Lu-EC0800 Combined with the Antifolate Pemetrexed: Preclinical Pilot Study of Folate Receptor Targeted Radionuclide Tumor Therapy

Total Page:16

File Type:pdf, Size:1020Kb

Lu-EC0800 Combined with the Antifolate Pemetrexed: Preclinical Pilot Study of Folate Receptor Targeted Radionuclide Tumor Therapy Published OnlineFirst September 12, 2013; DOI: 10.1158/1535-7163.MCT-13-0422-T Molecular Cancer Small Molecule Therapeutics Therapeutics 177Lu-EC0800 Combined with the Antifolate Pemetrexed: Preclinical Pilot Study of Folate Receptor Targeted Radionuclide Tumor Therapy Josefine Reber1, Stephanie Haller1, Christopher P. Leamon2, and Cristina Muller€ 1 Abstract Targeted radionuclide therapy has shown impressive results for the palliative treatment of several types of cancer diseases. The folate receptor has been identified as specifically associated with a variety of frequent tumor types. Therefore, it is an attractive target for the development of new radionuclide therapies using folate- based radioconjugates. Previously, we found that pemetrexed (PMX) has a favorable effect in reducing undesired renal uptake of radiofolates. Moreover, PMX also acts as a chemotherapeutic and radiosensitizing agent on tumors. Thus, the aim of our study was to investigate the combined application of PMX and the therapeutic radiofolate 177Lu-EC0800. Determination of the combination index (CI) revealed a synergistic inhibitory effect of 177Lu-EC0800 and PMX on the viability of folate receptor–positive cervical (KB) and ovarian (IGROV-1) cancer cells in vitro (CI < 0.8). In an in vivo study, tumor-bearing mice were treated with 177Lu-EC0800 (20 MBq) and a subtherapeutic (0.4 mg) or therapeutic amount (1.6 mg) of PMX. Application of 177Lu-EC0800 with PMXther resulted in a two- to four-fold enhanced tumor growth delay and a prolonged survival of KB and IGROV-1 tumor-bearing mice, as compared to the combination with PMXsubther or untreated control mice. 177 PMXsubther protected the kidneys from undesired side effects of Lu-EC0800 (20 MBq) by reducing the absorbed radiation dose. Intact kidney function was shown by determination of plasma parameters and quantitative single-photon emission computed tomography using 99mTc-DMSA. Our results confirmed the anticipated dual role of PMX. Its unique features resulted in an improved antitumor effect of folate-based radionuclide therapy and prevented undesired radio-nephrotoxicity. Mol Cancer Ther; 12(11); 2436–45. Ó2013 AACR. Introduction The development of new targeting strategies for the Targeted radionuclide therapy has shown impressive treatment of further tumor types is of high interest and results for the palliative treatment of several cancer dis- would have a critical impact on the future management of eases. It is based on the use of particle-emitting radio- these cancer diseases. In this respect, the folate receptor is isotopes (e.g., 177Lu, 90Y, 131I) in conjunction with tumor- an attractive target as it has been identified as specifically targeted biomolecules (e.g., peptides, antibodies; ref. 1). A associated with a variety of cancer types, such as ovarian, prominent example of a successfully used radiopharma- endometrial, lung, brain, breast, and colorectal cancer (4, ceutical in clinical routine are somatostatin-based radio- 5). The vitamin folic acid has been used as a targeting peptides (e.g., 177Lu-DOTATATE, 90Y-DOTATOC) for ligand because it binds to the folate receptor with high the treatment of neuroendocrine tumors (2). Moreover, affinity followed by endocytotic internalization of the radiolabeled antibodies such as 90Y-Ibritumomab (Zeva- therapeutic payload into cancer cells (6). While folic acid lin) and 131I-tositumomab (Bexxar) are approved for the conjugates of highly toxic chemotherapeutics have been treatment of non-Hodgkins lymphoma (3). successfully used in clinical trials (7, 8), application of therapeutic folic acid radioconjugates is currently being developed in preclinical studies. Substantial expression of the folate receptor