101 results on '"Zhanjun Hou"'
Search Results
2. Biology and therapeutic applications of the proton-coupled folate transporter
- Author
-
Larry H. Matherly, Mathew Schneider, Aleem Gangjee, and Zhanjun Hou
- Subjects
Pharmacology ,Folic Acid ,Neoplasms ,Humans ,Folic Acid Antagonists ,Antineoplastic Agents ,General Medicine ,Pemetrexed ,Toxicology ,Biology ,Proton-Coupled Folate Transporter - Abstract
The proton-coupled folate transporter (PCFT; SLC46A1) was discovered in 2006 as the principal mechanism by which folates are absorbed in the intestine and the causal basis for hereditary folate malabsorption (HFM). In 2011, it was found that PCFT is highly expressed in many tumors. This stimulated interest in using PCFT for cytotoxic drug targeting, taking advantage of the substantial levels of PCFT transport and acidic pH conditions commonly associated with tumors.We summarize the literature from 2006 to 2022 that explores the role of PCFT in the intestinal absorption of dietary folates and its role in HFM and as a transporter of folates and antifolates such as pemetrexed (Alimta) in relation to cancer. We provide the rationale for the discovery of a new generation of targeted pyrrolo[2,3-We summarize the promising future and potential challenges of implementing PCFT-targeted therapeutics for HFM and a variety of cancers.
- Published
- 2023
3. Enhancing anti-AML activity of venetoclax by isoflavone ME-344 through suppression of OXPHOS and purine biosynthesis
- Author
-
Natalia Baran, Katie Hurrish, Yongwei Su, Shraddha Patel, Cassandra Ramage, Jenna Carter, Holly Edwards, Steven Buck, Sandra Wiley, Maik Hüttemann, Lisa Polin, Juiwanna Kushner, Sijana Dzinic, Kathryn White, Xun Bao, Jing Li, Jay Yang, Julie Boerner, Zhanjun Hou, Gheath Al-Atrash, Sergej Konoplev, Jonathan Busquets, Stefano Tiziani, Larry Matherly, Jeffrey Taub, Marina Konopleva, and Yubin Ge
- Abstract
Venetoclax (VEN), in combination with low dose cytarabine (AraC) or a hypomethylating agent, is FDA approved to treat acute myeloid leukemia (AML) in patients who are over the age of 75 or cannot tolerate standard chemotherapy. Despite high response rates to these combination therapies, most patients succumb to the disease due to relapse and/or drug resistance, providing an unmet clinical need for novel therapies to improve AML patient survival. ME-344 is a potent isoflavone with demonstrated inhibitory activity toward oxidative phosphorylation (OXPHOS) and clinical activity in solid tumors. Given that OXPHOS inhibition enhances VEN antileukemic activity against AML, we hypothesized that ME-344 could enhance the anti-AML activity of VEN. Here we report that ME-344 synergized with VEN to target AML cell lines and primary patient samples while sparing normal hematopoietic cells. Cooperative suppression of OXPHOS was detected in a subset of AML cell lines and primary patient samples. Metabolomics analysis revealed a significant reduction of purine biosynthesis metabolites by ME-344. Further, lometrexol, an inhibitor of purine biosynthesis, synergistically enhanced VEN-induced apoptosis in AML cell lines. Interestingly, AML cells with acquired resistance to AraC showed significantly increased purine biosynthesis metabolites and sensitivities to ME-344. Furthermore, synergy between ME-344 and VEN was preserved in these AraC-resistant AML cells. These results translated into significantly prolonged survival upon combination of ME-344 and VEN in NSGS mice bearing parental or AraC-resistant MV4-11 leukemia. This study demonstrates that ME-344 enhances VEN antileukemic activity against preclinical models of AML by suppressing OXPHOS and/or purine biosynthesis.
- Published
- 2023
- Full Text
- View/download PDF
4. A Beta Strain-Based Spike Glycoprotein Vaccine Candidate Induces Broad Neutralization and Protection against SARS-CoV-2 Variants of Concern
- Author
-
Lei Cao, Jinyuan Guo, Hai Li, Hu Ren, Kang Xiao, Yan Zhang, Shuangli Zhu, Yang Song, Weijia Zhao, Dan Wu, Zhihui Chen, Yanan Zhang, Baicheng Xia, Tianjiao Ji, Dongmei Yan, Dongyan Wang, Qian Yang, Yangzi Zhou, Xiaolei Li, Zhanjun Hou, and Wenbo Xu
- Subjects
Microbiology (medical) ,Infectious Diseases ,General Immunology and Microbiology ,Ecology ,Physiology ,Genetics ,Cell Biology - Abstract
Continuous emergence of adaptive mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the use and development of existing vaccines and drugs. The value of variant-based vaccines that are capable of inducing a higher and broader protection immune response against SARS-CoV-2 variants is currently being evaluated.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Methods from Novel Pyrrolo[3,2-d]pyrimidine Compounds Target Mitochondrial and Cytosolic One-carbon Metabolism with Broad-spectrum Antitumor Efficacy
- Author
-
Larry H. Matherly, Aleem Gangjee, Charles E. Dann, Zhanjun Hou, Jing Li, Joshua D. Rabinowitz, Lisa Polin, Maik Hüttemann, Seongho Kim, Juiwanna Kushner, Kathryn White, Sijana H. Dzinic, Carrie O'Connor, Adrianne Wallace-Povirk, Jenney Liu, Josephine Frühauf, Xun Bao, Changwen Ning, Arpit Doshi, Md. Junayed Nayeen, Jennifer Wong-Roushar, Jade M. Katinas, Gregory S. Ducker, Khushbu Shah, and Aamod S. Dekhne
- Abstract
Supplementary Method Descriptions Molecular modeling and computational studies Enzyme expression and purification. In vitro enzymatic assays and Ki determinations In vivo efficacy trials with MIA PaCa-2 pancreatic cancer xenografts. Cytochrome c oxidase assay Synthesis of AGF94 and 5-substituted pyrrolo[3,2-d]pyrimidine compounds
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Novel Pyrrolo[3,2-d]pyrimidine Compounds Target Mitochondrial and Cytosolic One-carbon Metabolism with Broad-spectrum Antitumor Efficacy
- Author
-
Larry H. Matherly, Aleem Gangjee, Charles E. Dann, Zhanjun Hou, Jing Li, Joshua D. Rabinowitz, Lisa Polin, Maik Hüttemann, Seongho Kim, Juiwanna Kushner, Kathryn White, Sijana H. Dzinic, Carrie O'Connor, Adrianne Wallace-Povirk, Jenney Liu, Josephine Frühauf, Xun Bao, Changwen Ning, Arpit Doshi, Md. Junayed Nayeen, Jennifer Wong-Roushar, Jade M. Katinas, Gregory S. Ducker, Khushbu Shah, and Aamod S. Dekhne
- Abstract
Folate-dependent one-carbon (C1) metabolism is compartmentalized into the mitochondria and cytosol and supports cell growth through nucleotide and amino acid biosynthesis. Mitochondrial C1 metabolism, including serine hydroxymethyltransferase (SHMT) 2, provides glycine, NAD(P)H, ATP, and C1 units for cytosolic biosynthetic reactions, and is implicated in the oncogenic phenotype across a wide range of cancers. Whereas multitargeted inhibitors of cytosolic C1 metabolism, such as pemetrexed, are used clinically, there are currently no anticancer drugs that specifically target mitochondrial C1 metabolism. We used molecular modeling to design novel small-molecule pyrrolo[3,2-d]pyrimidine inhibitors targeting mitochondrial C1 metabolism at SHMT2. In vitro antitumor efficacy was established with the lead compounds (AGF291, AGF320, AGF347) toward lung, colon, and pancreatic cancer cells. Intracellular targets were identified by metabolic rescue with glycine and nucleosides, and by targeted metabolomics using a stable isotope tracer, with confirmation by in vitro assays with purified enzymes. In addition to targeting SHMT2, inhibition of the cytosolic purine biosynthetic enzymes, β-glycinamide ribonucleotide formyltransferase and/or 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase, and SHMT1 was also established. AGF347 generated significant in vivo antitumor efficacy with potential for complete responses against both early-stage and upstage MIA PaCa-2 pancreatic tumor xenografts, providing compelling proof-of-concept for therapeutic targeting of SHMT2 and cytosolic C1 enzymes by this series. Our results establish structure–activity relationships and identify exciting new drug prototypes for further development as multitargeted antitumor agents.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Tables S1-S2 from Dual Targeting of Epithelial Ovarian Cancer Via Folate Receptor α and the Proton-Coupled Folate Transporter with 6-Substituted Pyrrolo[2,3-d]pyrimidine Antifolates
- Author
-
Larry H. Matherly, Aleem Gangjee, Robert T. Morris, Juiwanna Kushner, Kathryn White, Lisa Polin, Steve Orr, Christina George, Adrianne Wallace-Povirk, Si Yang, Carrie O'Connor, Leda Gattoc, and Zhanjun Hou
- Abstract
Table S1: Epithelial ovarian cancer (EOC) tissue cDNA array for real-time RT-PCR (Figures 2A and 2B). Table S2. Epithelial ovarian cancer (EOC) tissue microarray (TMA) for immunohistochemistry (IHC; Figures 2C and 2D).
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Data from Novel Pyrrolo[3,2-d]pyrimidine Compounds Target Mitochondrial and Cytosolic One-carbon Metabolism with Broad-spectrum Antitumor Efficacy
- Author
-
Larry H. Matherly, Aleem Gangjee, Charles E. Dann, Zhanjun Hou, Jing Li, Joshua D. Rabinowitz, Lisa Polin, Maik Hüttemann, Seongho Kim, Juiwanna Kushner, Kathryn White, Sijana H. Dzinic, Carrie O'Connor, Adrianne Wallace-Povirk, Jenney Liu, Josephine Frühauf, Xun Bao, Changwen Ning, Arpit Doshi, Md. Junayed Nayeen, Jennifer Wong-Roushar, Jade M. Katinas, Gregory S. Ducker, Khushbu Shah, and Aamod S. Dekhne
- Abstract
Supplementary Figure S1 shows in vitro antiproliferative activity and targeted pathways of previously reported AICARFTase and GARFTase inhibitors. Supplementary Figure S2 shows docking of AGF291, AGF320, and AGF347 (along with 5-formyl-THF) in human SHMT2 and rabbit SHMT1. Supplementary Figure S3 shows plasma membrane folate transporter expression levels in H460, HCT116, and MIA PaCa-2 human tumor cell lines as compared to those in the IGROV-1 epithelial ovarian cancer cell line. Supplementary Figure S4 shows in vitro antiproliferative activity and targeted pathways of AGF291, AGF320, and AGF347 along with previously reported GARFTase inhibitor AGF94 in H460, HCT116, and MIA PaCa-2 cell lines. Supplementary Figure S5 shows targeted metabolomics data on total serine, serine isotope labeling patterns, total GAR, total AICAR, total adenine nucleotides, and total dTTP along with isotope labeling patterns of H460, HCT116, and MIA PaCa-2 cell lines treated with AGF291, AGF320, and AGF347 not shown in main text Figure 4. Supplementary Figure S6 shows a Western blot confirming knockdown of SHMT2 in H460 SHMT2 KD cell line and knockout of SHMT2 in HCT116 SHMT2 KO cell line. Supplementary Figure S7 shows in vivo efficacies of AGF347 and gemcitabine towards MIA PaCa-2 early and late stage tumor xenograft models where mouse serum folate levels were not depleted to approximate those found in humans. Supplementary Figure S8 shows a cytochrome c oxidase assay on tumors harvested from the metabolomics arm of the late stage in vivo AGF347 trial. Table S1 shows docking scores of novel compounds in human SHMT2 and rabbit SHMT1 (corresponding to Supplementary Figure S2). Table S2 shows quantitative data from in vivo early- and late-stage trials of AGF347 and gemcitabine against MIA PaCa-2 tumor xenografts in NCR SCID mice.
- Published
- 2023
- Full Text
- View/download PDF
9. EP217/#328 BRCA1 and BRCA2 mutations lead to differential Wnt signaling in ovarian cancer cells
- Author
-
Ayesha Alvero, Ramandeep Rattan, Zhanjun Hou, Sandra Galoforo, Alexandra Fox, Seongo Kim, Rouba Ali-Fehmi, Mira Kheil, Robert Morris, Larry Matherly, Gil Mor, and Radhika Gogoi
- Published
- 2022
- Full Text
- View/download PDF
10. Therapeutic Targeting of Mitochondrial One-Carbon Metabolism in Cancer
- Author
-
Larry H. Matherly, Zhanjun Hou, Aleem Gangjee, and Aamod Dekhne
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents ,Mitochondrion ,Biology ,Article ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,0302 clinical medicine ,Aminohydrolases ,Neoplasms ,Serine ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Epigenetics ,Amino acid synthesis ,Glycine Hydroxymethyltransferase ,Methylenetetrahydrofolate Dehydrogenase (NADP) ,chemistry.chemical_classification ,Cancer ,Metabolism ,medicine.disease ,Multifunctional Enzymes ,Carbon ,Mitochondria ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cytosol ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Serine hydroxymethyltransferase ,Cancer cell ,Energy Metabolism ,Biomarkers ,Metabolic Networks and Pathways - Abstract
One-carbon (1C) metabolism encompasses folate-mediated 1C transfer reactions and related processes, including nucleotide and amino acid biosynthesis, antioxidant regeneration, and epigenetic regulation. 1C pathways are compartmentalized in the cytosol, mitochondria, and nucleus. 1C metabolism in the cytosol has been an important therapeutic target for cancer since the inception of modern chemotherapy, and “antifolates” targeting cytosolic 1C pathways continue to be a mainstay of the chemotherapy armamentarium for cancer. Recent insights into the complexities of 1C metabolism in cancer cells, including the critical role of the mitochondrial 1C pathway as a source of 1C units, glycine, reducing equivalents, and ATP, have spurred the discovery of novel compounds that target these reactions, with particular focus on 5,10-methylene tetrahydrofolate dehydrogenase 2 and serine hydroxymethyltransferase 2. In this review, we discuss key aspects of 1C metabolism, with emphasis on the importance of mitochondrial 1C metabolism to metabolic homeostasis, its relationship with the oncogenic phenotype, and its therapeutic potential for cancer.
- Published
- 2020
- Full Text
- View/download PDF
11. Abstract 4903: Levels of folate transporters impact the compartmentalization of one-carbon metabolism in the mitochondria vs cytosol providing a unique vulnerability to SHMT inhibition
- Author
-
Mathew Joseph Schneider, Carrie O'connor, Xun Bao, Md. Junayed Nayeen, Zhanjun Hou, Jing Li, Aleem Gangjee, and Larry H. Matherly
- Subjects
Cancer Research ,Oncology - Abstract
These studies characterize the critical role of the Reduced Folate Carrier (RFC) and the Proton Coupled Folate Transporter (PCFT) as determinants of cancer cell reliance on mitochondrial vs cytosolic one carbon (C1) metabolism, to identify a unique susceptibility towards SHMT1 and SHMT2 inhibition in cancer cells. C1 metabolism is frequently reprogrammed in cancer cells to provide nucleotides, amino acids, and glutathione, and to maintain redox homeostasis for proliferation. The mitochondrial C1 converting enzymes serine hydroxymethyltransferase (SHMT) 2 and methylene tetrahydrofolate (THF) dehydrogenase 2 (MTHFD2) are among the top overexpressed metabolic enzymes in human cancers, suggesting these are important cancer-specific targets. We previously discovered novel 5-substituted pyrrolo[3,2-d]pyrimidine antifolates (with AGF347 being the lead) that potently inhibit mitochondrial SHMT2, as well as SHMT1. Folates are essential cofactors for C1 converting reactions; folates and antifolates primarily rely on the major facilitative folate transporters RFC and PCFT for internalization by tumor cells. In these studies, we explore the effects of folate transporter expression on the compartmentalization of C1 metabolism in the mitochondria and cytosol and the potential impact on SHMT1 and SHMT2 inhibition. We used a tetracycline inducible system for RFC expression in RFC/PCFT-null HeLa cells, with and without constitutive PCFT, and characterized the impact on transport and accumulation of folates and AGF347 in response to folate transporter expression. Increased accumulation of folates was observed with increasing RFC in the absence of PCFT. In the presence of PCFT, baseline folate accumulation was substantial and independent of RFC expression, indicating a major role of PCFT as a facilitative folate transporter. SHMT2 catalyzes the conversion of serine and THF to glycine and 5, 10-methylene THF in the mitochondria, while SHMT1 catalyzes the reverse reaction in the cytosol, producing THF and serine. To determine how folate transporter expression affects the C1 flux through SHMT1 vs SHMT2, [2,3,3-2H]serine tracer experiments were performed; increased mitochondrial C1 flux through SHMT2 was observed with increasing RFC in the absence of PCFT, whereas the mitochondrial flux through SHMT2 predominated in the presence of PCFT. By cell proliferation assays it was discovered that cells with lower RFC expression exhibit a hypersensitive phenotype towards AGF347 in the absence of PCFT, indicating a unique therapeutic opportunity for targeting SHMT forms in cancer cells with low folate transport activity. Here we identify RFC and PCFT as key determinants for the compartmentalization of C1 flux in the mitochondria vs the cytosol, in turn affecting the sensitivity of cancer cells towards SHMT inhibition. Citation Format: Mathew Joseph Schneider, Carrie O'connor, Xun Bao, Md. Junayed Nayeen, Zhanjun Hou, Jing Li, Aleem Gangjee, Larry H. Matherly. Levels of folate transporters impact the compartmentalization of one-carbon metabolism in the mitochondria vs cytosol providing a unique vulnerability to SHMT inhibition. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4903.
