1
|
Zheng Y, Cheng H, Jiang S, Tai W. Fc Multisite Conjugation and Prolonged Delivery of the Folate-Targeted Drug Conjugate EC140. Bioconjug Chem 2025; 36:762-769. [PMID: 40178505 DOI: 10.1021/acs.bioconjchem.5c00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Small molecule-drug conjugate (SMDC) is a targeted drug delivery technology that develops in parallel with the antibody-drug conjugate. However, the clinical translation of SMDC faces challenges due to its limited circulating half-life in vivo. The drawback in pharmacokinetics is that it restricts the exposure time of SMDC to tumor tissues and ultimately reduces the therapeutic efficacy. In this study, we chemically conjugated a folate-targeted SMDC EC140 to the long-circulating Fc protein at multiple sites, yielding a stable and high-DAR Fc-SMDC conjugate (Fc-EC140). Fc-EC140 can bear approximately 4 molecules of EC140 per Fc protein (drug-antibody ratio = 4.1) and display enhanced potency in folate receptor (FR)-positive tumor cells compared to the SMDC comparator. In addition, Fc-EC140 retains the FcRn-mediated recycling function and displays an extended half-life of 28 h in the mice. In vivo, antitumor experiments demonstrate that intravenous administration of Fc-EC140 (Q7D × 3 at a dose of 15 mg/kg) nearly cures the KB tumors, which is far more effective than the comparator EC140 administrated at equivalent doses. This study presents a new strategy for the targeted delivery of SMDC.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Sibo Jiang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, China
| | - Wanyi Tai
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
2
|
Bocci M, Zana A, Principi L, Lucaroni L, Prati L, Gilardoni E, Neri D, Cazzamalli S, Galbiati A. In vivo activation of FAP-cleavable small molecule-drug conjugates for the targeted delivery of camptothecins and tubulin poisons to the tumor microenvironment. J Control Release 2024; 367:779-790. [PMID: 38346501 DOI: 10.1016/j.jconrel.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Small molecule-drug conjugates (SMDCs) are increasingly considered as a therapeutic alternative to antibody-drug conjugates (ADCs) for cancer therapy. OncoFAP is an ultra-high affinity ligand of Fibroblast Activation Protein (FAP), a stromal tumor-associated antigen overexpressed in a wide variety of solid human malignancies. We have recently reported the development of non-internalizing OncoFAP-based SMDCs, which are activated by FAP thanks to selective proteolytic cleavage of the -GlyPro- linker with consequent release of monomethyl auristatin E (MMAE) in the tumor microenvironment. In this article, we describe the generation and the in vivo characterization of FAP-cleavable OncoFAP-drug conjugates based on potent topoisomerase I inhibitors (DXd, SN-38, and exatecan) and an anti-tubulin payload (MMAE), which are already exploited in clinical-stage and approved ADCs. The Glycine-Proline FAP-cleavable technology was directly benchmarked against linkers found in Adcetris™, Enhertu™, and Trodelvy™ structures by means of in vivo therapeutic experiments in mice bearing tumors with cellular or stromal FAP expression. OncoFAP-GlyPro-Exatecan and OncoFAP-GlyPro-MMAE emerged as the most efficacious anti-cancer therapeutics against FAP-positive cellular models. OncoFAP-GlyPro-MMAE exhibited a potent antitumor activity also against stromal models, and was therefore selected for clinical development.
Collapse
Affiliation(s)
- Matilde Bocci
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| | - Aureliano Zana
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Laura Lucaroni
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | - Luca Prati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Dario Neri
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, Zurich CH-8093, Switzerland; Philogen S.p.A., Siena 53100, Italy
| | | | - Andrea Galbiati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| |
Collapse
|
3
|
Collyer SE, Stack GD, Walsh JJ. Selective delivery of clinically approved tubulin binding agents through covalent conjugation to an active targeting moiety. Curr Med Chem 2022; 29:5179-5211. [DOI: 10.2174/0929867329666220401105929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
The efficacy and tolerability of tubulin binding agents are hampered by their low specificity for cancer cells, like most clinically used anticancer agents. To improve specificity, tubulin binding agents have been covalently conjugated to agents which target cancer cells to give actively targeted drug conjugates. These conjugates are designed to increase uptake of the drug by cancer cells, while having limited uptake by normal cells thereby improving efficacy and tolerability.
Approaches used include attachment to small molecules, polysaccharides, peptides, proteins and antibodies that exploit the overexpression of receptors for these substances. Antibody targeted strategies have been the most successful to date with six such examples having gained clinical approval. Many other conjugate types, especially those targeting the folate receptor, have shown promising efficacy and toxicity profiles in pre-clinical models and in early-stage clinical studies. Presented herein is a discussion of the success or otherwise of the recent strategies used to form these actively targeted conjugates.