in the Authors' Affiliations: 1Center for Radiopharmaceutical Sciences ETH- proximal tubule cells of the kidneys results in commonly PSI-USZ, Paul Scherrer Institute, Villigen-PSI, Switzerland; 2Endocyte Inc., West Lafayette, Indiana high and specific renal uptake of folate-based radio- conjugates (9, 10). As a consequence there is an inherent Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). risk of damage to the kidneys by particle radiation. How- ever, we have shown in several preclinical studies that Corresponding Author: Cristina Muller,€ Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzer- administration of the antifolate pemetrexed (PMX) re- land. Phone: 41-56-310-44-54; Fax: 41-56-310-28-49; E-mail: sulted in a tremendous reduction of the radiofolate’s [email protected] retention in the kidneys whereas accumulation in tumor doi: 10.1158/1535-7163.MCT-13-0422-T xenografts remained unaffected (11–13). The exact mech- Ó2013 American Association for Cancer Research. anism of this interaction is still not completely clear. 2436 Mol Cancer Ther; 12(11) November 2013 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 12, 2013; DOI: 10.1158/1535-7163.MCT-13-0422-T 177Lu-Folate Therapy in Combination with Pemetrexed However, in numerous preclinical studies, we observed organisms and Cell Cultures (DSMZ). IGROV-1 cells an interrelation between the kidney reducing effect and (human ovarian carcinoma cells) were a kind gift from the time point of preinjected PMX (14) as well as the molar Dr. Gerrit Jansen (Department of Rheumatology, VU amount of PMX and the folate radioconjugate, respective- University Medical Center, Amsterdam, the Nether- ly (15). However, the reduced kidney uptake of radio- lands). All 3 cell lines were authenticated by DMSZ based folates was not a result of PMX’s antifolate activity as the on DNA profiling. KB and IGROV-1 cells are known to effect was maintained even if PMX was applied in com- express the folate receptor (31, 32), whereas folate receptor bination with the antidote thymidine (16). These facts negative PC-3 cells (33) were used as a negative control. suggest that PMX’s kidney reducing effect is based on a PC-3 cells were cultured in RPMI 1640 medium (Amimed) competition among PMX and the folate radioconjugate for whereas KB and IGROV-1 cells were cultured in folate- folate receptor binding sites in the kidneys. free RPMI medium referred to as FFRPMI (without folic PMX is a multitargeted antifolate, which is clinically acid, vitamin B12, and phenol red; Cell Culture Technol- approved for the treatment of pleural mesothelioma ogies GmbH). Cell culture media were supplemented and non–small cell lung cancer in combination with with 10% fetal calf serum, L-glutamine, and antibiotics cisplatin (17–19). Moreover, PMX is currently being (penicillin/streptomycin/fungizone). Routine culture tested in a number of clinical trials for the treatment treatment was conducted twice a week. Uptake and inter- of several other cancer types (reviewed in ref. 20), nalization studies of 177Lu-EC0800 as well as clonogenic among those is also ovarian cancer (21). PMX has and MTT assays were conducted as previously reported been used in combinations with gemcitabine, tyrosine in the literature in detail in the Supplementary Methods kinase inhibitors, antibodies, or even external radiation (29, 34). (22–24). A combination of PMX with external radiother- apy was based on the observation that PMX acts as a MTT assay radiosensitizing agent in variable types of cancer cells Cell viability was assessed using an MTT assay (35). In in vitro and in vivo (25–28). brief, KB, IGROV-1, and PC-3 cells were treated with We hypothesized that PMX would have a dual role if it 200 mL FFRPMI medium (without supplements) contain- was combined with therapeutic folate radioconjugates. ing 177Lu-EC0800 (0.001–5.0 MBq/mL) and/or PMX First, it was expected to prevent radionephropathy by (0.01–100 mmol/L; pemetrexed, Alimta, LY231514; Eli reducing the absorbed radiation dose of folate