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract 4902: Mitochondrial and cytosolic folylpolyglutamate synthetase in one-carbon metabolism and anti-tumor efficacy of mitochondrial-targeted antifolates
- Author
-
Carrie O’Connor, Jade Katinas, Mathew Schneider, Md. Junayed Nayeen, Xun Bao, Jing Li, Charles Dann, Aleem Gangjee, Larry H. Matherly, and Zhanjun Hou
- Subjects
Cancer Research ,Oncology - Abstract
Folates mediate one-carbon (C1) transfers, which are essential for cellular homeostasis and survival. C1 metabolism encompasses distinct cytosol (Cyto) and mitochondria (Mito) pathways connected by an interchange between serine, glycine and formate. Mito C1 metabolism provides cellular glycine and C1 units (as formate) for de novo synthesis of thymidylate and purine nucleotides in the Cyto. Polyglutamyl folates are the predominant folate forms in cells and are generally preferred for C1 transfers. Thus, folate polyglutamylation is essential for cellular homeostasis. Folate polyglutamylation is catalyzed by folypolyglutamate synthetase (FPGS), including Cyto (cFPGS) and Mito (mFPGS) isoforms. C1 metabolism is critical for tumor growth and thus offers a plethora of therapeutic targets for cancer. Of particular interest are Mito C1 inhibitors including “non-classical” pyrazolopyran compounds SHIN1 and SHIN2, and “classical” pyrrolo[3,2-d]pyrimidine antifolate compounds typified by AGF347, all targeting serine hydroxymethyltransferase 2 (SHMT2). Both SHIN1 and AGF347 effected in vitro anti-tumor efficacy, and SHIN2 and AGF347 showed in vivo efficacy toward tumor xenografts. We systematically explored the roles of cFPGS and mFPGS levels as critical determinants of C1 inhibitor target engagement and net Cyto versus Mito C1 flux, resulting in anti-tumor efficacy. We found that FPGS transcript levels significantly correlated with the effects of AGF347 toward a panel of human pancreatic cancer cell lines including MIA PaCa-2. We engineered MIA PaCa-2 cells with FPGS gene knockout to stably express inducible mFPGS or cFPGS form. FPGS in mFPGS-transfected cells (mFPGS#32) was expressed mainly in Mito over Cyto, whereas FPGS in cFPGS-transfected cells (cFPGS#3) was expressed exclusively in Cyto. Accumulation of radiolabeled folate and AGF347 increased with increasing FPGS in both Cyto and Mito for mFPGS#32, but only in Cyto for cFPGS#3. Metabolomics with [2,3,3-2H]serine in cFPGS#3 and mFPGS#32 established FPGS levels in Mito versus Cyto as important determinants of C1 fluxes. Increased FPGS levels in mFPGS#32 enhanced C1 flux in Mito far greater than in Cyto, and greater than for the increased FPGS in cFPGS#3. As a result, Cyto-targeted antifolates pemetrexed and AGF94 were only modestly impacted by increasing FPGS levels in both mFPGS#32 and cFPGS#3 (~3x). In contrast, increasing FPGS levels dramatically enhanced the inhibitory effects of Mito-targeted antifolates such as AGF347 in mFPGS#32 (~8-25x), and to a greater extent than in cFPGS#3 (~6-8x). Conversely, increasing FPGS levels substantially attenuated the cytotoxic effect of SHIN1 in mFPGS#32 (~19x). In summary, FPGS levels are an important determinant of C1 fluxes, particularly in Mito where they contribute to cytotoxic potencies of Mito-targeted C1 inhibitors at SHMT2. Citation Format: Carrie O’Connor, Jade Katinas, Mathew Schneider, Md. Junayed Nayeen, Xun Bao, Jing Li, Charles Dann, Aleem Gangjee, Larry H. Matherly, Zhanjun Hou. Mitochondrial and cytosolic folylpolyglutamate synthetase in one-carbon metabolism and anti-tumor efficacy of mitochondrial-targeted antifolates. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4902.
- Published
- 2023
- Full Text
- View/download PDF
13. The evolving biology of the proton‐coupled folate transporter: New insights into regulation, structure, and mechanism
- Author
-
Zhanjun Hou, Aleem Gangjee, and Larry H. Matherly
- Subjects
Folic Acid ,Transcription, Genetic ,Neoplasms ,Genetics ,Animals ,Folic Acid Antagonists ,Humans ,Molecular Biology ,Biochemistry ,Article ,Proton-Coupled Folate Transporter ,Transcription Factors ,Biotechnology - Abstract
The human proton-coupled folate transporter (PCFT; SLC46A1) or hPCFT was identified in 2006 as the principal folate transporter involved in the intestinal absorption of dietary folates. A rare autosomal recessive hereditary folate malabsorption syndrome is attributable to human SLC46A1 variants. The recognition that hPCFT was highly expressed in many tumors stimulated substantial interest in its potential for cytotoxic drug targeting, taking advantage of its high-level transport activity under acidic pH conditions that characterize many tumors and its modest expression in most normal tissues. To better understand the basis for variations in hPCFT levels between tissues including human tumors, studies have examined the transcriptional regulation of hPCFT including the roles of CpG hypermethylation and critical transcription factors and cis elements. Additional focus involved identifying key structural and functional determinants of hPCFT transport that, combined with homology models based on structural homologies to the bacterial transporters GlpT and LacY, have enabled new structural and mechanistic insights. Recently, cryo-electron microscopy structures of chicken PCFT in a substrate-free state and in complex with the antifolate pemetrexed were reported, providing further structural insights into determinants of (anti)folate recognition and the mechanism of pH-regulated (anti)folate transport by PCFT. Like many major facilitator proteins, hPCFT exists as a homo-oligomer, and evidence suggests that homo-oligomerization of hPCFT monomeric proteins may be important for its intracellular trafficking and/or transport function. Better understanding of the structure, function and regulation of hPCFT should facilitate the rational development of new therapeutic strategies for conditions associated with folate deficiency, as well as cancer.
- Published
- 2022
- Full Text
- View/download PDF
14. Correction: Novel Pyrrolo[3,2-d]Pyrimidine Compounds Target Mitochondrial and Cytosolic One-Carbon Metabolism with Broad-spectrum Antitumor Efficacy
- Author
-
Aamod S. Dekhne, Khushbu Shah, Gregory S. Ducker, Jade M. Katinas, Jennifer Wong-Roushar, Md. Junayed Nayeen, Arpit Doshi, Changwen Ning, Xun Bao, Josephine Frühauf, Jenney Liu, Adrianne Wallace-Povirk, Carrie O'Connor, Sijana H. Dzinic, Kathryn White, Juiwanna Kushner, Seongho Kim, Maik Hüttemann, Lisa Polin, Joshua D. Rabinowitz, Jing Li, Zhanjun Hou, Charles E. Dann, Aleem Gangjee, and Larry H. Matherly
- Subjects
Cancer Research ,Oncology - Published
- 2023
- Full Text
- View/download PDF
15. Targeted therapy of pyrrolo[2,3-d]pyrimidine antifolates in a syngeneic mouse model of high grade serous ovarian cancer and the impact on the tumor microenvironment
- Author
-
Adrianne Wallace-Povirk, Lisa Rubinsak, Agnes Malysa, Sijana H. Dzinic, Manasa Ravindra, Mathew Schneider, James Glassbrook, Carrie O’Connor, Zhanjun Hou, Seongho Kim, Jessica Back, Lisa Polin, Robert T. Morris, Aleem Gangjee, Heather Gibson, and Larry H. Matherly
- Subjects
Ovarian Neoplasms ,Mice ,Multidisciplinary ,Pyrimidines ,Tumor Microenvironment ,Animals ,Folic Acid Antagonists ,Humans ,Antineoplastic Agents ,Female ,Carcinoma, Ovarian Epithelial - Abstract
Novel therapies are urgently needed for epithelial ovarian cancer (EOC), the most lethal gynecologic malignancy. In addition, therapies that target unique vulnerabilities in the tumor microenvironment (TME) of EOC have largely been unrealized. One strategy to achieve selective drug delivery for EOC therapy involves use of targeted antifolates via their uptake by folate receptor (FR) proteins, resulting in inhibition of essential one-carbon (C1) metabolic pathways. FRα is highly expressed in EOCs, along with the proton-coupled folate transporter (PCFT); FRβ is expressed on activated macrophages, a major infiltrating immune population in EOC. Thus, there is great potential for targeting both the tumor and the TME with agents delivered via selective transport by FRs and PCFT. In this report, we investigated the therapeutic potential of a novel cytosolic C1 6-substituted pyrrolo[2,3-d]pyrimidine inhibitor AGF94, with selectivity for uptake by FRs and PCFT and inhibition of de novo purine nucleotide biosynthesis, against a syngeneic model of ovarian cancer (BR-Luc) which recapitulates high-grade serous ovarian cancer in patients. In vitro activity of AGF94 was extended in vivo against orthotopic BR-Luc tumors. With late-stage subcutaneous BR-Luc xenografts, AGF94 treatment resulted in substantial anti-tumor efficacy, accompanied by significantly decreased M2-like FRβ-expressing macrophages and increased CD3+ T cells, whereas CD4+ and CD8+ T cells were unaffected. Our studies demonstrate potent anti-tumor efficacy of AGF94 in the therapy of EOC in the context of an intact immune system, and provide a framework for targeting the immunosuppressive TME as an essential component of therapy.
- Published
- 2021
16. Cellular Pharmacodynamics of a Novel Pyrrolo[3,2-d]pyrimidine Inhibitor Targeting Mitochondrial and Cytosolic One-Carbon Metabolism
- Author
-
Khushbu Shah, Maik Hüttemann, Seongho Kim, Adrianne Wallace-Povirk, Larry H. Matherly, Josphephine Fruhauf, Md. Junayed Nayeen, Carrie O'Connor, Chengwen Ning, Hasini A. Kalpage, Aleem Gangjee, Aamod Dekhne, and Zhanjun Hou
- Subjects
0301 basic medicine ,Purine ,Leucovorin ,Antineoplastic Agents ,Mitochondrion ,Serine ,Gene Knockout Techniques ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,0302 clinical medicine ,Cell Line, Tumor ,Humans ,Pyrroles ,Nucleotide ,Purine metabolism ,Purine Nucleotides ,Tetrahydrofolates ,Glycine Hydroxymethyltransferase ,Pharmacology ,chemistry.chemical_classification ,Cell Membrane ,Articles ,Glutathione ,Mitochondria ,Pancreatic Neoplasms ,Pyrimidines ,030104 developmental biology ,Biochemistry ,chemistry ,Serine hydroxymethyltransferase ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Reactive Oxygen Species ,030217 neurology & neurosurgery - Abstract
Folate-dependent one-carbon (C1) metabolism is compartmentalized in the mitochondria and cytosol and is a source of critical metabolites for proliferating tumors. Mitochondrial C1 metabolism including serine hydroxymethyltransferase 2 (SHMT2) generates glycine for de novo purine nucleotide and glutathione biosynthesis and is an important source of NADPH, ATP, and formate, which affords C1 units as 10-formyl-tetrahydrofolate and 5,10-methylene-tetrahydrofolate for nucleotide biosynthesis in the cytosol. We previously discovered novel first-in-class multitargeted pyrrolo[3,2-d]pyrimidine inhibitors of SHMT2 and de novo purine biosynthesis at glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase with potent in vitro and in vivo antitumor efficacy toward pancreatic adenocarcinoma cells. In this report, we extend our findings to an expanded panel of pancreatic cancer models. We used our lead analog AGF347 [(4-(4-(2-amino-4-oxo-3,4-dihydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl)butyl)-2-fluorobenzoyl)-l-glutamic acid] to characterize pharmacodynamic determinants of antitumor efficacy for this series and demonstrated plasma membrane transport into the cytosol, uptake from cytosol into mitochondria, and metabolism to AGF347 polyglutamates in both cytosol and mitochondria. Antitumor effects of AGF347 downstream of SHMT2 and purine biosynthesis included suppression of mammalian target of rapamycin signaling, and glutathione depletion with increased levels of reactive oxygen species. Our results provide important insights into the cellular pharmacology of novel pyrrolo[3,2-d]pyrimidine inhibitors as antitumor compounds and establish AGF347 as a unique agent for potential clinical application for pancreatic cancer, as well as other malignancies. SIGNIFICANCE STATEMENT: This study establishes the antitumor efficacies of novel inhibitors of serine hydroxymethyltransferase 2 and of cytosolic targets toward a panel of clinically relevant pancreatic cancer cells and demonstrates the important roles of plasma membrane transport, mitochondrial accumulation, and metabolism to polyglutamates of the lead compound AGF347 to drug activity. We also establish that loss of serine catabolism and purine biosynthesis resulting from AGF347 treatment impacts mammalian target of rapamycin signaling, glutathione pools, and reactive oxygen species, contributing to antitumor efficacy.
- Published
- 2019
- Full Text
- View/download PDF
17. Abstract 3785: ME-344, a novel isoflavone mitochondrial inhibitor, in combination with venetoclax constitutes a new metabolism-targeted approach to overcome resistance to Bcl-2 inhibition and standard of care treatment in AML
- Author
-
Katie H. Hurrish, Yongwei Su, Shraddha Patel, Sandra E. Wiley, Zhanjun Hou, Jenna Carter, Hasini Kalpage, Maik Hüttemann, Connie Weng, Holly Edwards, Lisa Polin, Jing Li, Jay Yang, Larry H. Matherly, Sergej Naumovich Konoplev, Jeffrey W. Taub, Marina Konopleva, Yubin Ge, and Natalia Baran
- Subjects
Cancer Research ,Oncology - Abstract
Acute myeloid Leukemia (AML) is an aggressive hematologic malignancy with poor prognosis. Despite chromosomal and genetic heterogeneity, AML are uniformly characterized by increased reliance on oxidative phosphorylation (OXPHOS). This key metabolic hallmark of leukemia was recently reported as a feature of resistance to Cytarabine (AraC)-based therapy. Also, an aggressive phenotype and poor response to chemotherapy is associated with increased levels of Bcl-2. Despite the introduction of the Bcl-2 inhibitor venetoclax (VEN), the overall survival, particularly in older patients, remains poor. Thus, approaches to improve the sensitivity of leukemic cells to AraC-based or Bcl-2 based therapies are urgently needed.Here, we investigated the preclinical activity of ME-344, a novel isoflavone OXPHOS inhibitor, on AML cell lines and relapsed/refractory (R/R) patient samples in vitro and examined the efficacy of ME-344 in combination with VEN in Ara-C sensitive and resistant AML cell lines and patient-derived xenografts (PDX) both in vitro and in vivo.ME-344 (0-300 nM, 24 hr) significantly reduced viability of AML cell lines with EC50 of 75-100 nM and R/R AML patient samples with EC50 of 200-300 nM respectively. The cytotoxic response in AML was enhanced when ME-344 was combined with VEN, producing strong synergistic viability reduction and induction of apoptosis, as evidenced by Annexin V assay and an increased level of cleaved caspase 3 and PARP (immunoblotting). The dual inhibition of OXPHOS/Bcl-2 reduced Mcl-1 levels and showed efficacy in Mcl-1 overexpressingand Ara-C resistant AML models.Functional metabolic characterization of AML by transcriptomics and mass spectrometry demonstrated that ME-344 effectively inhibited biosynthetic pathways of nucleotides uncovering the purine biosynthesis pathway as crucial for therapeutic efficacy. ME-344 induced a dose-dependent decrease in the oxygen consumption rate (by Seahorse assay), in both AraC-sensitive and -resistant AML cell lines, and in R/R AML patient samples, which was further significantly potentiated by combination with VEN.Finally, in an aggressive AML xenograft model, ME-344 (200 mpk, i.v.) combined with subtherapeutic doses of VEN (25 mpk) reduced circulating leukemia burden and extended survival. Ongoing in vivo studies in AML PDX models will address the impact of ME-344 in the context of acquired AraC- and Bcl-2- resistance. In summary, our findings indicate ME-344 alone or in combination with Bcl-2 inhibition constitutes an important therapeutic modality that targets a unique metabolic vulnerability of AML. Citation Format: Katie H. Hurrish, Yongwei Su, Shraddha Patel, Sandra E. Wiley, Zhanjun Hou, Jenna Carter, Hasini Kalpage, Maik Hüttemann, Connie Weng, Holly Edwards, Lisa Polin, Jing Li, Jay Yang, Larry H. Matherly, Sergej Naumovich Konoplev, Jeffrey W. Taub, Marina Konopleva, Yubin Ge, Natalia Baran. ME-344, a novel isoflavone mitochondrial inhibitor, in combination with venetoclax constitutes a new metabolism-targeted approach to overcome resistance to Bcl-2 inhibition and standard of care treatment in AML [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3785.
