Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept

Upregulation of hydrogen sulfide (H<sub>2</sub>S) biosynthesis, at least in part related to the upregulation of cystathionine β-synthetase (CBS) in cancer cells, serves as a tumor-promoting factor and has emerged as a possible molecular target for antitumor drug development. To facilitat...

Full description

Bibliographic Details
Main Authors: Mark R. Hellmich, Celia Chao, Katalin Módis, Ye Ding, John R. Zatarain, Ketan Thanki, Manjit Maskey, Nadiya Druzhyna, Ashley A. Untereiner, Akbar Ahmad, Yu Xue, Haiying Chen, William K. Russell, Jianmei Wang, Jia Zhou, Csaba Szabo
Format: Article
Language:English
Published: MDPI AG 2021-07-01
Series:Biomolecules
Subjects:
Online Access:https://www.mdpi.com/2218-273X/11/8/1073
_version_ 1797524582721126400
author Mark R. Hellmich
Celia Chao
Katalin Módis
Ye Ding
John R. Zatarain
Ketan Thanki
Manjit Maskey
Nadiya Druzhyna
Ashley A. Untereiner
Akbar Ahmad
Yu Xue
Haiying Chen
William K. Russell
Jianmei Wang
Jia Zhou
Csaba Szabo
author_facet Mark R. Hellmich
Celia Chao
Katalin Módis
Ye Ding
John R. Zatarain
Ketan Thanki
Manjit Maskey
Nadiya Druzhyna
Ashley A. Untereiner
Akbar Ahmad
Yu Xue
Haiying Chen
William K. Russell
Jianmei Wang
Jia Zhou
Csaba Szabo
author_sort Mark R. Hellmich
collection DOAJ
description Upregulation of hydrogen sulfide (H<sub>2</sub>S) biosynthesis, at least in part related to the upregulation of cystathionine β-synthetase (CBS) in cancer cells, serves as a tumor-promoting factor and has emerged as a possible molecular target for antitumor drug development. To facilitate future clinical translation, we have synthesized a variety of novel CBS-targeting, esterase-cleavable prodrugs based on the structure of the prototypical CBS inhibitor aminooxyacetic acid (AOAA). The pharmacological properties of these compounds were evaluated in cell-free assays with recombinant human CBS protein, the human colon cancer cell line HCT116, and in vivo using various tumor-bearing mice models. The prodrug YD0251 (the isopropyl ester derivative of AOAA) was selected for detailed characterization. YD0251 exhibits improved antiproliferative efficacy in cell culture models when compared to AOAA. It is up to 18 times more potent than AOAA at suppressing HCT116 tumor growth in vivo and is effective when administered to tumor-bearing mice either via subcutaneous injection or oral gavage. Patient-derived xenografts (PDTXs) with higher levels of CBS protein grew significantly larger than tumors with lower levels, and YD0251 treatment inhibited the growth of PDTXs with elevated CBS, whereas it had no significant effect on PDTXs with low CBS protein levels. The toxicity of YD0251 was assessed in mice subjected to subchronic administration of supratherapeutic doses the inhibitor; no significant alteration in circulating markers of organ injury or histopathological alterations were noted, up to 60 mg/kg/day × 5 days. In preparation to a future theranostic concept (to match CBS inhibitor therapy to high-CBS expressors), we identified a potential plasma marker of CBS-expressing tumors. Colon cancer cells produced significant levels of lanthionine, a rare metabolic intermediate of CBS-mediated H<sub>2</sub>S biosynthesis; forced expression of CBS into non-transformed epithelial cells increased lanthionine biogenesis in vitro and in vivo (measured in the urine of tumor-bearing mice). These current results may be useful to facilitate the translation of a CBS inhibition-based antitumor concept into the clinical space.
first_indexed 2024-03-10T08:59:30Z
format Article
id doaj.art-7a180ec2f5f14c47af1604b5f70b4469
institution Directory Open Access Journal
issn 2218-273X
language English
last_indexed 2024-03-10T08:59:30Z
publishDate 2021-07-01
publisher MDPI AG
record_format Article
series Biomolecules
spelling doaj.art-7a180ec2f5f14c47af1604b5f70b44692023-11-22T06:54:22ZengMDPI AGBiomolecules2218-273X2021-07-01118107310.3390/biom11081073Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition ConceptMark R. Hellmich0Celia Chao1Katalin Módis2Ye Ding3John R. Zatarain4Ketan Thanki5Manjit Maskey6Nadiya Druzhyna7Ashley A. Untereiner8Akbar Ahmad9Yu Xue10Haiying Chen11William K. Russell12Jianmei Wang13Jia Zhou14Csaba Szabo15Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Biochemistry and Molecular Biology, University of Texas, Medical Branch, Galveston, TX 77555, USACollege of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USADepartment of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USADepartment