radio- Lilly, Supplementary Fig. S1). The control cells were conjugates in the kidneys. Second, PMX was believed to incubated with FFRPMI medium only. After 4 hours enhance the therapeutic efficacy of folate-based radio- incubation at 37C, the cells were washed once with nuclide tumor therapy by its action as a chemotherapeutic 200 mL PBS followed by addition of FFRPMI or RPMI and radiosensitizing agent. medium (with supplements). Cells were then allowed to The goal of this study was to show the anticipated dual grow for 4 days at 37C without changing medium. role of PMX. For this purpose, we used an established Analysis of the viability was conducted as previously DOTA-folate conjugate (EC0800; ref. 29), which was reported using an MTT reagent and a microplate reader 177 T ¼ E radiolabeled with Lu ( 1/2 6.7 days, av(b-): 134 keV, (Victor X3; Perkin Elmer; ref. 36). To quantify cell viability, E 177 (g): 113 keV, 208 keV). Lu-EC0800 was applied in the absorbance of the test samples was expressed as combination with PMX using 2 folate receptor positive percentage of the absorbance of the control cell samples human cancer cells (KB and IGROV-1) in vitro and as (¼100%). Dose–response curves were analyzed using the human tumor xenografts in athymic nude mice. More- software GraphPad Prism (version 4.0). Inhibition of cell over, the protective effect of PMX on the kidneys to reduce viability was expressed as the half-maximal inhibitory the absorbed radiation dose was investigated in non– concentration of PMX (IC50 in mmol/L) and as the half- tumor-bearing mice. maximal inhibitory activity concentration of 177Lu- EC0800 (IAC50 in MBq/mL) by measuring dose–response Materials and Methods curves of PMX and 177Lu-EC0800. The dose–response Preparation of 177Lu-EC0800 curves were used to determine the combination index The DOTA-folate conjugate (EC0800; ref. 29) was kindly (CI) according to Chou and colleagues (Supplementary provided by Endocyte Inc. (Supplementary Fig. S1). The Methods; ref. 37). radiosynthesis of 177Lu-EC0800 and the evaluation of its stability were carried out as previously reported (Supple- Animal studies 177 in vivo mentary Methods; ref.
Recommended publications
  • Pemetrexed-Lilly-Epar-Product-Information En.Pdf
    ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS 1 1. NAME OF THE MEDICINAL PRODUCT Pemetrexed Lilly 100 mg powder for concentrate for solution for infusion Pemetrexed Lilly 500 mg powder for concentrate for solution for infusion 2. QUALITATIVE AND QUANTITATIVE COMPOSITION Pemetrexed Lilly 100 mg powder for concentrate for solution for infusion Each vial contains 100 mg of pemetrexed (as pemetrexed disodium). Excipient with known effect Each vial contains approximately 11 mg sodium. Pemetrexed Lilly 500 mg powder for concentrate for solution for infusion Each vial contains 500 mg of pemetrexed (as pemetrexed disodium). Excipient with known effect Each vial contains approximately 54 mg sodium. After reconstitution (see section 6.6), each vial contains 25 mg/mL of pemetrexed. For the full list of excipients, see section 6.1. 3. PHARMACEUTICAL FORM Powder for concentrate for solution for infusion. White to either light yellow or green-yellow lyophilised powder. 4. CLINICAL PARTICULARS 4.1 Therapeutic indications Malignant pleural mesothelioma Pemetrexed Lilly in combination with cisplatin is indicated for the treatment of chemotherapy naïve patients with unresectable malignant pleural mesothelioma. Non-small cell lung cancer Pemetrexed Lilly in combination with cisplatin is indicated for the first line treatment of patients with locally advanced or metastatic non-small cell lung cancer other than predominantly squamous cell histology (see section 5.1). Pemetrexed Lilly is indicated as monotherapy for the maintenance treatment of locally advanced or metastatic non-small cell lung cancer other than predominantly squamous cell histology in patients whose disease has not progressed immediately following platinum-based chemotherapy (see section 5.1).