- Published
- 2022
- Full Text
- View/download PDF
18. Fluorine-Substituted Pyrrolo[2,3-d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis
- Author
-
Adrianne Wallace-Povirk, Mohammad A. Karim, Juiwanna Kushner, Larry H. Matherly, Kathryn White, Manasa P. Ravindra, Carrie O'Connor, Nian Tong, Lisa Polin, Aleem Gangjee, Mike R. Wilson, and Zhanjun Hou
- Subjects
0301 basic medicine ,Antineoplastic Agents ,CHO Cells ,HeLa ,Mice ,03 medical and health sciences ,Pyrimidine analogue ,Cricetulus ,Folic Acid ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Folate Receptor 1 ,Receptor ,Purine Nucleotides ,Cell Proliferation ,biology ,Chemistry ,Chinese hamster ovary cell ,Biological Transport ,Transporter ,Fluorine ,biology.organism_classification ,Xenograft Model Antitumor Assays ,In vitro ,Pyrimidines ,030104 developmental biology ,Biochemistry ,Folate receptor ,030220 oncology & carcinogenesis ,Molecular Medicine ,Proton-Coupled Folate Transporter - Abstract
Novel fluorinated 2-amino-4-oxo-6-substituted pyrrolo[2,3- d]pyrimidine analogues 7-12 were synthesized and tested for selective cellular uptake by folate receptors (FRs) α and β or the proton-coupled folate transporter (PCFT) and for antitumor efficacy. Compounds 8, 9, 11, and 12 showed increased in vitro antiproliferative activities (∼11-fold) over the nonfluorinated analogues 2, 3, 5, and 6 toward engineered Chinese hamster ovary and HeLa cells expressing FRs or PCFT. Compounds 8, 9, 11, and 12 also inhibited proliferation of IGROV1 and A2780 epithelial ovarian cancer cells; in IGROV1 cells with knockdown of FRα, 9, 11, and 12 showed sustained inhibition associated with uptake by PCFT. All compounds inhibited glycinamide ribonucleotide formyltransferase, a key enzyme in the de novo purine biosynthesis pathway. Molecular modeling studies validated in vitro cell-based results. NMR evidence supports the presence of an intramolecular fluorine-hydrogen bond. Potent in vivo efficacy of 11 was established with IGROV1 xenografts in severe compromised immunodeficient mice.
- Published
- 2018
- Full Text
- View/download PDF
19. Folate Transport and One-Carbon Metabolism in Targeted Therapies of Epithelial Ovarian Cancer
- Author
-
Adrianne Wallace-Povirk, Zhanjun Hou, Md. Junayed Nayeen, Aleem Gangjee, and Larry H. Matherly
- Subjects
epithelial ovarian cancer ,Cancer Research ,endocrine system diseases ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Review ,proton-coupled folate transporter ,folate ,one-carbon metabolism ,female genital diseases and pregnancy complications ,folate receptor ,Oncology ,tumor microenvironment ,folate transport ,RC254-282 - Abstract
Simple Summary New therapies are urgently needed for ovarian cancer, the most lethal malignancy in women. To identify new approaches for targeting ovarian cancer, metabolic vulnerabilities must be discovered and strategies for the selective delivery of therapeutic agents must be established. New approaches that are tumor-selective and that facilitate the internalization of novel drugs or provide targets for therapy are being developed for treating ovarian cancer involving folate receptors and the proton-coupled folate transporter. New drugs are being discovered that target key metabolic processes in tumors and neighboring immune cells which contribute to tumor progression. In this review, we describe the remarkable advances in this rapidly evolving area and their extraordinary potential to improve the lives of women diagnosed with this devastating disease. Abstract New therapies are urgently needed for epithelial ovarian cancer (EOC), the most lethal gynecologic malignancy. To identify new approaches for targeting EOC, metabolic vulnerabilities must be discovered and strategies for the selective delivery of therapeutic agents must be established. Folate receptor (FR) α and the proton-coupled folate transporter (PCFT) are expressed in the majority of EOCs. FRβ is expressed on tumor-associated macrophages, a major infiltrating immune population in EOC. One-carbon (C1) metabolism is partitioned between the cytosol and mitochondria and is important for the synthesis of nucleotides, amino acids, glutathione, and other critical metabolites. Novel inhibitors are being developed with the potential for therapeutic targeting of tumors via FRs and the PCFT, as well as for inhibiting C1 metabolism. In this review, we summarize these exciting new developments in targeted therapies for both tumors and the tumor microenvironment in EOC.
- Published
- 2021
- Full Text
- View/download PDF
20. Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3-d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis
- Author
-
Aleem Gangjee, Christina George, Juiwanna Kushner, Adrianne Wallace-Povirk, Carrie O'Connor, Lisa Polin, Mike R. Wilson, Zhanjun Hou, Manasa P. Ravindra, Mohammad A. Karim, Kathryn White, and Larry H. Matherly
- Subjects
0301 basic medicine ,Pyrimidine ,Antineoplastic Agents ,CHO Cells ,Mice, SCID ,Article ,Mice ,03 medical and health sciences ,Pyrimidine analogue ,chemistry.chemical_compound ,Cricetulus ,0302 clinical medicine ,In vivo ,Drug Discovery ,Animals ,Humans ,Folate Receptor 1 ,Pyrroles ,Folate Receptor 2 ,Receptor ,Purine Nucleotides ,IC50 ,Cell Proliferation ,biology ,Chemistry ,Chinese hamster ovary cell ,biology.organism_classification ,Pyrimidines ,030104 developmental biology ,Biochemistry ,Folate receptor ,030220 oncology & carcinogenesis ,Folic Acid Antagonists ,Heterografts ,Molecular Medicine ,Proton-Coupled Folate Transporter - Abstract
Tumor-targeted specificities of 6-substituted pyrrolo-[2,3-d]pyrimidine analogues of 1, where the phenyl side-chain is replaced by 3′,6′ (5, 8), 2′,5′ (6, 9), and 2′,6′ (7, 10) pyridyls, were analyzed. Proliferation inhibition of isogenic Chinese hamster ovary (CHO) cells expressing folate receptors (FRs) α and β were in rank order, 6 > 9 > 5 > 7 > 8, with 10 showing no activity, and 6 > 9 > 5 > 8, with 10 and 7 being inactive, respectively. Antiproliferative effects toward FRα- and FRβ-expressing cells were reflected in competitive binding with [(3)H]folic acid. Only compound 6 was active against proton-coupled folate receptor (PCFT)-expressing CHO cells (~4-fold more potent than 1) and inhibited [(3)H]methotrexate uptake by PCFT. In KB and IGROV1 tumor cells, 6 showed
- Published
- 2018
- Full Text
- View/download PDF
21. A Novel Isoflavone, ME-344, Enhances Venetoclax Antileukemic Activity Against AML Via Suppression of Oxidative Phosphorylation and Purine Biosynthesis
- Author
-
Jeffrey W. Taub, Jing Li, Katie Hege Hurrish, Jay Yang, Jenna L. Carter, Zhanjun Hou, Yongwei Su, Yubin Ge, Lisa Polin, Hasini A. Kalpage, Holly Edwards, Maik Hüttemann, Larry H. Matherly, and Sandra E. Wiley
- Subjects
chemistry.chemical_compound ,Biochemistry ,Chemistry ,Venetoclax ,Immunology ,Cell Biology ,Hematology ,Oxidative phosphorylation ,Purine metabolism - Abstract
The 5-year survival rate for adult patients with acute myeloid leukemia (AML) treated with cytarabine-based chemotherapy remains less than 30%, due to drug resistance and disease relapse. Recently, a selective inhibitor of anti-apoptotic Bcl-2, venetoclax, was approved by the FDA in combination with low dose cytarabine or hypomethylating agents for treating newly diagnosed AML patients who are 75 years of age or older or for those who are unfit for standard chemotherapy, providing more treatment options for this group of patients. Although the response rate to these newly approved combination therapies is reported to be 70%, the median overall survival is only 10-18 months showing that the duration of response is limited. Therefore, novel therapeutic agents are in demand to enhance venetoclax activity against AML and to combat AML resistant to cytarabine-based chemotherapy. Cytarabine-resistant AML cells lead to relapse and rely on oxidative phosphorylation (OXPHOS) for survival. In addition, it has been reported that targeting OXPHOS can enhance venetoclax activity against preclinical models of AML. Thus, we hypothesize that OXPHOS suppressing agents would be good candidates to combine with and enhance venetoclax antileukemic activity against newly diagnosed AML and those with resistance to cytarabine. A novel isoflavone, ME-344, has been shown to suppress OXPHOS in cell lines derived from solid tumors by inhibiting Complex I of the electron transport chain. We hypothesized that combining ME-344 with venetoclax would result in synergistic antileukemic activity against AML. Consistent with our hypothesis, combining ME-344 with venetoclax resulted in synergistic induction of apoptosis in AML cell lines, including those with acquired cytarabine resistance. The combination of these two agents also resulted in synergistic antileukemic activity in one primary AML patient sample, as determined by MTT assay. The combination of ME-344 and venetoclax prolonged the median survival of MV4-11 leukemia- bearing NSGS mice by 37% (median survival of 48 days compared to 35 days for vehicle control treated mice, n=5 per arm, p Disclosures Wiley: MEIPharma: Current Employment.
- Published
- 2021
- Full Text
- View/download PDF
22. Role of proton-coupled folate transporter in pemetrexed resistance of mesothelioma: clinical evidence and new pharmacological tools
- Author
-
Michal Stark, Elisa Giovannetti, Gerrit Jansen, Larry H. Matherly, Maria Gemelli, Godefridus J. Peters, Erik Thunnissen, Ittai B. Muller, Zhanjun Hou, P. A. Zucali, E. A. Struys, Niccola Funel, M. Perrino, Yehuda G. Assaraf, Medical oncology laboratory, CCA - Cancer biology and immunology, Pathology, Clinical chemistry, AGEM - Inborn errors of metabolism, AGEM - Endocrinology, metabolism and nutrition, Rheumatology, and AGEM - Digestive immunity
- Subjects
Male ,Mesothelioma ,0301 basic medicine ,Oncology ,Thymidylate synthase ,Immunoenzyme Techniques ,Reduced Folate Carrier Protein ,chemistry.chemical_compound ,0302 clinical medicine ,Tumor Cells, Cultured ,Aged, 80 and over ,biology ,Hematology ,Methylation ,Middle Aged ,Prognosis ,Survival Rate ,Pemetrexed ,030220 oncology & carcinogenesis ,Antifolate ,Immunohistochemistry ,Female ,Proton-Coupled Folate Transporter ,medicine.drug ,Adult ,medicine.medical_specialty ,Pleural Neoplasms ,03 medical and health sciences ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Gene silencing ,Survival rate ,Aged ,Cell Proliferation ,business.industry ,Original Articles ,Thymidylate Synthase ,medicine.disease ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,biology.protein ,Folic Acid Antagonists ,business ,Follow-Up Studies - Abstract
Background Thymidylate synthase (TS) has a predictive role in pemetrexed treatment of mesothelioma; however, additional chemoresistance mechanisms are poorly understood. Here, we explored the role of the reduced-folate carrier (RFC/SLC19A1) and proton-coupled folate transporter (PCFT/SLC46A1) in antifolate resistance in mesothelioma. Patients and methods PCFT,RFC andTS RNA and PCFT protein levels were determined by quantitative RT-PCR of frozen tissues and immunohistochemistry of tissue-microarrays, respectively, in two cohorts of pemetrexed-treated patients. Data were analyzed byt-test, Fisher’s/log-rank test and Cox proportional models. The contribution of PCFT expression andPCFT-promoter methylation to pemetrexed activity were evaluated in mesothelioma cells and spheroids, through 5-aza-2′-deoxycytidine-mediated demethylation and siRNA-knockdown. Results Pemetrexed-treated patients with lowPCFT had significantly lower rates of disease control, and shorter overall survival (OS), in both the test (N = 73, 11.3 versus 20.1 months,P = 0.01) and validation (N = 51, 12.6 versus 30.3 months,P = 0.02) cohorts. Multivariate analysis confirmedPCFT-independent prognostic role. Low-PCFT protein levels were also associated with shorter OS. Patients with both low-PCFT and high-TS levels had the worst prognosis (OS, 5.5 months), whereas associations were neither found forRFC nor in pemetrexed-untreated patients.PCFT silencing reduced pemetrexed sensitivity, whereas 5-aza-2′-deoxycytidine overcame resistance. Conclusions These findings identify for the first time PCFT as a novel mesothelioma prognostic biomarker, prompting prospective trials for its validation. Moreover, preclinical data suggest that targetingPCFT-promoter methylation might eradicate pemetrexed-resistant cells characterized by low-PCFT expression.