of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USAUpregulation of hydrogen sulfide (H<sub>2</sub>S) biosynthesis, at least in part related to the upregulation of cystathionine β-synthetase (CBS) in cancer cells, serves as a tumor-promoting factor and has emerged as a possible molecular target for antitumor drug development. To facilitate future clinical translation, we have synthesized a variety of novel CBS-targeting, esterase-cleavable prodrugs based on the structure of the prototypical CBS inhibitor aminooxyacetic acid (AOAA). The pharmacological properties of these compounds were evaluated in cell-free assays with recombinant human CBS protein, the human colon cancer cell line HCT116, and in vivo using various tumor-bearing mice models. The prodrug YD0251 (the isopropyl ester derivative of AOAA) was selected for detailed characterization. YD0251 exhibits improved antiproliferative efficacy in cell culture models when compared to AOAA. It is up to 18 times more potent than AOAA at suppressing HCT116 tumor growth in vivo and is effective when administered to tumor-bearing mice either via subcutaneous injection or oral gavage. Patient-derived xenografts (PDTXs) with higher levels of CBS protein grew significantly larger than tumors with lower levels, and YD0251 treatment inhibited the growth of PDTXs with elevated CBS, whereas it had no significant effect on PDTXs with low CBS protein levels. The toxicity of YD0251 was assessed in mice subjected to subchronic administration of supratherapeutic doses the inhibitor; no significant alteration in circulating markers of organ injury or histopathological alterations were noted, up to 60 mg/kg/day × 5 days. In preparation to a future theranostic concept (to match CBS inhibitor therapy to high-CBS expressors), we identified a potential plasma marker of CBS-expressing tumors. Colon cancer cells produced significant levels of lanthionine, a rare metabolic intermediate of CBS-mediated H<sub>2</sub>S biosynthesis; forced expression of CBS into non-transformed epithelial cells increased lanthionine biogenesis in vitro and in vivo (measured in the urine of tumor-bearing mice). These current results may be useful to facilitate the translation of a CBS inhibition-based antitumor concept into the clinical space.https://www.mdpi.com/2218-273X/11/8/1073hydrogen sulfideanticancerbiomarkerprodrug
spellingShingle Mark R. Hellmich
Celia Chao
Katalin Módis
Ye Ding
John R. Zatarain
Ketan Thanki
Manjit Maskey
Nadiya Druzhyna
Ashley A. Untereiner
Akbar Ahmad
Yu Xue
Haiying Chen
William K. Russell
Jianmei Wang
Jia Zhou
Csaba Szabo
Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
Biomolecules
hydrogen sulfide
anticancer
biomarker
prodrug
title Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
title_full Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
title_fullStr Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
title_full_unstemmed Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
title_short Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept
title_sort efficacy of novel aminooxyacetic acid prodrugs in colon cancer models towards clinical translation of the cystathionine β synthase inhibition concept
topic hydrogen sulfide
anticancer
biomarker
prodrug
url https://www.mdpi.com/2218-273X/11/8/1073
work_keys_str_mv AT markrhellmich efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT celiachao efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT katalinmodis efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT yeding efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT johnrzatarain efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT ketanthanki efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT manjitmaskey efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT nadiyadruzhyna efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT ashleyauntereiner efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT akbarahmad efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT yuxue efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT haiyingchen efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT williamkrussell efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT jianmeiwang efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT jiazhou efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept
AT csabaszabo efficacyofnovelaminooxyaceticacidprodrugsincoloncancermodelstowardsclinicaltranslationofthecystathioninebsynthaseinhibitionconcept