    [Show full text]
  • Folic Acid Antagonists: Antimicrobial and Immunomodulating Mechanisms and Applications
    International Journal of Molecular Sciences Review Folic Acid Antagonists: Antimicrobial and Immunomodulating Mechanisms and Applications Daniel Fernández-Villa 1, Maria Rosa Aguilar 1,2 and Luis Rojo 1,2,* 1 Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas, CSIC, 28006 Madrid, Spain; [email protected] (D.F.-V.); [email protected] (M.R.A.) 2 Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, 28029 Madrid, Spain * Correspondence: [email protected]; Tel.: +34-915-622-900 Received: 18 September 2019; Accepted: 7 October 2019; Published: 9 October 2019 Abstract: Bacterial, protozoan and other microbial infections share an accelerated metabolic rate. In order to ensure a proper functioning of cell replication and proteins and nucleic acids synthesis processes, folate metabolism rate is also increased in these cases. For this reason, folic acid antagonists have been used since their discovery to treat different kinds of microbial infections, taking advantage of this metabolic difference when compared with human cells. However, resistances to these compounds have emerged since then and only combined therapies are currently used in clinic. In addition, some of these compounds have been found to have an immunomodulatory behavior that allows clinicians using them as anti-inflammatory or immunosuppressive drugs. Therefore, the aim of this review is to provide an updated state-of-the-art on the use of antifolates as antibacterial and immunomodulating agents in the clinical setting, as well as to present their action mechanisms and currently investigated biomedical applications. Keywords: folic acid antagonists; antifolates; antibiotics; antibacterials; immunomodulation; sulfonamides; antimalarial 1.
    [Show full text]
  • Pemetrexed (Alimta) Is Covered Pemetrexed (Alimta®) Is Considered Medically Necessary for the Treatment of Patients With
    Corporate Medical Policy ® Pemetrexed (Alimta ) File Name: pemetrexed_alimta Origination: 8/2016 Last CAP Review: 4/2020 Next CAP Review: 4/2021 Last Review: 4/2020 Description of Procedure or Service ® Pemetrexed (Alimta ) is a folate analog metabolic inhibitor indicated for non-squamous non-small cell lung cancer, mesothelioma, urothelial carcinoma, epithelial ovarian cancer and thymic carcinoma. Alimta is indicated in combination with cisplatin therapy for the initial treatment of patients with locally advanced or metastatic non-squamous non-small cell lung cancer, and in combination with pembrolizumab and platinum chemotherapy for the initial treatment of patients with metastatic non- squamous non-small cell lung cancer and with no EGFR or ALK genomic tumor aberrations. Alimta is indicated for the maintenance treatment of patients with locally advanced or metastatic non-squamous non-small cell lung cancer whose disease has not progressed after four cycles of platinum-based first- line chemotherapy. Alimta is indicated as a single agent for the treatment of patients with recurrent, metastatic non- squamous non-small cell lung cancer after prior chemotherapy. Alimta in combination with cisplatin is indicated for the treatment of patients with malignant pleural mesothelioma whose disease is unresectable or who are otherwise not candidates for curative surgery. ***Note: This Medical Policy is complex and technical. For questions concerning the technical language and/or specific clinical indications for its use, please consult your physician. Policy ® BCBSNC will provide coverage for Pemetrexed (Alimta ) when it is determined to be medically necessary because the medical criteria and guidelines noted below are met. Benefits Application This medical policy relates only to the services or supplies described herein.