- Published
- 2017
- Full Text
- View/download PDF
23. Dual Targeting of Epithelial Ovarian Cancer Via Folate Receptor α and the Proton-Coupled Folate Transporter with 6-Substituted Pyrrolo[2,3-d]pyrimidine Antifolates
- Author
-
Leda Gattoc, Aleem Gangjee, Juiwanna Kushner, Carrie O'Connor, Christina George, Si Yang, Larry H. Matherly, Adrianne Wallace-Povirk, Steve Orr, Kathryn White, Robert T. Morris, Zhanjun Hou, and Lisa Polin
- Subjects
0301 basic medicine ,Cancer Research ,Carcinoma, Ovarian Epithelial ,Biology ,Article ,Mice ,03 medical and health sciences ,Folic Acid ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Folate Receptor 1 ,Neoplasms, Glandular and Epithelial ,Cell Proliferation ,Ovarian Neoplasms ,Cisplatin ,Gene knockdown ,Cancer ,Transporter ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,030104 developmental biology ,Cell killing ,Oncology ,Biochemistry ,Drug Resistance, Neoplasm ,Folate receptor ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Folic Acid Antagonists ,Proton-Coupled Folate Transporter ,medicine.drug - Abstract
Folate uptake in epithelial ovarian cancer (EOC) involves the reduced folate carrier (RFC) and the proton-coupled folate transporter (PCFT), both facilitative transporters and folate receptor (FR) α. Although in primary EOC specimens, FRα is widely expressed and increases with tumor stage, PCFT was expressed independent of tumor stage (by real-time RT-PCR and IHC). EOC cell line models, including cisplatin sensitive (IGROV1 and A2780) and resistant (SKOV3 and TOV112D) cells, expressed a 17-fold range of FRα and similar amounts (within ∼2-fold) of PCFT. Novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates AGF94 and AGF154 exhibited potent antiproliferative activities toward all of the EOC cell lines, reflecting selective cellular uptake by FRα and/or PCFT over RFC. When IGROV1 cells were pretreated with AGF94 at pH 6.8, clonogenicity was potently inhibited, confirming cell killing. FRα was knocked down in IGROV1 cells with lentiviral shRNAs. Two FRα knockdown clones (KD-4 and KD-10) showed markedly reduced binding and uptake of [3H]folic acid and [3H]AGF154 by FRα, but maintained high levels of [3H]AGF154 uptake by PCFT compared to nontargeted control cells. In proliferation assays, KD-4 and KD-10 cells preserved in vitro inhibition by AGF94 and AGF154, compared to a nontargeted control, attributable to residual FRα- and substantial PCFT-mediated uptake. KD-10 tumor xenografts in severe-compromised immune-deficient mice were likewise sensitive to AGF94. Collectively, our results demonstrate the substantial therapeutic potential of novel 6-substituted pyrrolo[2,3-d]pyrimidine antifolates with dual targeting of PCFT and FRα toward EOCs that express a range of FRα, along with PCFT, as well as cisplatin resistance. Mol Cancer Ther; 16(5); 819–30. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
24. Abstract 2348: Targeting mitochondrial and cytosolic one-carbon metabolism in epithelial ovarian cancer via folate receptor alpha
- Author
-
Jennifer Wong-Roushar, Aamod Dekhne, Charles E. Dann, Lisa Polin, Xun Bao, Seongho Kim, Jade M. Katinas, Adrianne Wallace-Povirk, Jose Cardiel Nunez, Md. Junayed Nayeen, Khushbu Shah, Zhanjun Hou, Aleem Gangjee, Carrie O'Connor, Jing Li, and Larry Matherly
- Subjects
Folate Receptor Alpha ,Cancer Research ,One-carbon metabolism ,Cytosol ,Oncology ,Chemistry ,Cancer research ,Epithelial ovarian cancer - Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy. Though most patients initially respond to platinum-based therapy, the likelihood of disease reoccurrence is nearly 100%. Thus, new tumor-selective therapies for EOC are urgently needed. One such treatment option involves targeting tumors via folate receptor α (FRα), which is overexpressed in up to 90% of EOCs and shows increasing expression with higher stage and grade of disease. Our laboratory discovered novel 5-substituted pyrrolo[3,2-d]pyrimidine analogs (AGF347, AGF359, AGF362 and AGF363) that inhibit mitochondrial one-carbon (C1) metabolism at serine hydroxymethyltransferase (SHMT) 2, with secondary inhibition at cytosolic enzyme targets including those in de novo purine biosynthesis. Potent inhibition was seen with several FRα-expressing EOC tumor cells. Inhibitory potencies were in order, AGF347 > AGF359 > AGF362 > AGF363. Drug effects were substantially reduced with excess folic acid (FA), confirming FRα-mediated drug uptake. Toward cisplatin resistant SKOV3, TOV112D and A2780 EOC cells, inhibition in the nanomolar range was detected with all compounds. Targeted metabolomics, using L-[2,3,3-2H]serine as a tracer in wild-type or SHMT2 knockdown SKOV3 cells, confirmed all compounds inhibited cytosolic and mitochondrial C1-metabolism (at SHMT2). Apoptosis was detected for all compounds by Annexin V/PI, with partial rescue of apoptosis observed upon addition of glutathione (GSH). Glutathione pools (GSH and total GSH+GSSG) were significantly perturbed by drug treatment with all inhibitors in SKOV3 cells, comparable to GSH levels observed in SHMT2 KD cells. In vivo efficacy studies with SKOV3 xenografts treated with either AGF347 or cisplatin in SCID mice showed cisplatin resistance, while AGF347 demonstrated efficacy and delay in disease progression with a median tumor growth delay of 10 days, with the longest delay being 15 days. Our studies describe a series of novel inhibitors targeting mitochondrial and cytosolic C1-metabolism, selectively delivered via FRα, which show direct cytotoxic effects against cisplatin resistant EOC in vitro and in vivo, and display additional mechanisms of cytotoxicity mediated through glycine depletion. Citation Format: Adrianne C. Wallace-Povirk, Carrie O'Connor, Xun Bao, Jade Katinas, Jennifer Wong-Roushar, Aamod Dekhne, Zhanjun Hou, Md. Junayed Nayeen, Khushbu Shah, Jose Cardiel Nunez, Jing Li, Seongho Kim, Lisa Polin, Charles E. Dann, Aleem Gangjee, Larry H. Matherly. Targeting mitochondrial and cytosolic one-carbon metabolism in epithelial ovarian cancer via folate receptor alpha [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2348.
- Published
- 2021
- Full Text
- View/download PDF
25. Functional and mechanistic roles of the human proton-coupled folate transporter transmembrane domain 6–7 linker
- Author
-
Zhanjun Hou, Mike R. Wilson, Larry H. Matherly, Lucas J. Wilson, and Jun Ye
- Subjects
0301 basic medicine ,Blotting, Western ,Mutant ,Biotin ,Biochemistry ,Article ,Protein Structure, Secondary ,03 medical and health sciences ,Folic Acid ,Thiamine transporter ,Humans ,Biotinylation ,Amino Acid Sequence ,Molecular Biology ,Protein secondary structure ,Alanine ,chemistry.chemical_classification ,Microscopy, Confocal ,biology ,Membrane Proteins ,Biological Transport ,Cell Biology ,Fusion protein ,Cell biology ,Amino acid ,Transmembrane domain ,030104 developmental biology ,chemistry ,Mutagenesis ,Symporter ,biology.protein ,Proton-Coupled Folate Transporter ,HeLa Cells - Abstract
The proton-coupled folate transporter (PCFT; SLC46A1) is a folate–proton symporter expressed in solid tumors and is used for tumor-targeted delivery of cytotoxic antifolates. Topology modeling suggests that the PCFT secondary structure includes 12 transmembrane domains (TMDs) with TMDs 6 and 7 linked by an intracellular loop (positions 236–265) including His247, implicated as functionally important. Single-cysteine (Cys) mutants were inserted from positions 241 to 251 in Cys-less PCFT and mutant proteins were expressed in PCFT-null (R1-11) HeLa cells; none were reactive with 2-aminoethyl methanethiosulfonate biotin, suggesting that the TMD6–7 loop is intracellular. Twenty-nine single alanine mutants spanning the entire TMD6–7 loop were expressed in R1-11 cells; activity was generally preserved, with the exception of the 247, 250, and 251 mutants, partly due to decreased surface expression. Coexpression of PCFT TMD1–6 and TMD7–12 half-molecules in R1-11 cells partially restored transport activity, although removal of residues 252–265 from TMD7–12 abolished transport. Chimeric proteins, including a nonhomologous sequence from a thiamine transporter (ThTr1) inserted into the PCFT TMD6–7 loop (positions 236–250 or 251–265), were active, although replacement of the entire loop with the ThTr1 sequence resulted in substantial loss of activity. Amino acid replacements (Ala, Arg, His, Gln, and Glu) or deletions at position 247 in wild-type and PCFT–ThTr1 chimeras resulted in differential effects on transport. Collectively, our findings suggest that the PCFT TMD6–7 connecting loop confers protein stability and may serve a unique functional role that depends on secondary structure rather than particular sequence elements.
- Published
- 2016
- Full Text
- View/download PDF
26. Targeting one-carbon metabolism in syngeneic mouse model of BRCA-mutated high-grade serous ovarian cancer
- Author
-
Adrianne Wallace-Povirk, Larry H. Matherly, R.T. Morris, Carrie O'Connor, Zhanjun Hou, Aleem Gangjee, and Lisa Rubinsak
- Subjects
One-carbon metabolism ,Oncology ,business.industry ,Cancer research ,Serous ovarian cancer ,Obstetrics and Gynecology ,Medicine ,Syngeneic mouse ,business - Published
- 2020
- Full Text
- View/download PDF
27. Abstract 511: Targeted one-carbon (1C) metabolism inhibitors: Design, synthesis and biological evaluation of novel 5-substituted pyrrolo[3,2-d]pyrimidines with pyridyl- and ortho-fluoropyridyl glutamate side chains as selective folate receptors substrates
- Author
-
Zhanjun Hou, Arpit Doshi, Larry Matherly, Aleem Gangjee, Adrianne Wallace-Povirk, and Carrie O'Connor
- Subjects
Cancer Research ,Oncology ,Design synthesis ,Chemistry ,Glutamate receptor ,Side chain ,chemistry.chemical_element ,Metabolism ,Receptor ,Carbon ,Combinatorial chemistry ,Biological evaluation - Abstract
One-carbon (1C) metabolism, in tumor cells, provides basic metabolites needed for proliferation and is compartmentalized into the cytosol and mitochondria. The key enzymes in 1C metabolism are de novo purine biosynthesis catalyzing enzymes, glycinamide ribonucleotide formyl transferase (GARFTase) and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFTase) and mitochondrial serine hydroxymethyltransferase 2 (SHMT2) involved in glycine synthesis and maintaining 1C pools. We have previously designed and evaluated a series of multitargeted 5-substituted pyrrolo[3,2-d]pyrimidine antifolates with a phenyl glutamate side-chain (AGF291, AGF300 and AGF299), which inhibited KB tumor cell growth. AGF291 inhibited the enzymes crucial for both cytosolic and mitochondrial 1C metabolism. However, these compounds also were transported via the ubiquitously expressed reduced folate carrier (RFC) which can lead to dose-limiting toxicities as seen for classical antifolates such as pemetrexed (PMX). In this study, we designed conformationally restricted 5-substituted pyrrolo[3,2-d]pyrimidine antifolates with pyridyl glutamate side-chains (AGF363, AGF369 and AGF370) and ortho-fluoropyridyl glutamate side-chains (AGF377 and AGF379) to decrease normal cell uptake by the ubiquitously present RFC and maintain or increase transport via tumor-selective folate receptor α (FRα) and folate receptor β (FRβ). Replacing the phenyl with the pyridyl side-chain abrogated the transport via RFC, thereby improved (AGF363 vs AGF291) or maintained (AGF369 vs AGF300 and AGF370 vs AGF299) selective uptake by FRα and FRβ over RFC and showed potent KB tumor cell inhibition (AGF363, IC50 = 10.78 nM and AGF369, IC50 = 6.97 nM). The ortho-fluorinated pyridyl analogs were also selective for FRα and FRβ over RFC but showed moderate potency towards KB tumor cells. Overall, this study identified two key pharmacophores, i.e. the pyridyl and the o-fluoropyridyl side-chains, to obtain absolute selectivity for FRα and FRβ over RFC in the 5-substituted pyrrolo[3,2-d]pyrimidine series of 1Cmetabolism inhibitors. Further preclinical studies are underway to afford viable candidates for future clinical application of selective compounds without toxicities as an improvement of currently used agents. Citation Format: Arpit Doshi, Adrianne Wallace-Povirk, Carrie O'Connor, Zhanjun Hou, Larry Matherly, Aleem Gangjee. Targeted one-carbon (1C) metabolism inhibitors: Design, synthesis and biological evaluation of novel 5-substituted pyrrolo[3,2-d]pyrimidines with pyridyl- and ortho-fluoropyridyl glutamate side chains as selective folate receptors substrates [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 511.
- Published
- 2020
- Full Text
- View/download PDF
28. Regulation of differential proton-coupled folate transporter gene expression in human tumors: transactivation by KLF15 with NRF-1 and the role of Sp1
- Author
-
Aleem Gangjee, Zhanjun Hou, Steve Orr, Carrie O'Connor, Larry H. Matherly, Seongho Kim, and Josephine Frühauf
- Subjects
Sp1 Transcription Factor ,Kruppel-Like Transcription Factors ,Response Elements ,Biochemistry ,Article ,Transactivation ,Neoplasms ,Gene expression ,Transcriptional regulation ,Animals ,Humans ,Electrophoretic mobility shift assay ,Molecular Biology ,Regulation of gene expression ,Reporter gene ,Chemistry ,Nuclear Respiratory Factor 1 ,Cell Biology ,Transfection ,Hep G2 Cells ,Cell biology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Drosophila melanogaster ,Chromatin immunoprecipitation ,Proton-Coupled Folate Transporter ,HeLa Cells - Abstract
Tumors can be therapeutically targeted with novel antifolates (e.g. AGF94) that are selectively transported by the human proton-coupled folate transporter (hPCFT). Studies were performed to determine the transcription regulation of hPCFT in tumors and identify possible mechanisms that contribute to the highly disparate levels of hPCFT in HepG2 versus HT1080 tumor cells. Transfection of hPCFT-null HT1080 cells with hPCFT restored transport and sensitivity to AGF94. Progressive deletions of the hPCFT promoter construct (−2005 to +96) and reporter gene assays in HepG2 and HT1080 cells confirmed differences in hPCFT transactivation and localized a minimal promoter to between positions −50 and +96. The minimal promoter included KLF15, GC-Box and NRF-1 cis-binding elements whose functional importance was confirmed by promoter deletions and mutations of core consensus sequences and reporter gene assays. In HepG2 cells, NRF-1, KLF15 and Sp1 transcripts were increased over HT1080 cells by ∼5.1-, ∼44-, and ∼2.4-fold, respectively. In Drosophila SL2 cells, transfection with KLF15 and NRF-1 synergistically activated the hPCFT promoter; Sp1 was modestly activating or inhibitory. Chromatin immunoprecipitation and electrophoretic mobility shift assay (EMSA) and supershifts confirmed differential binding of KLF15, Sp1, and NRF-1 to the hPCFT promoter in HepG2 and HT1080 cells that paralleled hPCFT levels. Treatment of HT1080 nuclear extracts (NE) with protein kinase A increased Sp1 binding to its consensus sequence by EMSA, suggesting a role for Sp1 phosphorylation in regulating hPCFT transcription. A better understanding of determinants of hPCFT transcriptional control may identify new therapeutic strategies for cancer by modulating hPCFT levels in combination with hPCFT-targeted antifolates.
- Published
- 2018
29. Targeting Nonsquamous Nonsmall Cell Lung Cancer via the Proton-Coupled Folate Transporter with 6-Substituted Pyrrolo[2,3-d]Pyrimidine Thienoyl Antifolates
- Author
-
Si Yang, Mike R. Wilson, Larry H. Matherly, Jenny Huang, Manohar Ratnam, Lisa Polin, Aleem Gangjee, Juiwanna Kushner, Kathryn White, and Zhanjun Hou
- Subjects
0301 basic medicine ,Lung Neoplasms ,Pyrimidine ,Antineoplastic Agents ,Mice, SCID ,Biology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,0302 clinical medicine ,In vivo ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Animals ,Humans ,Pharmacology ,Mice, Inbred ICR ,Gene knockdown ,Tumor microenvironment ,Dose-Response Relationship, Drug ,Transporter ,Articles ,Xenograft Model Antitumor Assays ,Molecular biology ,respiratory tract diseases ,Blot ,Pyrimidines ,030104 developmental biology ,Biochemistry ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Antifolate ,Folic Acid Antagonists ,Molecular Medicine ,Female ,Proton-Coupled Folate Transporter - Abstract
Pemetrexed (PMX) is a 5-substituted pyrrolo[2,3-d]pyrimidine antifolate used for therapy of nonsquamous nonsmall cell lung cancer (NS-NSCLC). PMX is transported by the reduced folate carrier (RFC) and proton-coupled folate transporter (PCFT). Unlike RFC, PCFT is active at acidic pH levels characterizing the tumor microenvironment. By real-time reverse-transcription polymerase chain reaction (RT-PCR) and immunohistochemistry, PCFT transcripts and proteins were detected in primary NS-NSCLC specimens. In six NS-NSCLC cell lines (A549, H1437, H460, H1299, H1650, and H2030), PCFT transcripts and proteins were detected by real-time RT-PCR and western blots, respectively. 6-Substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates related to PMX [compound 1 (C1) and compound 2 (C2), respectively] are selective substrates for PCFT over RFC. In the NS-NSCLC cell lines, both [(3)H]PMX and [(3)H]C2 were transported by PCFT. C1 and C2 inhibited proliferation of the NS-NSCLC cell lines; A549, H460, and H2030 cells were more sensitive to C1 than to PMX. C1 and C2 inhibited glycinamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis. When treated at pH 6.8, which favors PCFT uptake, C1 and C2 inhibited clonogenicity of H460 cells greater than PMX; PMX inhibited clonogenicity more than C1 or C2 at pH 7.2, which favors RFC transport over PCFT. Knockdown of PCFT in H460 cells resulted in decreased [(3)H]PMX and [(3)H]C2 transport and decreased growth inhibition by C1 and C2, and to a lesser extent by PMX. In vivo efficacy of C1 was seen toward H460 tumor xenografts in severe-combined immunodeficient mice. Our results suggest that 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates offer significant promise for treating NS-NSCLC by selective uptake by PCFT.
