Bioengineered human arginase I with enhanced activity and stability controls hepatocellular and pancreatic carcinoma xenografts

Evan S Glazer, Everett M Stone, Cihui Zhu, Katherine L Massey, Amir N Hamir, Steven A Curley, Evan S Glazer, Everett M Stone, Cihui Zhu, Katherine L Massey, Amir N Hamir, Steven A Curley

Abstract

Hepatocellular carcinoma (HCC) and pancreatic carcinoma (PC) cells often have inherent urea cycle defects rendering them auxotrophic for the amino acid l-arginine (l-arg). Most HCC and PC require extracellular sources of l-arg and undergo cell cycle arrest and apoptosis when l-arg is restricted. Systemic, enzyme-mediated depletion of l-arg has been investigated in mouse models and human trials. Non-human enzymes elicit neutralizing antibodies, whereas human arginases display poor pharmacological properties in serum. Co(2+) substitution of the Mn(2+) metal cofactor in human arginase I (Co-hArgI) was shown to confer more than 10-fold higher catalytic activity (k(cat)/K(m)) and 5-fold greater stability. We hypothesized that the Co-hArgI enzyme would decrease tumor burden by systemic elimination of l-arg in a murine model. Co-hArgI was conjugated to 5-kDa PEG (Co-hArgI-PEG) to enhance circulation persistence. It was used as monotherapy for HCC and PC in vitro and in vivo murine xenografts. The mechanism of cell death was also investigated. Weekly treatment of 8 mg/kg Co-hArgI-PEG effectively controlled human HepG2 (HCC) and Panc-1 (PC) tumor xenografts (P = .001 and P = .03, respectively). Both cell lines underwent apoptosis in vitro with significant increased expression of activated caspase-3 (P < .001). Furthermore, there was evidence of autophagy in vitro and in vivo. We have demonstrated that Co-hArgI-PEG is effective at controlling two types of l-arg-dependent carcinomas. Being a nonessential amino acid, arginine deprivation therapy through Co-hArgI-PEG holds promise as a new therapy in the treatment of HCC and PC.

Figures

Figure 1
Figure 1
After day 1, the IC50 was 0.75 ± 0.36 and 0.44 ± 0.04 nM for HepG2 (A) and Panc-1 (B) cells, respectively, based on mitochondrial reductase activity (MTT). Flow cytometry demonstrated that HepG2 cells treated with 1 nM arginase (C) increased the apoptotic fraction (Annexin V-positive only, P = .001) and dead cell fraction (double positive for Annexin V and 7-AAD, P = .0003) with an appropriate decrease in viability (double negative, P = .0001). A similar decrease in the viability of Panc-1 cells was found (D), albeit less so when compared with the HepG2 cells (P = .004). Necrotic cells were identified as being Annexin V-negative, 7-AAD-positive cellular events.
Figure 2
Figure 2
Protein immunoblot demonstrated increased levels of LC3B-II after a single day of treatment with 1 nM Co-hArgI or placement in l-arg-free medium in HepG2 cells (A). This continued but decreased by day 4 after treatment without medium replacement. Panc-1 cells initially had decreased levels of LC3B-II 1 day after treatment but increased expression by day 4. Activated (cleaved large fragment) caspase-3 increased slightly after 1 day of treatment in HepG2 cells, whereas this was more clearly seen for Panc-1 cells 4 days after treatment. Investigation with flow cytometry demonstrated that more individual Panc-1 cells (B) contained similar levels of ASS-1 (P = .13), increased levels of activated caspase-3 (P = .0009), and similar Ki-67 expression (P = .66) 4 days after l-arg treatment compared with control cells. Likewise, more HepG2 cells (C) expressed ASS-1 (P = .0001), activated caspase-3 (P < .0001), and Ki-67 (P = .0002). All media contained 10% FBS which may contain some l-arg.
Figure 3
Figure 3
(A) Panc-1 subcutaneous xenografts in nude BALB/c mice demonstrated decreased tumor volume due to treatment with 8 mg/kg Co-hArgI-PEG (P = .03). (B) Likewise, HepG2 subcutaneous xenografts also demonstrated significantly smaller tumors, but they did so within 1 week of treatment (P = .001). (C) At the end of the experiment, PEG-Co-hArgI-treated tumors were much smaller as exemplified in the HepG2 tumors.
Figure 4
Figure 4
Although there is some necrosis (*) in HepG2 tumors from mice treated with PBS controls (A; hematoxylin and eosin stain, bright red areas are intravascular blood), there was significantly higher grades of necrosis in the tumors from arginase-treated mice (B). Likewise, a few cells in control tumors expressed activated caspase-3 (C; immunohistochemistry [IHC]), although significantly more cells expressed activated caspase-3 after Co-hArgI treatment (D). Finally, tumors from control-treated mice demonstrated far fewer autophagosomes (E; immunofluorescence [IF]; green label is LC3B-II, whereas blue label is DAPI) than tumors from arginase-treated mice (F). Larger nuclei (blue) are seen with Co-hArgI treatment (D, F).
Figure 5
Figure 5
Arginase-related toxicity was found in mice treated at doses of 15 mg/kg or greater. On necropsy, the only evidence of normal tissue toxicity was massive bone marrow devastation (A), whereas control bone marrow remained normal (B). In addition, weight loss was measured as a function of dose-dependent toxicity (0–32 mg/kg, n = 5 each) after IP injection of l-arg. The greatest observed weight loss occurred 4 days after treatment (C). The 16- and 32-mg/kg groups had a BCS of 1 and were killed for humane reasons. The mice in the 8-mg/kg group had a BCS score of 2 to 3 and regained their original body mass in an additional 2 to 3 days.

Source: PubMed

3
Subscribe