    [Show full text]
  • Original Article Pemetrexed and Cyclophosphamide Combination Therapy for the Treatment of Non-Small Cell Lung Cancer
    Int J Clin Exp Pathol 2015;8(11):14693-14700 www.ijcep.com /ISSN:1936-2625/IJCEP0012945 Original Article Pemetrexed and cyclophosphamide combination therapy for the treatment of non-small cell lung cancer Dong Li1, Song He2 1Department of Cardiothoracic Surgery, Central Hospital of Zibo, Zibo 250012, Shandong, China; 2Maanshan Center for Clinical Laboratory, Maanshan Municipal Hospital Group, Maanshan, Anhui, China Received July 15, 2015; Accepted October 13, 2015; Epub November 1, 2015; Published November 15, 2015 Abstract: Lung cancer is the leading cause of cancer-related mortality. This study was undertaken to investigate the efficacy and safety of adding regulatory T cell inhibitor cyclophosphamide to pemetrexed therapy for the second-line treatment of NSCLC with wild-type epidermal growth factor receptor (EGFR). A total of 70 patients were screened between March 2011 and December 2013, out of which 62 patients were enrolled in the study. Patients were randomized to receive 500 mg/m2 pemetrexed in combination with 20 mg/kg cyclophosphamide in a 21 day cycle (n=30) or 500 mg/m2 pemetrexed (n=32), and followed up for 30 months. Disease progression was observed in 23 patients in the pemetrexed plus cyclophosphamide arm and 27 patients in the pemetrexed monotherapy arm. Median progression-free survival was 3.6 months (95% confidence interval [CI], 1.3 to 5.9 months) in the peme- trexed plus cyclophosphamide arm and 2.2 months (95% CI, 1.3 to 3.1 months) in the pemetrexed monotherapy arm. The 6-month PFS rates were 22% (95% CI, 10 to 34) and 14.5% (95% CI, 6 to 23) in the pemetrexed plus cyclophosphamide arm and pemetrexed monotherapy arm, respectively.
    [Show full text]
  • A Preclinical Evaluation of Pemetrexed and Irinotecan Combination As Second-Line Chemotherapy in Pancreatic Cancer
    British Journal of Cancer (2007) 96, 1358 – 1367 & 2007 Cancer Research UK All rights reserved 0007 – 0920/07 $30.00 www.bjcancer.com A preclinical evaluation of pemetrexed and irinotecan combination as second-line chemotherapy in pancreatic cancer A Mercalli1, V Sordi1, R Formicola1, M Dandrea2, S Beghelli2, A Scarpa2, V Di Carlo1, M Reni3,4 and L Piemonti*,1,4 1 2 Laboratory of Experimental Surgery, San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy; Section of Anatomic Pathology, Department 3 of Pathology, University of Verona, Strada Le Grazie 8, Verona 37134, Italy; Department of Oncology, San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy Translational Therapeutics Gemcitabine (GEM)-based chemotherapy is regarded as the standard treatment of pancreatic adenocarcinoma, but yields a very limited disease control. Very few studies have investigated salvage chemotherapy after failure of GEM or GEM-containing chemotherapy and preclinical studies attempting to widen the therapeutic armamentarium, not including GEM, are warranted. MIA PaCa2, CFPAC-1 and Capan-1 pancreatic cancer cell lines were treated with GEM, fluouracil (5-FU), docetaxel (DCT), oxaliplatin (OXP), irinotecan (CPT-11), pemetrexed (PMX) and raltitrexed (RTX) as single agent. Pemetrexed, inducing apoptosis with IC50s under the Cmax in the three lines tested, appeared the most effective drug as single agent. Based on these results, schedule- and concentration-dependent drug interactions (assessed using the combination index) of PMX/GEM, PMX/DCT and PMX–CPT-11 were evaluated. The combinatory study clearly indicated the PMX and CPT-11 combination as the most active against pancreatic cancer. To confirm the efficacy of PMX–CPT-11 combination, we extended the study to a panel of 10 pancreatic cancer cell lines using clinically relevant concentrations (PMX 10 mM; CPT-11 1 mm).