- Published
- 2016
- Full Text
- View/download PDF
30. Novel 5-Substituted Pyrrolo[2,3-d]pyrimidines as Dual Inhibitors of Glycinamide Ribonucleotide Formyltransferase and 5-Aminoimidazole-4-carboxamide Ribonucleotide Formyltransferase and as Potential Antitumor Agents
- Author
-
Steven Orr, Sudhir Raghavan, Zhanjun Hou, Larry H. Matherly, Aleem Gangjee, Christina George, Yiqiang Wang, and Shermaine Mitchell-Ryan
- Subjects
Phosphoribosylglycinamide formyltransferase ,Purine ,Ribonucleotide ,Stereochemistry ,Antineoplastic Agents ,CHO Cells ,Thymidylate synthase ,KB Cells ,Phosphoribosylaminoimidazolecarboxamide Formyltransferase ,Article ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cricetulus ,Drug Discovery ,medicine ,Animals ,Humans ,Pyrroles ,Nucleotide ,Enzyme Inhibitors ,Cells, Cultured ,Cell Proliferation ,Phosphoribosylglycinamide Formyltransferase ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,Phosphoribosylaminoimidazolecarboxamide formyltransferase ,biology ,Chemistry ,Pyrimidines ,Pemetrexed ,Biochemistry ,Folate receptor ,biology.protein ,Molecular Medicine ,Drug Screening Assays, Antitumor ,medicine.drug - Abstract
A new series of 5-substituted thiopheneyl pyrrolo[2,3-d]pyrimidines 6–11 with varying chain lengths (n = 1–6) were designed and synthesized as hybrids of the clinically used anticancer drug pemetrexed (PMX) and our 6-substituted thiopheneyl pyrrolo[2,3-d]pyrimidines 2c and 2d with folate receptor (FR) α and proton-coupled folate transporter (PCFT) uptake specificity over the reduced folate carrier (RFC) and inhibition of de novo purine nucleotide biosynthesis at glycinamide ribonucleotide formyltransferase (GARFTase). Compounds 6–11 inhibited KB human tumor cells in the order 9 = 10 > 8 > 7 > 6 = 11. Compounds 8–10 were variously transported by FRα, PCFT, and RFC and, unlike PMX, inhibited de novo purine nucleotide rather than thymidylate biosynthesis. The antiproliferative effects of 8 and 9 appeared to be due to their dual inhibitions of both GARFTase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase. Our studies identify a unique structure–activity relationship for transport and dual target inhibition.
- Published
- 2015
- Full Text
- View/download PDF
31. Structure–Activity Profiles of Novel 6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Antifolates with Modified Amino Acids for Cellular Uptake by Folate Receptors α and β and the Proton-Coupled Folate Transporter
- Author
-
Adrianne Wallace, Aleem Gangjee, Christina George, Sai Zhao, Sudhir Raghavan, Steven Orr, Mike R. Wilson, Larry H. Matherly, Lalit K. Golani, and Zhanjun Hou
- Subjects
Models, Molecular ,Stereochemistry ,CHO Cells ,KB Cells ,Structure-Activity Relationship ,Cricetulus ,Cricetinae ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Folate Receptor 1 ,Folate Receptor 2 ,chemistry.chemical_classification ,Chemistry ,Chinese hamster ovary cell ,Biological activity ,Addition/Correction ,3. Good health ,Amino acid ,Pyrimidines ,Biochemistry ,Docking (molecular) ,Folate receptor ,Folic Acid Antagonists ,Molecular Medicine ,Folate receptor 1 ,Folic Acid Transporters - Abstract
Structure-activity relationships for cellular uptake and inhibition of cell proliferation were studied for 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates in which the terminal l-glutamate of the parent structure (7) was replaced by natural or unnatural amino acids. Compounds 7 and 10-13 were selectively inhibitory toward folate receptor (FR) α-expressing Chinese hamster ovary (CHO) cells. Antiproliferative effects of compounds 7 and 9-13 toward FRα- and FRβ-expressing CHO cells were only partly reflected in binding affinities to FRα and FRβ or in the docking scores with molecular models of FRα and FRβ. Compounds 7 and 11 were potent inhibitors of glycinamide ribonucleotide formyltransferase in de novo purine biosynthesis in KB human tumor cells. These studies establish for the first time the importance of the α- and γ-carboxylic acid groups, the length of the amino acid, and the conformation of the side chain for transporter binding and biological activity of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates.
- Published
- 2014
- Full Text
- View/download PDF
32. Substituted Cysteine Accessibility Reveals a Novel Transmembrane 2–3 Reentrant Loop and Functional Role for Transmembrane Domain 2 in the Human Proton-coupled Folate Transporter
- Author
-
Larry H. Matherly, Zhanjun Hou, and Mike R. Wilson
- Subjects
Guanine ,Blotting, Western ,Molecular Sequence Data ,Methotrexate transport ,Pemetrexed ,Tritium ,Biochemistry ,Protein Structure, Secondary ,Folic Acid ,Protein structure ,Glutamates ,Membrane Biology ,Humans ,Biotinylation ,Amino Acid Sequence ,Cysteine ,Binding site ,Molecular Biology ,Binding Sites ,Chemistry ,Cell Membrane ,Biological Transport ,Cell Biology ,Transmembrane protein ,Kinetics ,Transmembrane domain ,Methotrexate ,Amino Acid Substitution ,Membrane topology ,Mutation ,Symporter ,Folic Acid Antagonists ,Proton-Coupled Folate Transporter ,HeLa Cells - Abstract
The proton-coupled folate transporter (PCFT) is a folate-proton symporter highly expressed in solid tumors that can selectively target cytotoxic antifolates to tumors under acidic microenvironment conditions. Predicted topology models for PCFT suggest that the loop domain between transmembrane domains (TMDs) 2 and 3 resides in the cytosol. Mutations involving Asp-109 or Arg-113 in the TMD2-3 loop result in loss of activity. By structural homology to other solute carriers, TMD2 may form part of the PCFT substrate binding domain. In this study we mutated the seven cysteine (Cys) residues of human PCFT to serine, creating Cys-less PCFT. Thirty-three single-Cys mutants spanning TMD2 and the TMD2-3 loop in a Cys-less PCFT background were transfected into PCFT-null HeLa cells. All 33 mutants were detected by Western blotting, and 28 were active for [(3)H]methotrexate uptake at pH 5.5. For the active residues, we performed pulldown assays with membrane-impermeable 2-aminoethyl methanethiosulfonate-biotin and streptavidin beads to determine their aqueous-accessibilities. Multiple residues in TMD2 and the TMD2-3 loop domain reacted with 2-aminoethyl methanethiosulfonate-biotin, establishing aqueous accessibilities. Pemetrexed pretreatment inhibited biotinylation of TMD2 mutants G93C and F94C, and biotinylation of these residues inhibited methotrexate transport activity. Our results suggest that the TMD 2-3 loop domain is aqueous-accessible and forms a novel reentrant loop structure. Residues in TMD2 form an aqueous transmembrane pathway for folate substrates, and Gly-93 and Phe-94 may contribute to a substrate binding domain. Characterization of PCFT structure is essential to understanding the transport mechanism including the critical determinants of substrate binding.
- Published
- 2014
- Full Text
- View/download PDF
33. Discovery of amide-bridged pyrrolo[2,3-d]pyrimidines as tumor targeted classical antifolates with selective uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis
- Author
-
Aleem Gangjee, Weiguo Xiang, Aamod Dekhne, Zhanjun Hou, Larry H. Matherly, Arpit Doshi, and Carrie O'Connor
- Subjects
Models, Molecular ,Purine ,Pyrimidine ,Pyridines ,Stereochemistry ,Clinical Biochemistry ,AICA ribonucleotide ,Pharmaceutical Science ,Antineoplastic Agents ,CHO Cells ,01 natural sciences ,Biochemistry ,Article ,chemistry.chemical_compound ,Cricetulus ,Cell Line, Tumor ,Neoplasms ,Amide ,Drug Discovery ,medicine ,Animals ,Humans ,Folate Receptor 1 ,Pyrroles ,Purine Nucleotides ,Molecular Biology ,010405 organic chemistry ,Organic Chemistry ,Ligand (biochemistry) ,Amides ,Adenosine ,Biosynthetic Pathways ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry ,Folate receptor ,Antifolate ,Folic Acid Antagonists ,Molecular Medicine ,medicine.drug - Abstract
We previously showed that classical 6-substituted pyrrolo[2,3-d]pyrimidine antifolates bind to folate receptor (FR) α and the target purine biosynthetic enzyme glycinamide ribonucleotide formyltransferase (GARFTase) with different cis and trans conformations. In this study, we designed novel analogs of this series with an amide moiety in the bridge region that can adopt both the cis and trans lowest energy conformations. This provides entropic benefit, by restricting the number of side-chain conformations of the unbound ligand to those most likely to promote binding to FRα and the target enzyme required for antitumor activity. NMR of the most active compound 7 showed both cis and trans amide bridge conformations in ~1:1 ratio. The bridge amide group in the best docked poses of 7 in the crystal structures of FRα and GARFTase adopted both cis and trans conformations, with the lowest energy conformations predicted by Maestro and evidenced by NMR within 1 kcal/mol. Compound 7 showed ~3-fold increased inhibition of FRα-expressing cells over its non-restricted parent analog 1 and was selectively internalized by FRα over the reduced folate carrier (RFC), resulting in significant in vitro antitumor activity toward FRα-expressing KB human tumor cells. Antitumor activity of 7 was abolished by treating cells with adenosine but was incompletely protected by 5-aminoimidazole-4-carboxamide (AICA) at higher drug concentrations, suggesting GARFTase and AICA ribonucleotide formyltransferase (AICARFTase) in de novo purine biosynthesis as the likely intracellular targets. GARFTase inhibition by compound 7 was confirmed by an in situ cell-based activity assay. Our results identify a “first-in-class” classical antifolate with a novel amide linkage between the scaffold and the side chain aryl L-glutamate that affords exclusive selectivity for transport via FRα over RFC and antitumor activity resulting from inhibition of GARFTase and likely AICARFTase. Compound 7 offers significant advantages over clinically used inhibitors of this class that are transported by the ubiquitous RFC, resulting in dose-limiting toxicities.
- Published
- 2019
- Full Text
- View/download PDF
34. Abstract 2992: Cellular pharmacodynamics of mitochondrial one-carbon metabolism-targeting 5-substituted pyrrolo[3,2-d]pyrimidine antifolates
- Author
-
Aamod Dekhne, Khushbu Shah, Gregory S. Ducker, Md. Junayed Nayeen, Jade M. Katinas, Jennifer Wong, Arpit Doshi, Xun Bao, Hasini Kalpage, Jenney Liu, Seongho Kim, Adrianne Wallace-Povirk, Changwen Ning, Carrie O'Connor, Zhanjun Hou, Lisa Polin, Jing Li, Maik Hüttemann, Joshua D. Rabinowitz, Charles E. Dann, Aleem Gangjee, and Larry Matherly
- Subjects
Cancer Research ,Oncology - Abstract
Folate-dependent one-carbon metabolism (1CM) is compartmentalized in the mitochondria and cytosol and generates a number of metabolites critical to tumor propagation. Folates are taken up by the plasma membrane facilitative transporters, reduced folate carrier (RFC; major tissue transporter) and proton-coupled folate transporter (PCFT; narrow physiological niche, but commonly expressed in solid tumors), and then transported into mitochondria via the mitochondrial folate transporter (MFT; SLC25A32). Although drug-targeting of cytosolic 1CM remains a clinical mainstay for a variety of cancers, development of clinically-useful agents targeting mitochondrial 1CM remains elusive. Of particular pharmacological interest is the mitochondrial 1CM enzyme, serine hydroxymethyltransferase2 (SHMT2). SHMT2 expression correlates with the oncogenic phenotype in a host of different cancers and, overall, SHMT2 is the fifth-most differentially expressed metabolic enzyme in cancer versus normal tissues. Despite its unequivocal oncogenic importance and therapeutic potential, there are no clinically relevant inhibitors of SHMT2. In this study, we characterized cellular pharmacodynamics of novel 5-substituted pyrrolo[3,2-d]pyrimidine antifolates (AGF291, AGF320, and AGF347) which show in vitroantitumor efficacy toward H460 lung, HCT-116 colon, and MIA PaCa-2 pancreatic cancer cells. Inhibition of mitochondrial SHMT2 and cytosolic 1CM at the purine nucleotide biosynthesis enzymes glycinamide ribonucleotide formyltransferase (GARFTase) and/or 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFTase) by this series was established by in vitro targeted metabolomics in H460, HCT-116, and MIA PaCa-2 cells and in vitro cell-free assays with purified enzymes. By depleting SHMT2-derived formate, these compounds potentiate their own direct inhibition of GARFTase and AICARFTase. Depletion of adenine nucleotide pools in vitro by all compounds led to inhibition of mTOR signaling to S6K1 in HCT116 cells. Subcellular fractionation of MIA PaCa-2 and MFT-null and human MFT-transfected glyB Chinese hamster ovary cells confirmed synthesis of polyglutamyl forms of AGF347 in both cytosol and mitochondria with mitochondrial uptake of AGF347 in part mediated by MFT. Treatment by all compounds decreased the cellular GSH/GSSG ratio, indicating depleted ability to combat oxidative stress. In vivo, AGF347 demonstrated potent antitumor efficacy against MIA PaCa-2 xenografts in SCID mice (n=5) with median tumor growth delay (T-C) in 4 mice >38 days and 1 of 5 tumor-free survivors (cured). In vivo metabolomics on tumor xenografts confirmed inhibition of serine catabolism and purine biosynthesis. Collectively, our studies establish the exceptional therapeutic potential of inhibitors dual-targeting mitochondrial and cytosolic 1CM. Citation Format: Aamod Dekhne, Khushbu Shah, Gregory S. Ducker, Md. Junayed Nayeen, Jade M. Katinas, Jennifer Wong, Arpit Doshi, Xun Bao, Hasini Kalpage, Jenney Liu, Seongho Kim, Adrianne Wallace-Povirk, Changwen Ning, Carrie O'Connor, Zhanjun Hou, Lisa Polin, Jing Li, Maik Hüttemann, Joshua D. Rabinowitz, Charles E. Dann, Aleem Gangjee, Larry Matherly. Cellular pharmacodynamics of mitochondrial one-carbon metabolism-targeting 5-substituted pyrrolo[3,2-d]pyrimidine antifolates [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2992.
