13:397-411
Agnello, Giulia; Chang, Leslie L; Lamb, Candice M et al. (2013) Discovery of a substrate selectivity motif in amino acid decarboxylases unveils a taurine biosynthesis pathway in prokaryotes. ACS Chem Biol 8:2264-71
Li, Wenzong; Cantor, Jason R; Yogesha, S D et al. (2012) Uncoupling intramolecular processing and substrate hydrolysis in the N-terminal nucleophile hydrolase hASRGL1 by circular permutation. ACS Chem Biol 7:1840-7

Showing the most recent 10 out of 11 publications