    [Show full text]
  • Cancer Drug Pharmacology Table
    CANCER DRUG PHARMACOLOGY TABLE Cytotoxic Chemotherapy Drugs are classified according to the BC Cancer Drug Manual Monographs, unless otherwise specified (see asterisks). Subclassifications are in brackets where applicable. Alkylating Agents have reactive groups (usually alkyl) that attach to Antimetabolites are structural analogues of naturally occurring molecules DNA or RNA, leading to interruption in synthesis of DNA, RNA, or required for DNA and RNA synthesis. When substituted for the natural body proteins. substances, they disrupt DNA and RNA synthesis. bendamustine (nitrogen mustard) azacitidine (pyrimidine analogue) busulfan (alkyl sulfonate) capecitabine (pyrimidine analogue) carboplatin (platinum) cladribine (adenosine analogue) carmustine (nitrosurea) cytarabine (pyrimidine analogue) chlorambucil (nitrogen mustard) fludarabine (purine analogue) cisplatin (platinum) fluorouracil (pyrimidine analogue) cyclophosphamide (nitrogen mustard) gemcitabine (pyrimidine analogue) dacarbazine (triazine) mercaptopurine (purine analogue) estramustine (nitrogen mustard with 17-beta-estradiol) methotrexate (folate analogue) hydroxyurea pralatrexate (folate analogue) ifosfamide (nitrogen mustard) pemetrexed (folate analogue) lomustine (nitrosurea) pentostatin (purine analogue) mechlorethamine (nitrogen mustard) raltitrexed (folate analogue) melphalan (nitrogen mustard) thioguanine (purine analogue) oxaliplatin (platinum) trifluridine-tipiracil (pyrimidine analogue/thymidine phosphorylase procarbazine (triazine) inhibitor)
    [Show full text]
  • BC Cancer Benefit Drug List September 2021
    Page 1 of 65 BC Cancer Benefit Drug List September 2021 DEFINITIONS Class I Reimbursed for active cancer or approved treatment or approved indication only. Reimbursed for approved indications only. Completion of the BC Cancer Compassionate Access Program Application (formerly Undesignated Indication Form) is necessary to Restricted Funding (R) provide the appropriate clinical information for each patient. NOTES 1. BC Cancer will reimburse, to the Communities Oncology Network hospital pharmacy, the actual acquisition cost of a Benefit Drug, up to the maximum price as determined by BC Cancer, based on the current brand and contract price. Please contact the OSCAR Hotline at 1-888-355-0355 if more information is required. 2. Not Otherwise Specified (NOS) code only applicable to Class I drugs where indicated. 3. Intrahepatic use of chemotherapy drugs is not reimbursable unless specified. 4. For queries regarding other indications not specified, please contact the BC Cancer Compassionate Access Program Office at 604.877.6000 x 6277 or [email protected] DOSAGE TUMOUR PROTOCOL DRUG APPROVED INDICATIONS CLASS NOTES FORM SITE CODES Therapy for Metastatic Castration-Sensitive Prostate Cancer using abiraterone tablet Genitourinary UGUMCSPABI* R Abiraterone and Prednisone Palliative Therapy for Metastatic Castration Resistant Prostate Cancer abiraterone tablet Genitourinary UGUPABI R Using Abiraterone and prednisone acitretin capsule Lymphoma reversal of early dysplastic and neoplastic stem changes LYNOS I first-line treatment of epidermal
    [Show full text]
  • Arsenic Trioxide Targets MTHFD1 and SUMO-Dependent Nuclear De Novo Thymidylate Biosynthesis