- Published
- 2019
- Full Text
- View/download PDF
35. Abstract 4800: Targeting mitochondrial and cytosolic one-carbon metabolism in epithelial ovarian cancer via folate receptor alpha
- Author
-
Aleem Gangjee, Adrianne Wallace-Povirk, Aamod Dekhne, Larry H. Matherly, Zhanjun Hou, Khushbu Shah, Md. Junayed Nayeen, and Carrie O'Connor
- Subjects
Folate Receptor Alpha ,Cancer Research ,Cell growth ,Chemistry ,Chinese hamster ovary cell ,Cell ,Mitochondrion ,Cytosol ,medicine.anatomical_structure ,Oncology ,Serine hydroxymethyltransferase ,medicine ,Cancer research ,Purine metabolism - Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy. Even though most patients initially respond to platinum-based therapy, the likelihood of disease reoccurrence is virtually 100%. Thus, there is an urgent need for new tumor-selective therapies for EOC. One such treatment option involves targeting tumors via folate receptor alpha (FRα) which is overexpressed in up to 90% of EOCs and shows increasing expression with stage and grade of disease. Our laboratory discovered novel 5-substituted pyrrolo[3,2-d]pyrimidine analogs (AGF291, AGF320, AGF347, AGF359 and AGF362) which inhibit mitochondrial one-carbon (C1) metabolism at serine hydroxymethyltransferase (SHMT) 2, with secondary inhibitions at cytosolic enzyme targets including those in de novo purine biosynthesis and SHMT1. Potent inhibition was seen toward isogenic Chinese hamster ovary (CHO) cell lines individually expressing FRα, the reduced folate carrier (RFC, ubiquitously expressed tissue folate transporter) and the proton-coupled folate transporter (PCFT, expressed in a limited number of normal tissues and inactive at normal pH, and several solid tumors at acidic pH including EOC), and with FRα-expressing tumor cells, including KB and IGROV1, a human EOC cell line. Inhibitory potencies were in order, AGF347 > AGF362 >> AGF291 = AGF320 = AGF359. Drug effects were substantially reduced with excess folic acid, confirming FRα-mediated drug uptake. Toward cisplatin resistant SKOV3, TOV112D and A2780 EOC cells, inhibition in the nanomolar range was detected with all compounds. Excess folic acid abrogated drug effects to varying degrees, suggesting significant uptake by the PCFT and/or the RFC, in addition to FRα. Short-term (5 minutes) cell uptake assays with the EOC cell lines and [3H]AGF347 confirmed transport by PCFT and RFC. With sustained exposures resulting in steady state AGF347 accumulations under both physiologic (pH 7.2) and acidic (pH 6.8, approximating the tumor microenvironment) conditions, FRα uptake predominated. [3H]AGF347 treatment of IGROV1 EOC resulted in substantial drug accumulation in both cytosol and mitochondria. Inhibition of cell proliferation for all analogs was reversed by addition of both glycine and adenosine, implicating C1 metabolism in mitochondria including SHMT2 and de novo purine biosynthesis in the cytosol as the targeted pathways; protection by 5-aminoimidazole-4-carboxamide (AICA) with or without glycine was incomplete, implying direct targeting of AICA ribonucleotide formyltransferase (AICARFTase), the second folate-dependent enzyme in purine biosynthesis. Our studies describe first-in-class FRα-selective compounds targeting mitochondrial and cytosolic C1 metabolism, with potent activity against EOC, including cisplatin resistant EOC. Citation Format: Adrianne Wallace-Povirk, Carrie O’Connor, Aamod Dekhne, Zhanjun Hou, Md. Junayed Nayeen, Khushbu Shah, Aleem Gangjee, Larry Matherly. Targeting mitochondrial and cytosolic one-carbon metabolism in epithelial ovarian cancer via folate receptor alpha [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4800.
- Published
- 2019
- Full Text
- View/download PDF
36. Abstract 811: Multi-targeted inhibitors of mitochondrial one-carbon metabolism and cytosolic de novo purine synthesis enzymes as anti-tumor agents
- Author
-
Aleem Gangjee, Junayed Nayeen, Aamod Dekhne, Changwon Ning, Zhanjun Hou, Larry H. Matherly, Carrie O'Connor, Khushbu Shah, Jennifer Wong, Charles E. Dann, and Jade M. Katinas
- Subjects
De novo synthesis ,chemistry.chemical_classification ,Cancer Research ,Pyrimidine analogue ,Ribonucleotide ,Oncology ,Chemistry ,Cell growth ,Serine hydroxymethyltransferase ,Glycine ,Cancer cell ,Nucleotide ,Molecular biology - Abstract
Targeting cellular one-carbon (1C) metabolism in cancer cells with pyrrolo[3,2-d]pyrimidine analogs provides in vivo antitumor efficacy. We discovered a first-in-class series of 5-substituted pyrrolo[3,2-d]pyrimidine analogs (AGF291, AGF320, AGF347) with inhibition of serine hydroxymethyltransferase 2 (SHMT2), a key enzyme in mitochondrial 1C metabolism and a reported oncodriver. In addition, AGF291, AGF320 and AGF347 inhibited de novo purine nucleotide biosynthesis at glycinamide ribonucleotide formyltransferase (GARFTase) and 5-aminoimidazole-4-carboxamide (AICA) ribonucleotide formyltransferase (AICARFTase), and serine hydroxymethyltransferase 1 (SHMT1). In vivo studies demonstrated excellent antitumor efficacy for AGF347 against MIA PaCa-2 pancreatic adenocarcinoma xenografts in SCID mice with a median tumor growth delay (T-C) of >38 days for 4 mice, and 1 of 5 mice tumor-free 122 days post-treatment. AGF359, a new analog in this series, was a potent inhibitor of KB human tumor cell proliferation in vitro. AGF359 inhibition of KB human tumor cells was reversed with glycine and adenosine, establishing mitochondrial 1C metabolism and de novo purine biosynthesis as the targeted pathways; AICA plus glycine was incompletely protective, implicating AICARFTase as a direct cellular target. AGF359, like AGF291, AGF320, and AGF347, inhibited purified human SHMT2 (Ki = 0.399 ± 0.174 µM), SHMT1 (Ki = 0.70 ± 0.088 µM) and AICARFTase (Ki = 8.86 ± 2.83 µM). There was no inhibition of 5,10-methylene tetrahydrofolate dehydrogenase 2 (MTHFD2). Further structural modifications of these multi-targeted agents afforded pyrrolo[3,2-d]pyrimidine analogs AGF307 and AGF312. Cell-based glycine/nucleotide rescue experiments in KB tumor cells established that, as with AGF359, AGF307 and AGF312 were dual inhibitors of the mitochondrial 1C metabolism (AGF307) and de novo purine biosynthesis (AGF307, AGF312). These compounds are currently in further preclinical evaluation as a prelude to possible clinical development as antitumor agents. Citation Format: Md Junayed Nayeen, Khushbu Shah, Aamod Dekhne, Changwon Ning, Carrie O'Connor, Jade M. Katinas, Jennifer Wong, Zhanjun Hou, Charles E. Dann III, Larry H. Matherly, Aleem Gangjee. Multi-targeted inhibitors of mitochondrial one-carbon metabolism and cytosolic de novo purine synthesis enzymes as anti-tumor agents [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 811.
- Published
- 2019
- Full Text
- View/download PDF
37. Abstract 4794: Targeting mitochondrial and cytosolic one carbon metabolism of pancreatic adenocarcinoma via the proton-coupled folate transporter with novel 5-substituted pyrrolo[3,2-d]pyrimidine analogs
- Author
-
Jennifer Wong, Adrianne Wallace-Povirk, Jade M. Katinas, Larry H. Matherly, Zhanjun Hou, Jing Li, Xun Bao, Charles E. Dann, Josephine Frühauf, Carrie O'Connor, Changwen Ning, Aleem Gangjee, Md. Junayed Nayeen, and Aamod Dekhne
- Subjects
Purine ,chemistry.chemical_classification ,Cancer Research ,Metabolism ,Mitochondrion ,Molecular biology ,chemistry.chemical_compound ,Cytosol ,Enzyme ,Oncology ,chemistry ,Cell culture ,Serine hydroxymethyltransferase ,Purine metabolism - Abstract
Pancreatic cancer (PaC) represents the 4th leading cause of cancer-related deaths in the US with a mortality rate of 99%. The 5-year overall survival rate for PaC is currently 8%. One-carbon (C1) metabolism is frequently altered in cancer. For PaC, TCGA data sets show that elevated expression of key enzymes involved in cytosolic [5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFTase) and serine hydroxymethyltransferase (SHMT)1] and mitochondrial [SHMT2 and methylene tetrahydrofolate dehydrogenase 2 (MTHFD2)] C1 metabolism is associated with poor survival. Antifolate therapeutics disrupt cytosolic C1 pathways required for syntheses of thymidylate, purines, and certain amino acids, and are a mainstay for therapy of several cancers. Antifolate uptake into tumors and tissues involves the reduced folate carrier, the major tissue folate transporter, and the proton-coupled folate transporter (PCFT), which shows a more limited tissue distribution but is widely expressed in human solid tumors and is active only at acidic pHs characterizing the tumor microenvironment. We discovered novel 5-substituted pyrrolo[3,2-d]pyrimidine analogs (AGF347, AGF359) with PCFT transport that potently inhibited proliferation of PaC cell lines (AsPC-1, BxPC-3, CFPAC-1, HPAC, MIA PaCa-2 and PANC-1), of which HPAC (KRAS mutant) and BxPC-3 (KRAS wild-type) cells were most sensitive. The PaC cell lines all expressed PCFT transcripts and proteins that were active for PCFT transport with 3H-AGF347 at acid pH. When HPAC cells were incubated with 3H-AGF347 over 48 h, drug accumulated in both cytosol and mitochondria. 3H-AGF347 was extensively metabolized to polyglutamates. Treatment of PaC cells with AGF347 and AGF359 inhibited proliferation by inducing glycine and adenosine auxotrophy that was rescued by excess glycine and adenosine. This implied that both mitochondria and cytosolic C1 metabolism was inhibited. Inhibition of mitochondrial SHMT2 and cytosolic SHMT1, glycinamide ribonucleotide formyltransferase and/or AICARFTase was confirmed by in vitro targeted metabolomics and assays with purified enzymes. Tumor cell killing was confirmed (with HPAC and BxPC-3) by colony-forming assays with AGF347 and AGF359 and drug-induced apoptosis with AGF347 was demonstrated (with HPAC) by annexin V-PI staining and flow cytometry. AGF347 and AGF359 depleted purine nucleotides and inhibited mTOR signaling via S6K1 at least in part (for BxPC-3) via activation of AMPK, likely due to elevated ZMP accompanying suppression of AICARFTase. Collectively, our studies identify first-in-class inhibitors and establish the considerable therapeutic potential of dual-targeting mitochondrial and cytosolic C1 metabolism in PaC independent of KRAS mutation status and reflecting cellular uptake by PCFT. Citation Format: Changwen Ning, Aamod Dekhne, Md. Junayed Nayeen, Jade M. Katinas, Jennifer Wong, Josephine Frühauf, Xun Bao, Carrie O’Connor, Adrianne Wallace-Povirk, Jing Li, Charles E. Dann, Aleem Gangjee, Larry H. Matherly, Zhanjun Hou. Targeting mitochondrial and cytosolic one carbon metabolism of pancreatic adenocarcinoma via the proton-coupled folate transporter with novel 5-substituted pyrrolo[3,2-d]pyrimidine analogs [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4794.
- Published
- 2019
- Full Text
- View/download PDF
38. Abstract 789: Multi-targeted novel 5-substituted pyrrolo[3,2-d]pyrimidines with tumor-selective targeting and inhibition of cytosolic de novo purine biosynthesis and mitochondrial one-carbon metabolism
- Author
-
Nian Tong, Khushbu Shah, Aleem Gangjee, Carrie O’Connor, Adrianne W. Porvirk, Aamod Dekhne, Zhanjun Hou, and Larry H. Matherly
- Subjects
Cancer Research ,Oncology - Abstract
One-carbon (C1) metabolism supports a number of physiological and pathophysiological processes ranging from stem cell renewal to cancer progression. Clinically used antifolates are transported into both tumor and normal cells by the ubiquitously expressed reduced folate carrier (RFC). Uptake of targeted agents via tumor-specific folate receptors (FRs) over RFC would permit tumor-selectivity, while limiting dose-limiting toxicities associated with standard chemotherapy. Serine catabolism in mitochondria is the major source of glycine and C1 units for cytosolic biosynthesis, preserves redox balance and minimizes reactive oxygen species, and is an important source of ATP. Among the mitochondrial C1 enzymes, serine hydroxymethyltransferase 2 (SHMT2) and 5,10-methylene tetrahydrofolate (me-THF) dehydrogenase 2 (MTHFD2) are highly expressed in tumors versus normal tissues. SHMT2 has been suggested to be an important oncodriver. However, there are no clinically relevant inhibitors of these enzymes. To generate potential inhibitors of these enzymes, we synthesized 5-substituted pyrrolo[3,2-d]pyrimidine analogs as structural hybrids of cytotoxic 5-substituted pyrrolo[2,3-d]pyrimidines and me-THF. The 5-substituted pyrrolo[3,2-d] pyrimidine with a four carbon bridged phenyl side chain AGF300 afforded selective uptake via FRα over RFC, with inhibition of mitochondrial C1 metabolism and de novo purine biosynthesis, resulting in inhibition of KB human tumor cell proliferation. Inhibition of KB cells by AGF300 was reversed by glycine and adenosine. As previous studies of related 5-substituted pyrrolo[2,3-d]pyrimidines established that the nature and length of the bridge plays an important role in determining tumor cell potency and transport selectivity, we replaced the carbon adjacent to the phenyl ring in AGF300 with heteroatoms, including O (AGF323), S (AGF346) or NH (AGF350). These compounds were tested as growth inhibitors against engineered Chinese hamster ovary (CHO) cells singly expressing human FRα (RT16) or RFC (PC43-10). Incorporation of the O, S and NH in the pyrrolo[3,2-d]pyrimidine analogs preserved excellent inhibition of FRα-containing CHO cells (IC50s of 57 nM, 77 nM and 50 nM, respectively); there was no inhibition of cells with RFC uptake up to 1000 nM. AGF323, AGF346 and AGF350 inhibited proliferation of KB cells which was reversed by excess glycine and adenosine. This establishes that for AGF323, AGF346 and AGF350, both mitochondrial and cytosolic C1 metabolism were inhibited. The development of novel compounds targeting mitochondrial and cytosolic C1 pathways with tumor-selective uptake is highly significant in that this would overcome the drawbacks of currently used cytotoxic agents for cancer. Citation Format: Nian Tong, Khushbu Shah, Aleem Gangjee, Carrie O’Connor, Adrianne W. Porvirk, Aamod Dekhne, Zhanjun Hou, Larry H. Matherly. Multi-targeted novel 5-substituted pyrrolo[3,2-d]pyrimidines with tumor-selective targeting and inhibition of cytosolic de novo purine biosynthesis and mitochondrial one-carbon metabolism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 789.
- Published
- 2019
- Full Text
- View/download PDF
39. Abstract 4791: Impact of folate transport redundancies on the therapy with tumor-targeted and untargeted antifolates
- Author
-
Aleem Gangjee, Carrie O'Connor, Md. Junayed Nayeen, Larry H. Matherly, Zhanjun Hou, Changwen Ning, Adrianne Wallace-Povirk, and Josephine Frühauf
- Subjects
Cancer Research ,biology ,Chemistry ,Cell growth ,Cancer ,Transporter ,medicine.disease ,biology.organism_classification ,HeLa ,chemistry.chemical_compound ,Oncology ,Cell culture ,Antifolate ,Cancer cell ,Cancer research ,medicine ,Receptor - Abstract
Folates participate in one-carbon (C1) transfer reactions in normal and cancer cells. Antifolate therapeutics disrupt cytosolic C1 metabolism, and have long been a mainstay for the therapy of a number of cancers. There are three major folate transporter systems in human tissues and tumors, including the reduced folate carrier (RFC), folate receptors (FRs) and proton-coupled folate transporter (PCFT). RFC is broadly expressed in tissues and tumors and is characterized by a neutral pH optimum. PCFT has more limited tissue distribution but is widely expressed in human solid tumors and exhibits an acidic pH optimum. FRα is expressed in a subset of solid tumors including epithelial ovarian cancer (EOC) but shows limited expression in most normal tissues. We previously discovered novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl compounds typified by AGF94 which show selective cellular uptake by FRα and/or PCFT over RFC, in contrast to the classical antifolates methotrexate (MTX) and pemetrexed (PMX) which are transported by both PCFT and RFC, and to a lesser extent by FRα. We showed that AGF94 had enhanced antitumor activities by targeting cytosolic C1 metabolism to PCFT-expressing tumors but lacking RFC, reflecting contraction of cellular tetrahydrofolate pools. In EOC cells, AGF94 showed substantial in vitro inhibition of cell proliferation independent of FRα expression, as long as PCFT was present. We systematically explored the impact of folate transporter redundancies among PCFT, FRα and RFC in determining anti-tumor efficacy of novel tumor-targeted and classical antifolates. Towards this goal, we engineered cell line models from PCFT-, FR-, and RFC-null HeLa cells to express doxycycline (DOX)-inducible FRα or RFC. We used these models to constitutively express PCFT together with DOX-inducible FRα or RFC. These were characterized for transporter expression and function, as well as intracellular folate levels, with/without DOX induction. The relative contributions of RFC, PCFT and FRα to transport function were evaluated from pH 5.5 to 7.4, with radiolabeled MTX and AGF347 which targets both cytosolic and mitochondrial C1 metabolism. We assessed antiproliferative activities of classical antifolates (PMX, MTX, PT523) versus tumor-targeted compounds (e.g., AGF94, AGF102, AGF347) in these models with/without DOX. Our results establish that co-expression of the major folate transporters can have variable and surprisingly disparate and substantial impacts on anti-tumor efficacies of both classical and tumor-targeted antifolates. Our study identified critical determinants of anti-tumor activity with classical and tumor-targeted antifolates, including relative levels of folate transporter expression and transporter specificity, and the impact of intracellular folate levels and extracellular pH. Citation Format: Zhanjun Hou, Carrie O’Connor, Changwen Ning, Adrianne Wallace-Povirk, Josephine Frühauf, Md. Junayed Nayeen, Aleem Gangjee, Larry H. Matherly. Impact of folate transport redundancies on the therapy with tumor-targeted and untargeted antifolates [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4791.
- Published
- 2019
- Full Text
- View/download PDF
40. Discovery of 5-Substituted Pyrrolo[2,3-d]pyrimidine Antifolates as Dual-Acting Inhibitors of Glycinamide Ribonucleotide Formyltransferase and 5-Aminoimidazole-4-carboxamide Ribonucleotide Formyltransferase in De Novo Purine Nucleotide Biosynthesis: Implications of Inhibiting 5-Aminoimidazole-4-carboxamide Ribonucleotide Formyltransferase to AMPK Activation and Antitumor Activity
- Author
-
Aleem Gangjee, Manasa P. Ravindra, Shermaine Mitchell-Ryan, Larry H. Matherly, Steven Orr, Eric C. Hales, Sudhir Raghavan, Christina Cherian, Zhanjun Hou, and Yiqiang Wang
- Subjects
Hydroxymethyl and Formyl Transferases ,Models, Molecular ,Ribonucleotide ,medicine.drug_class ,AICA ribonucleotide ,Antineoplastic Agents ,Apoptosis ,Carboxamide ,CHO Cells ,AMP-Activated Protein Kinases ,KB Cells ,Article ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cricetulus ,Drug Discovery ,medicine ,Animals ,Humans ,Pyrroles ,Enzyme Inhibitors ,Purine Nucleotides ,Cell Proliferation ,Dose-Response Relationship, Drug ,Molecular Structure ,Chinese hamster ovary cell ,Adenosine ,Pyrimidines ,chemistry ,Biochemistry ,Antifolate ,Folic Acid Antagonists ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Growth inhibition ,Thymidine ,medicine.drug - Abstract
We synthesized 5-substituted pyrrolo[2,3-d]pyrimidine antifolates (compounds 5-10) with one-to-six bridge carbons and a benozyl ring in the side chain as antitumor agents. Compound 8 with a 4-carbon bridge was the most active analogue and potently inhibited proliferation of folate receptor (FR) α-expressing Chinese hamster ovary and KB human tumor cells. Growth inhibition was reversed completely or in part by excess folic acid, indicating that FRα is involved in cellular uptake, and resulted in S-phase accumulation and apoptosis. Antiproliferative effects of compound 8 toward KB cells were protected by excess adenosine but not thymidine, establishing de novo purine nucleotide biosynthesis as the targeted pathway. However, 5-aminoimidazole-4-carboxamide (AICA) protection was incomplete, suggesting inhibition of both AICA ribonucleotide formyltransferase (AICARFTase) and glycinamide ribonucleotide formyltransferase (GARFTase). Inhibition of GARFTase and AICARFTase by compound 8 was confirmed by cellular metabolic assays and resulted in ATP pool depletion. To our knowledge, this is the first example of an antifolate that acts as a dual inhibitor of GARFTase and AICARFTase as its principal mechanism of action.
- Published
- 2013
- Full Text
- View/download PDF
41. Therapeutic targeting malignant mesothelioma with a novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate via its selective uptake by the proton-coupled folate transporter
- Author
-
Aleem Gangjee, Sita Kugel Desmoulin, Lisa Polin, Larry H. Matherly, Lei Wang, Christina Cherian, Kathryn White, Juiwanna Kushner, Zhanjun Hou, and Mark Stout
- Subjects
Mesothelioma ,Cancer Research ,Mice, SCID ,Biology ,Toxicology ,Article ,HeLa ,Mice ,chemistry.chemical_compound ,In vivo ,Animals ,Humans ,Pyrroles ,Pharmacology (medical) ,Cell Proliferation ,Phosphoribosylglycinamide Formyltransferase ,Pharmacology ,Polyglutamate ,Cell growth ,Membrane transport ,biology.organism_classification ,Xenograft Model Antitumor Assays ,In vitro ,Pyrimidines ,Polyglutamic Acid ,Oncology ,Biochemistry ,chemistry ,Antifolate ,Cancer research ,Folic Acid Antagonists ,Female ,Nucleoside ,Proton-Coupled Folate Transporter ,HeLa Cells - Abstract
The 5-substituted pyrrolo[2,3-d]pyrimidine antifolate pemetrexed (Pmx) is an active agent for malignant pleural mesothelioma (MPM). Pmx is transported into MPM cells by the reduced folate carrier (RFC) and proton-coupled folate transporter (PCFT). We tested the notion that a novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate (compound 2) might be an effective treatment for MPM, reflecting its highly selective membrane transport by PCFT over RFC. Compound 2 selectively inhibited proliferation of a HeLa subline expressing exclusively PCFT (R1-11-PCFT4) over an isogenic subline expressing only RFC (R1-11-RFC6). By outgrowth, H2452 human MPM cells were highly sensitive to the inhibitory effects of compound 2. By colony-forming assays, following an intermittent (24 h) drug exposure, 2 was cytotoxic. Cytotoxic activity by 2 was due to potent inhibition of glycinamide ribonucleotide formyltransferase (GARFTase) in de novo purine biosynthesis, as confirmed by nucleoside protection and in situ GARFTase assays with [14C]glycine. Assays with [3H]compound 2 and R1-11-PCFT4 or R1-11-RFC6 cells directly confirmed selective membrane transport by PCFT over RFC. PCFT transport was also confirmed for H2452 cells. In R1-11-PCFT4 and H2452 cells, [3H]compound 2 was metabolized to polyglutamates. Potent in vivo efficacy was confirmed toward early- and upstage H2452 xenografts in severe combined immunodeficient mice administered intravenous compound 2. Our results demonstrate potent antitumor efficacy of compound 2 toward H2452 MPM in vitro and in vivo, reflecting its efficient membrane transport by PCFT over RFC, synthesis of polyglutamates, and inhibition of GARFTase. Selectivity for non-RFC cellular uptake processes by novel tumor-targeted antifolates such as compound 2 presents an exciting new opportunity for treating solid tumors.
- Published
- 2013
- Full Text
- View/download PDF
42. Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis
- Author
-
Adrianne Wallace-Povirk, Sudhir Raghavan, Larry H. Matherly, Lalit K. Golani, Jennifer Wong, Lisa Polin, Parker W. de Waal, Xinxin Li, Siobhan M. Deis, Mike R. Wilson, Karsten Melcher, Charles E. Dann, Jiyuan Ke, H. Eric Xu, Juiwanna Kushner, Carrie O'Connor, Xin Gu, Kathryn White, Aleem Gangjee, and Zhanjun Hou
- Subjects
0301 basic medicine ,Phosphoribosylglycinamide formyltransferase ,Pyrimidine ,Stereochemistry ,Heteroatom ,Antineoplastic Agents ,Mice, SCID ,Crystallography, X-Ray ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,Folate Receptor 1 ,Pyrroles ,Purine Nucleotides ,Phosphoribosylglycinamide Formyltransferase ,Chemistry ,Chinese hamster ovary cell ,Transporter ,Molecular Docking Simulation ,030104 developmental biology ,Pyrimidines ,Biochemistry ,Folate receptor ,030220 oncology & carcinogenesis ,Molecular Medicine ,Folic Acid Antagonists ,Heterografts ,Female ,Folate receptor 1 ,Drug Screening Assays, Antitumor ,Neoplasm Transplantation ,Proton-Coupled Folate Transporter - Abstract
Targeted antifolates with heteroatom replacements of the carbon vicinal to the phenyl ring in 1 by N (4), O (8), or S (9), or with N-substituted formyl (5), acetyl (6), or trifluoroacetyl (7) moieties, were synthesized and tested for selective cellular uptake by folate receptor (FR) α and β or the proton-coupled folate transporter. Results show increased in vitro antiproliferative activity toward engineered Chinese hamster ovary cells expressing FRs by 4-9 over the CH2 analogue 1. Compounds 4-9 inhibited de novo purine biosynthesis and glycinamide ribonucleotide formyltransferase (GARFTase). X-ray crystal structures for 4 with FRα and GARFTase showed that the bound conformations of 4 required flexibility for attachment to both FRα and GARFTase. In mice bearing IGROV1 ovarian tumor xenografts, 4 was highly efficacious. Our results establish that heteroatom substitutions in the 3-atom bridge region of 6-substituted pyrrolo[2,3-d]pyrimidines related to 1 provide targeted antifolates that warrant further evaluation as anticancer agents.
- Published
- 2016
43. Structural and Enzymatic Analysis of Tumor-Targeted Antifolates That Inhibit Glycinamide Ribonucleotide Formyltransferase
- Author
-
Siobhan M. Deis, Zhanjun Hou, Charles E. Dann, Aleem Gangjee, Larry H. Matherly, and Arpit Doshi
- Subjects
0301 basic medicine ,Purine ,Phosphoribosylglycinamide formyltransferase ,Models, Molecular ,Protein Conformation ,Antineoplastic Agents ,Biology ,Biochemistry ,KB Cells ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,Nucleotide ,Receptor ,Phosphoribosylglycinamide Formyltransferase ,chemistry.chemical_classification ,Transporter ,Xenograft Model Antitumor Assays ,In vitro ,030104 developmental biology ,Pemetrexed ,chemistry ,030220 oncology & carcinogenesis ,Folic Acid Antagonists ,medicine.drug - Abstract
Pemetrexed and methotrexate are antifolates used for cancer chemotherapy and inflammatory diseases. These agents have toxic side effects resulting, in part, from nonspecific cellular transport by the reduced folate carrier (RFC), a ubiquitously expressed facilitative transporter. We previously described 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine antifolates with modifications of the side chain linker and aromatic ring that are poor substrates for RFC but are efficiently transported via folate receptors (FRs) and the proton-coupled folate transporter (PCFT). These targeted antifolates are cytotoxic in vitro toward FR- and PCFT-expressing tumor cells and in vivo with human tumor xenografts in immune-compromised mice, reflecting selective cellular uptake. Antitumor efficacy is due to inhibition of glycinamide ribonucleotide (GAR) formyltransferase (GARFTase) activity in de novo synthesis of purine nucleotides. This study used purified human GARFTase (formyltransferase domain) to assess in vitro inhibition by eight novel thieno- and pyrrolo[2,3-d]pyrimidine antifolates. Seven analogues (AGF23, AGF71, AGF94, AGF117, AGF118, AGF145, and AGF147) inhibited GARFTase with Ki values in the low- to mid-nanomolar concentration range, whereas AGF50 inhibited GARFTase with micromolar potency similar to that of PMX. On the basis of crystal structures of ternary complexes with GARFTase, β-GAR, and the monoglutamyl antifolates, differences in inhibitory potencies correlated well with antifolate binding and the positions of the terminal carboxylates. Our data provide a mechanistic basis for differences in inhibitory potencies between these novel antifolates and a framework for future structure-based drug design. These analogues could be more efficacious than clinically used antifolates, reflecting their selective cellular uptake by FRs and PCFT and potent GARFTase inhibition.
- Published
- 2016
44. Correction to Structure-Activity Profiles of Novel 6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Antifolates with Modified Amino Acids for Cellular Uptake by Folate Receptors α and β and the Proton-Coupled Folate Transporter
- Author
-
Larry H. Matherly, Lalit K. Golani, Aleem Gangjee, Sai Zhao, Christina George, Zhanjun Hou, Steven Orr, Sudhir Raghavan, Mike R. Wilson, and Adrianne Wallace
- Subjects
chemistry.chemical_classification ,chemistry.chemical_compound ,Biochemistry ,Pyrimidine ,chemistry ,Stereochemistry ,Drug Discovery ,Molecular Medicine ,Proton-coupled folate transporter ,Receptor ,Article ,Amino acid - Abstract
Structure–activity relationships for cellular uptake and inhibition of cell proliferation were studied for 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates in which the terminal l-glutamate of the parent structure (7) was replaced by natural or unnatural amino acids. Compounds 7 and 10–13 were selectively inhibitory toward folate receptor (FR) α-expressing Chinese hamster ovary (CHO) cells. Antiproliferative effects of compounds 7 and 9–13 toward FRα- and FRβ-expressing CHO cells were only partly reflected in binding affinities to FRα and FRβ or in the docking scores with molecular models of FRα and FRβ. Compounds 7 and 11 were potent inhibitors of glycinamide ribonucleotide formyltransferase in de novo purine biosynthesis in KB human tumor cells. These studies establish for the first time the importance of the α- and γ-carboxylic acid groups, the length of the amino acid, and the conformation of the side chain for transporter binding and biological activity of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates.
- Published
- 2016
45. Therapeutic Targeting of a Novel 6-Substituted Pyrrolo [2,3-d]pyrimidine Thienoyl Antifolate to Human Solid Tumors Based on Selective Uptake by the Proton-Coupled Folate Transporter
- Author
-
Sita Kugel Desmoulin, Aleem Gangjee, Christina Cherian, Juiwanna Kushner, Eric C. Hales, Larry H. Matherly, Lisa Polin, Lei Wang, Kathryn White, Zhanjun Hou, and Mark Stout
- Subjects
Mice, SCID ,HeLa ,Mice ,chemistry.chemical_compound ,Drug Delivery Systems ,In vivo ,Neoplasms ,Animals ,Humans ,Pharmacology ,Mice, Inbred ICR ,Tumor microenvironment ,Dose-Response Relationship, Drug ,biology ,Transporter ,Hep G2 Cells ,Articles ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Pyrimidines ,chemistry ,Biochemistry ,Cell culture ,Apoptosis ,Symporter ,Antifolate ,Cancer research ,Folic Acid Antagonists ,Molecular Medicine ,Female ,Proton-Coupled Folate Transporter ,HeLa Cells - Abstract
The proton-coupled folate transporter (PCFT) is a proton-folate symporter with an acidic pH optimum. By real-time reverse transcription-polymerase chain reaction, PCFT was expressed in the majority of 53 human tumor cell lines, with the highest levels in Caco-2 (colorectal adenocarcinoma), SKOV3 (ovarian), and HepG2 (hepatoma) cells. A novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate (compound 1) was used to establish whether PCFT can deliver cytotoxic drug under pH conditions that mimic the tumor microenvironment. Both 1 and pemetrexed (Pmx) inhibited proliferation of R1-11-PCFT4 HeLa cells engineered to express PCFT without the reduced folate carrier (RFC) and of HepG2 cells expressing both PCFT and RFC. Unlike Pmx, 1 did not inhibit proliferation of R1-11-RFC6 HeLa cells, which express RFC without PCFT. Treatment of R1-11-PCFT4 cells at pH 6.8 with 1 or Pmx inhibited colony formation with dose and time dependence. Transport of [(3)H]compound 1 into R1-11-PCFT4 and HepG2 cells was optimal at pH 5.5 but appreciable at pH 6.8. At pH 6.8, [(3)H]compound 1 was metabolized to (3)H-labeled polyglutamates. Glycinamide ribonucleotide formyltransferase (GARFTase) in R1-11-PCFT4 cells was inhibited by 1 at pH 6.8, as measured by an in situ GARFTase assay, and was accompanied by substantially reduced ATP levels. Compound 1 caused S-phase accumulation and a modest level of apoptosis. An in vivo efficacy trial with severe combined immunodeficient mice implanted with subcutaneous HepG2 tumors showed that compound 1 was active. Our findings suggest exciting new therapeutic possibilities to selectively deliver novel antifolate drugs via transport by PCFT over RFC by exploiting the acidic tumor microenvironment.