    Arsenic trioxide targets MTHFD1 and SUMO-dependent PNAS PLUS nuclear de novo thymidylate biosynthesis Elena Kamyninaa, Erica R. Lachenauera,b, Aislyn C. DiRisioa, Rebecca P. Liebenthala, Martha S. Fielda, and Patrick J. Stovera,b,c,1 aDivision of Nutritional Sciences, Cornell University, Ithaca, NY 14853; bGraduate Field of Biology and Biomedical Sciences, Cornell University, Ithaca, NY 14853; and cGraduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853 Contributed by Patrick J. Stover, February 12, 2017 (sent for review December 1, 2016; reviewed by I. David Goldman and Anne Parle-McDermott) Arsenic exposure increases risk for cancers and is teratogenic in levels. Decreased rates of de novo dTMP synthesis can be caused animal models. Here we demonstrate that small ubiquitin-like by the action of chemotherapeutic drugs (19), through inborn modifier (SUMO)- and folate-dependent nuclear de novo thymidylate errors of folate transport and metabolism (15, 18, 20, 21), by (dTMP) biosynthesis is a sensitive target of arsenic trioxide (As2O3), inhibiting translocation of the dTMP synthesis pathway enzymes leading to uracil misincorporation into DNA and genome instability. into the nucleus (2) and by dietary folate deficiency (22, 23). Im- Methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) and serine paired dTMP synthesis leads to genome instability through well- hydroxymethyltransferase (SHMT) generate 5,10-methylenetetrahy- characterized mechanisms associated with uracil misincorporation drofolate for de novo dTMP biosynthesis and translocate to the nu- into nuclear DNA and subsequent futile cycles of DNA repair (24, cleus during S-phase, where they form a multienzyme complex with 25). Nuclear DNA is surveyed for the presence of uracil by a thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), as family of uracil glycosylases including: uracil N-glycolase (UNG), well as the components of the DNA replication machinery.
    [Show full text]
  • Resistance to Antifolates
    Oncogene (2003) 22, 7431–7457 & 2003 Nature Publishing Group All rights reserved 0950-9232/03 $25.00 www.nature.com/onc Resistance to antifolates Rongbao Zhao1 and I David Goldman*,1 1Departments of Medicine and Molecular, Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA The antifolates were the first class of antimetabolites to the kinetics of the interaction between MTX and DHFR enter the clinics more than 50 years ago. Over the was fully understood, and not until the late 1970s and following decades, a full understanding of their mechan- early 1980s when polyglutamate derivatives of MTX were isms of action and chemotherapeutic potential evolved detected and their pharmacologic importance clarified. along with the mechanisms by which cells develop Likewise, an understanding of tumor cell resistance to resistance to these drugs. These principals served as a antifolates evolved slowly, often paralleling the emergence basis for the subsequent exploration and understanding of of new molecular concepts. As the mechanisms of the mechanisms of resistance to a variety of diverse resistance to antifolates were characterized, this provided antineoplastics with different cellular targets. This section insights and principles that were broadly applicable to describes the bases for intrinsic and acquired antifolate other antineoplastics. Ultimately, this knowledge led to the resistance within the context of the current understanding development of a new generation of antifolates, in the late of the mechanisms of actions and cytotoxic determinants 1980s and 1990s, which are potent direct inhibitors of of these agents. This encompasses impaired drug transport tetrahydrofolate (THF)-cofactor-dependent enzymes. Sev- into cells, augmented drug export, impaired activation of eral of these drugs are now in clinical trials, and the antifolates through polyglutamylation, augmented hydro- activity of one, pemetrexed, has been confirmed in a large lysis of antifolate polyglutamates, increased expression Phase III trial (Vogelzang et al., 2003).