- Published
- 2011
- Full Text
- View/download PDF
46. Abstract 1656: First-in-class, fluorinated folate receptor specific agents that target tumor cells via inhibition of serine hydroxymethyltransferase 2 (SHMT2) and 5-aminoimidazole-4-carboxamide formyltransferase (AICARFTase)
- Author
-
Aleem Gangjee, Aamod Dekhne, Junayed Nayeen, Larry H. Matherly, Khushbu Shah, and Zhanjun Hou
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Cancer Research ,AICA ribonucleotide ,In vitro ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Enzyme ,Oncology ,chemistry ,Folate receptor ,030220 oncology & carcinogenesis ,Serine hydroxymethyltransferase ,Glycine ,Cancer research ,Cytotoxic T cell ,Cytotoxicity - Abstract
Tumor-specific folate receptor (FR) targeting over the ubiquitous reduced folate carrier (RFC) allows for specific tumor targeting without the attending dose-limiting toxicity of all clinically used cytotoxic agents. In an attempt to provide selectivity for FRα and/or β, folic acid has been conjugated with a variety of cytotoxic payloads to alleviate dose-limiting toxicities since folic acid is not transported via RFC (the major tissue folate transporter) to any significant extent. These conjugates have been unsuccessful in clinical trials likely in part due to the instability of the linkers used for conjugation. Premature cleavage results in the cytotoxic component being released prior to tumor penetration, thus precluding any possibility of selective tumor targeting. We designed novel pyrrolo[3,2-d]pyrimidine compounds in which both the targeting component and the cytotoxic component are in the same molecule. These compounds do not require any conjugation or linkers and hence do not suffer any premature cleavage. In this report, we synthesized and evaluated a “first-in-class” series of fluorinated analogues that target FRα/β and have limited transport via RFC, thus providing selectivity for tumors expressing FRα and/or FRβ. Two lead molecules, AGF347 and AGF355, were potent inhibitors of KB human tumor cell proliferation in vitro (IC50 = 1.05 nM and 6.25 nM respectively). Based on protection experiments with glycine, thymidine, adenosine and 5-aminoimidazole-4-carboxamide (AICA), AGF347 and AGF355 were identified as dual inhibitors of serine hydroxymethyltransferase 2 (SHMT2), a key enzyme in mitochondrial one-carbon metabolism and reported oncodriver, and AICA ribonucleotide formyltransferase (AICARFTase), the 2nd folate-dependent step in de novo purine biosynthesis. These compounds are currently being evaluated in preclinical studies as potential candidates for clinical trials. Citation Format: Md Junayed Nayeen, Khushbu Shah, Aleem Gangjee, Aamod Dekhne, Zhanjun Hou, Larry H. Matherly. First-in-class, fluorinated folate receptor specific agents that target tumor cells via inhibition of serine hydroxymethyltransferase 2 (SHMT2) and 5-aminoimidazole-4-carboxamide formyltransferase (AICARFTase) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1656.
- Published
- 2018
- Full Text
- View/download PDF
47. Abstract 3980: Dual targeting mitochondrial and cytosolic one-carbon metabolism via the proton-coupled folate transporter with novel 5-substituted pyrrolo[3,2-d]pyrimidine antifolates
- Author
-
Carrie O'Connor, Joshua D. Rabinowitz, Aleem Gangjee, Aamod Dekhne, Khushbu Shah, Gregory S. Ducker, Zhanjun Hou, Larry H. Matherly, Adrianne Wallace-Povirk, Md. Junayed Nayeen, Josephine Frühauf, and Lisa Polin
- Subjects
Purine ,chemistry.chemical_classification ,Cancer Research ,Chinese hamster ovary cell ,AICA ribonucleotide ,Transporter ,Serine ,chemistry.chemical_compound ,Enzyme ,Oncology ,chemistry ,Biochemistry ,Cell culture ,Cancer cell - Abstract
Cellular one-carbon (1C) metabolism generates a host of metabolites especially critical to cancer cells. Classical 1C inhibitors often primarily target a single enzyme, commonly leading to drug resistance and necessitating the development of novel compounds targeting multiple enzymes. We synthesized several novel 5-substituted pyrrolo[3,2-d]pyrimidine compounds (AGF291, AGF320, and AGF347) with transport by the proton-coupled folate transporter (PCFT), a folate transporter with a narrow physiological niche, but commonly expressed in many solid tumors. These analogs showed substantial inhibition in proliferation assays with Chinese hamster ovary sublines engineered to individually express PCFT or the ubiquitously-expressed reduced folate carrier (the major tissue folate transporter), as well as in the PCFT-expressing human cancer cell lines H460 (large cell lung carcinoma), HCT-116 (colorectal carcinoma), and MIA PaCa-2 (pancreatic ductal adenocarcinoma). Full abrogation of inhibitory effects for all compounds required co-treatment with both adenosine and glycine, suggesting dual-targeting of cytosolic de novo purine biosynthesis and mitochondrial 1C/glycine metabolism. For cytosolic de novo purine biosynthesis, lack of protection by 5-aminoimidazole-4-carboxamide (AICA) suggested AICA ribonucleotide formyltransferase (AICARFTase), the second folate-dependent enzyme, as the likely enzyme target. AICARFTase inhibition was confirmed in intact H460 and HCT-116 cells by measuring accumulation of AICAR by targeted metabolomics. Targeting of mitochondrial enzymes in H460 and HCT-116 cells was suggested by selective inhibition of incorporation of [3-14C]serine over [14C]formate into the purine intermediate [14C]formyl glycinamide ribonucleotide. [2,3,3-2H]Serine isotope label scrambling analysis confirmed the mitochondrial target to be serine hydroxymethyltransferase2 (SHMT2). SHMT2 is a potentially promising anticancer drug target as its expression is highly correlated with the malignant phenotype across a broad spectrum of cancers beyond lung and colon, including breast, glioma, and liver. Indeed, our initial in-vivo studies with AGF291 suggest potent efficacy toward both MIA PaCa-2 and H460 xenograft mouse models. Our studies demonstrate the potential for tumor-selective dual targeting of both mitochondrial and cytosolic 1C metabolism to overcome resistance to classical 1C inhibitors. Citation Format: Aamod S. Dekhne, Gregory S. Ducker, Josephine Frühauf, Khushbu Shah, Md. Junayed Nayeen, Adrianne Wallace-Povirk, Carrie O'Connor, Zhanjun Hou, Lisa Polin, Aleem Gangjee, Joshua D. Rabinowitz, Larry H. Matherly. Dual targeting mitochondrial and cytosolic one-carbon metabolism via the proton-coupled folate transporter with novel 5-substituted pyrrolo[3,2-d]pyrimidine antifolates [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3980.
- Published
- 2018
- Full Text
- View/download PDF
48. Abstract 1658: 5N-substituted pyrrolo[3,2-d]pyrimidines: Tumor-targeted agents with first-in-class dual inhibition of serine hydroxymethyl transferase 2 and 5-amino-4-imidazolecarboxamide ribonucleotide formyl transferase enzymes
- Author
-
Aamod Dekhne, Aleem Gangjee, Larry Matherly, Khushbu Shah, and Zhanjun Hou
- Subjects
chemistry.chemical_classification ,Serine ,Dual inhibition ,Cancer Research ,chemistry.chemical_compound ,Enzyme ,Ribonucleotide ,Oncology ,chemistry ,Biochemistry ,Transferase ,Hydroxymethyl ,Tumor targeted - Abstract
Design, modeling and synthesis of pyrrolo[3,2-d]pyrimidine analogs were carried out based on the concept of dual mechanisms-of-action for achieving maximal antitumor efficacy. Tumor-targeted delivery via agents that are selectively transported by the folate receptor (FRα/β) and/or PCFT, overexpressed in tumor cells such as epithelial ovarian cancer and non-small cell lung cancer, over the ubiquitously expressed reduced folate carrier (RFC) would afford tumor selectivity. Gangjee, Matherly and coworkers previously reported 5-substituted pyrrolo[2,3-d]pyrimidines as inhibitors of de novo purine nucleotide biosynthesis glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide (AICA) ribonucleotide formyltransferase (AICARFTase). To synthesize potential dual inhibitors of de novo purine biosynthesis at cytosolic AICARFTase and mitochondrial one-carbon metabolism at serine hydroxymethyl transferase (SHMT) 2, we generated structural hybrids of these inhibitors and 5,10-methylenetetrahydrofolate (the SHMT2 cofactor), 5-substituted pyrrolo[3,2-d]pyrimidine analogs AGF291, AGF299, AGF300, AGF318, AGF320, and AGF331. Molecular modeling of these compounds utilizing X-ray crystal structures of the transporters (FRα/β) and enzyme targets (AICARFTase, SHMT2) predicted excellent activities. The influence of bridge lengths and the aromatic moieties of these novel compounds were evaluated by testing their anti-proliferative activities in Chinese hamster ovary cells expressing human FRα or PCFT, and in FRα- and/or PCFT expressing nasopharyngeal carcinoma (KB) and lung cancer (H460) cells. All the pyrrolo[3,2-d]pyrimidine analogs were inhibitory toward FR- and/or PCFT-expressing cells. Compared to its pyrrolo[2,3-d]pyrimidine analog, AGF300 increased inhibition of cells via FR uptake from IC50 8.6 nM to 2.1 nM (by 4-fold) and decreased uptake through RFC from 56.5 nM to 516 nM (by 10-fold), suggesting a FR-targeted delivery of the agent. Toward H460 xenografts in SCID mice, AGF291 was efficacious. Protection studies with thymidine, glycine, adenosine and AICA and radiotracer studies with [3-14C]serine identified the likely intracellular targets of the pyrrolo[3,2-d]pyrimidine analogs as SHMT2 and AICARFTase. Mitochondrial one-carbon metabolism originating with SHMT2 is a critical source of reducing equivalents and one-carbon units for cytosolic biosynthesis and SHMT2 has been implicated as an oncodriver in several tumor types. While inhibition of SHMT2 was the subject of exploratory studies, our novel pyrrolo[3,2-d]pyrimidine series are “first-in-class” in terms of their in vitro and in vivo antitumor efficacies attributable to dual targeting SHMT2 and AICARFTase. Citation Format: Khushbu Shah, Aamod Dekhne, Zhanjun Hou, Larry Matherly, Aleem Gangjee. 5N-substituted pyrrolo[3,2-d]pyrimidines: Tumor-targeted agents with first-in-class dual inhibition of serine hydroxymethyl transferase 2 and 5-amino-4-imidazolecarboxamide ribonucleotide formyl transferase enzymes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1658.
- Published
- 2018
- Full Text
- View/download PDF
49. Abstract 3983: Tumor-targeting with novel dual-targeted 6-substituted thieno[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α, and inhibition of de novo purine nucleotide biosynthesis
- Author
-
Carrie O'Connor, Zhanjun Hou, Nian Tong, Aleem Gangjee, Xilin Zhou, Larry H. Matherly, and Adrianne Wallace-Povirk
- Subjects
Purine ,chemistry.chemical_classification ,Folate Receptor Alpha ,Cancer Research ,Ribonucleotide ,Cell growth ,Chinese hamster ovary cell ,AICA ribonucleotide ,Biology ,chemistry.chemical_compound ,Oncology ,chemistry ,Biochemistry ,Folate receptor ,Cancer research ,Nucleotide - Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecologic cancer, with limited progress in targeted therapy or in combating drug resistance. Reflecting this, there is an urgent need to develop novel inhibitors with minimal toxicity. EOC is associated with increasing expression of folate receptor alpha (FRα) with increasing stage and grade of disease. Of particular interest are targeted therapeutics toward FRα which spare transport by the ubiquitously expressed reduced folate carrier (RFC). FRα is a viable target for tumor selectivity with limited expression in normal tissues, where it is only expressed on apical membranes. We previously described 6-substituted thieno[2,3-d]pyrimidine (6TP) benzoyl antifolates with bridge lengths of 2-8 carbons as potent and selective inhibitors of tumor cells including EOCs that express FRs. The most potent compound (AGF50) included a 4-carbon bridge and showed FRα selectivity and inhibition of de novo purine biosynthesis. We synthesized AGF50 analogs with various aryl side-chain modifications (e.g., thiophene, furan, pyridine) including 2'fluorine substitutions. Toward isogenic Chinese hamster ovary cell lines individually expressing FRα, RFC and the proton-coupled folate transporter (PCFT), the 6TP analogs were completely selective toward FRα over RFC and PCFT, manifested as inhibition of cell proliferation with nmolar IC50s. Activity of this series extended to KB human tumor cells and human EOC cell lines (e.g., IGROV1). Drug effects were abolished with excess folic acid, confirming FRα uptake. Adenosine completely protected cells from the inhibitory effects of 6TPs in vitro, suggesting inhibition of one-carbon transfers mediated by glycinamide ribonucleotide (GAR) formyltransferase (GARFTase) and/or 5-aminoimidazole-4-carboxamide (AICA) ribonucleotide formyltransferase (AICARFTase), leading to synthesis of purine nucleotides. Protection from 6TPs by AICA was incomplete (in contrast to a “pure” GARFTase inhibitor AGF94), suggesting inhibition of AICARFTase. Whereas AGF94 potently inhibited GARFTase (>98%) in KB cells, reflected in [14C]glycine incorporation into [14C]formyl GAR, inhibition by the 6TPs was incomplete (75-90%) up to 1 μM drug. AICARFTase inhibition was confirmed by measuring accumulation of AICA ribonucleotide (ZMP), although this varied for different 6TP analogs. Our studies establish an important structure-activity relationship for novel FRα-targeted 6TPs with aromatic side-chain modifications and imply the therapeutic potential of targeting AICARFTase for EOC. Citation Format: Adrianne Wallace-Povirk, Nian Tong, Carrie O'Connor, Zhanjun Hou, Aleem Gangjee, Larry Matherly, Xilin Zhou. Tumor-targeting with novel dual-targeted 6-substituted thieno[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α, and inhibition of de novo purine nucleotide biosynthesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3983.
- Published
- 2018
- Full Text
- View/download PDF
50. Identification of the Minimal Functional Unit of the Homo-oligomeric Human Reduced Folate Carrier
- Author
-
Christina Cherian, Jianmei Wu, Zhanjun Hou, Larry H. Matherly, and Joseph D. Drews
- Subjects
Dimer ,Blotting, Western ,Mutant ,Biology ,Biochemistry ,Oligomer ,Reduced Folate Carrier Protein ,Structure-Activity Relationship ,chemistry.chemical_compound ,Protein structure ,Membrane Biology ,Humans ,Immunoprecipitation ,Structure–activity relationship ,Molecular Biology ,Polyacrylamide gel electrophoresis ,Mesylates ,Microscopy, Confocal ,Wild type ,Membrane Transport Proteins ,Cell Biology ,chemistry ,Mutation ,Mutagenesis, Site-Directed ,Electrophoresis, Polyacrylamide Gel ,Protein Multimerization ,Intracellular ,HeLa Cells ,Signal Transduction - Abstract
The reduced folate carrier (RFC) is the major transport system for folates in mammals. We previously demonstrated the existence of human RFC (hRFC) homo-oligomers and established the importance of these higher order structures to intracellular trafficking and carrier function. In this report, we examined the operational significance of hRFC oligomerization and the minimal functional unit for transport. In negative dominance experiments, multimeric transporters composed of different ratios of active (either wild type (WT) or cysteine-less (CLFL)) and inactive (either inherently inactive (Y281L and R373A) due to mutation, or resulting from inactivation of the Y126C mutant by (2-sulfonatoethyl) methanethiosulfonate (MTSES)) hRFC monomers were expressed in hRFC-null HeLa (R5) cells, and residual WT or CLFL activity was measured. In either case, residual transport activity with increasing levels of inactive mutant correlated linearly with the fraction of WT or CLFL hRFC in plasma membranes. When active covalent hRFC dimers, generated by fusing CLFL and Y126C monomers, were expressed in R5 cells and treated with MTSES, transport activity of the CLFL-CLFL dimer was unaffected, whereas Y126C-Y126C was potently (64%) inhibited; heterodimeric CLFL-Y126C and Y126C-CLFL were only partly (27 and 23%, respectively) inhibited by MTSES. In contrast to Y126C-Y126C, trans-stimulation of methotrexate uptake by intracellular folates for Y126C-CLFL and CLFL-Y126C was nominally affected by MTSES. Collectively, these results strongly support the notion that each hRFC monomer comprises a single translocation pathway for anionic folate substrates and functions independently of other monomers (i.e. despite an oligomeric structure, hRFC functions as a monomer).
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.