    [Show full text]
  • DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents
    molecules Review DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents Maria Valeria Raimondi 1,*,† , Ornella Randazzo 1,†, Mery La Franca 1 , Giampaolo Barone 1 , Elisa Vignoni 2, Daniela Rossi 2 and Simona Collina 2,* 1 Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; [email protected] (O.R.); [email protected] (M.L.F.); [email protected] (G.B.) 2 Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, via Taramelli 12, 27100 Pavia, Italy; [email protected] (E.V.); [email protected] (D.R.) * Correspondence: [email protected] (M.V.R.); [email protected] (S.C.); Tel.: +390-912-389-1915 (M.V.R.); +390-382-987-379 (S.C.) † These Authors contributed equally to this work. Academic Editors: Simona Collina and Mariarosaria Miloso Received: 25 February 2019; Accepted: 20 March 2019; Published: 22 March 2019 Abstract: Dihydrofolate reductase inhibitors are an important class of drugs, as evidenced by their use as antibacterial, antimalarial, antifungal, and anticancer agents. Progress in understanding the biochemical basis of mechanisms responsible for enzyme selectivity and antiproliferative effects has renewed the interest in antifolates for cancer chemotherapy and prompted the medicinal chemistry community to develop novel and selective human DHFR inhibitors, thus leading to a new generation of DHFR inhibitors. This work summarizes the mechanism of action, chemical, and anticancer profile of the DHFR inhibitors discovered in the last six years. New strategies in DHFR drug discovery are also provided, in order to thoroughly delineate the current landscape for medicinal chemists interested in furthering this study in the anticancer field.
    [Show full text]
  • Protein Kinase C As a Therapeutic Target in Non-Small Cell Lung Cancer
    International Journal of Molecular Sciences Review Protein Kinase C as a Therapeutic Target in Non-Small Cell Lung Cancer Mohammad Mojtaba Sadeghi 1,2, Mohamed F. Salama 2,3,4 and Yusuf A. Hannun 1,2,3,* 1 Department of Biochemistry, Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA; [email protected] 2 Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY 11794, USA; [email protected] 3 Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA 4 Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Dakahlia Governorate, Egypt * Correspondence: [email protected] Abstract: Driver-directed therapeutics have revolutionized cancer treatment, presenting similar or better efficacy compared to traditional chemotherapy and substantially improving quality of life. Despite significant advances, targeted therapy is greatly limited by resistance acquisition, which emerges in nearly all patients receiving treatment. As a result, identifying the molecular modulators of resistance is of great interest. Recent work has implicated protein kinase C (PKC) isozymes as mediators of drug resistance in non-small cell lung cancer (NSCLC). Importantly, previous findings on PKC have implicated this family of enzymes in both tumor-promotive and tumor-suppressive biology in various tissues. Here, we review the biological role of PKC isozymes in NSCLC through extensive analysis of cell-line-based studies to better understand the rationale for PKC inhibition. Citation: Sadeghi, M.M.; Salama, PKC isoforms α, ", η, ι, ζ upregulation has been reported in lung cancer, and overexpression correlates M.F.; Hannun, Y.A. Protein Kinase C with worse prognosis in NSCLC patients.
    [Show full text]
  • Toward a Better Understanding of Methotrexate
    ARTHRITIS & RHEUMATISM Vol. 50, No. 5, May 2004, pp 1370–1382 DOI 10.1002/art.20278 © 2004, American College of Rheumatology REVIEW Toward a Better Understanding of Methotrexate Joel M. Kremer More is known about the metabolism, toxicity, literature. It is the goal of this contribution to review the pharmacokinetics, and clinical profile of methotrexate major and significant concepts regarding MTX metabo- (MTX) than any other drug currently in use in either lism which may be relevant to the treatment of rheu- rheumatology or oncology. In the 56 years since Farber matic disease, not to summarize publications about its et al first described clinical remissions in children with clinical effects. It will become apparent in the course of acute leukemia after treatment with the folate antago- the discussion that most of what we know about the nist aminopterin (1), antifolate drugs, dominated by metabolism of MTX is derived from the oncology liter- MTX, have been used to treat millions of patients with ature. Clinicians who have become familiar with the malignant and autoimmune diseases. It is estimated that concepts presented should be better equipped to pre- MTX is now prescribed to at least 500,000 patients with scribe the drug in a more effective and rational manner. rheumatoid arthritis (RA) worldwide, making it by far In addition, emerging insights into the role of naturally the most commonly used disease-modifying antirheu- occurring genetic variations in cellular pathways of MTX matic drug (DMARD). Indeed, MTX is prescribed for metabolism hold promise for predicting both efficacy more patients with RA than are all of the biologic drugs and toxicity of the drug.
    [Show full text]