Collapse
Affiliation(s)
- Samuel E. Collyer
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Gary D. Stack
- Department of Nursing and Healthcare, Technological University of the Shannon: Midlands Midwest, Athlone, Ireland
| | - John J. Walsh
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Martín-Sabroso C, Torres-Suárez AI, Alonso-González M, Fernández-Carballido A, Fraguas-Sánchez AI. Active Targeted Nanoformulations via Folate Receptors: State of the Art and Future Perspectives. Pharmaceutics 2021; 14:14. [PMID: 35056911 PMCID: PMC8781617 DOI: 10.3390/pharmaceutics14010014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023] Open
Abstract
In normal tissues, the expression of folate receptors is low and limited to cells that are important for embryonic development or for folate reabsorption. However, in several pathological conditions some cells, such as cancer cells and activated macrophages, overexpress folate receptors (FRs). This overexpression makes them a potential therapeutic target in the treatment of cancer and inflammatory diseases to obtain a selective delivery of drugs at altered cells level, and thus to improve the therapeutic efficacy and decrease the systemic toxicity of the pharmacological treatments. Two strategies have been used to achieve this folate receptor targeting: (i) the use of ligands with high affinity to FRs (e.g., folic acid or anti-FRs monoclonal antibodies) linked to the therapeutic agents or (ii) the use of nanocarriers whose surface is decorated with these ligands and in which the drug is encapsulated. This manuscript analyzes the use of FRs as a target to develop new therapeutic tools in the treatment of cancer and inflammatory diseases with an emphasis on the nanoformulations that have been developed for both therapeutic and imaging purposes.
Collapse
Affiliation(s)
- Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain; (C.M.-S.); (A.I.T.-S.); (M.A.-G.); (A.F.-C.)
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain; (C.M.-S.); (A.I.T.-S.); (M.A.-G.); (A.F.-C.)
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| | - Mario Alonso-González
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain; (C.M.-S.); (A.I.T.-S.); (M.A.-G.); (A.F.-C.)
| | - Ana Fernández-Carballido
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain; (C.M.-S.); (A.I.T.-S.); (M.A.-G.); (A.F.-C.)
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| | - Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain; (C.M.-S.); (A.I.T.-S.); (M.A.-G.); (A.F.-C.)
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
5
|
Zhou Y, Mowlazadeh Haghighi S, Liu Z, Wang L, Hruby VJ, Cai M. Development of Ligand-Drug Conjugates Targeting Melanoma through the Overexpressed Melanocortin 1 Receptor. ACS Pharmacol Transl Sci 2020; 3:921-930. [PMID: 33073191 DOI: 10.1021/acsptsci.0c00072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Melanoma is a lethal form of skin cancer. Despite recent breakthroughs of BRAF-V600E and PD-1 inhibitors showing remarkable clinical responses, melanoma can eventually survive these targeted therapies and become resistant. To solve the drug resistance issue, we designed and synthesized ligand-drug conjugates that couple cytotoxic drugs, which have a low cancer resistance issue, with the melanocortin 1 receptor (MC1R) agonist melanotan-II (MT-II), which provides specificity to MC1R-overexpressing melanoma. The drug-MT-II conjugates maintain strong binding interactions to MC1R and induce selective drug delivery to A375 melanoma cells through its MT-II moiety in vitro. Furthermore, using camptothecin as the cytotoxic drug, camptothecin-MT-II (compound 1) can effectively inhibit A375 melanoma cell growth with an IC50 of 16 nM. By providing selectivity to melanoma cells through its MT-II moiety, this approach of drug-MT-II conjugates enables us to have many more options for cytotoxic drug selection, which can be the key to solving the cancer resistant problem for melanoma.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Saghar Mowlazadeh Haghighi
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Zekun Liu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Lingzhi Wang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Minying Cai
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
6
|
He R, Pan J, Mayer JP, Liu F. The Chemical Methods of Disulfide Bond Formation and Their Applications to Drug Conjugates. CURR ORG CHEM 2020. [DOI: 10.2174/1385272823666191202111723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
The disulfide bond possesses unique chemical and biophysical properties which
distinguish it as one of the key structural elements of bioactive proteins and peptides, important
drugs and other materials. The chemo-selective synthesis of these structures and
the exploration of their function have been of longstanding interest to the chemistry community.
The past decades have witnessed significant progress in both areas. This review
will summarize the historically established and recently developed chemical methods in
disulfide bond formation. The discussion will also be extended to the use of the disulfide
linkers in small molecules, and peptide- and protein-drug conjugates. It is hoped that the
combined overview of the fundamental chemistries and applications to drug discovery
will inspire creative thinking and stimulate future novel uses of these versatile chemistries.
Collapse
Affiliation(s)
- Rongjun He
- Novo Nordisk Research Center Indianapolis, 5225 Exploration Drive, Indianapolis, IN 46241, United States
| | - Jia Pan
- Novo Nordisk Research Centre China, 20 Life Science Road, Beijing, China
| | - John P. Mayer
- Department of Molecular, Developmental & Cell Biology, University of Colorado, Boulder, CO 80309, United States
| | - Fa Liu
- Novo Nordisk Research Center, 530 Fairview Avenue North, Seattle, WA 98109, United States
| |
Collapse
|
7
|
Narmani A, Rezvani M, Farhood B, Darkhor P, Mohammadnejad J, Amini B, Refahi S, Abdi Goushbolagh N. Folic acid functionalized nanoparticles as pharmaceutical carriers in drug delivery systems. Drug Dev Res 2019; 80:404-424. [PMID: 31140629 DOI: 10.1002/ddr.21545] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/02/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
Conventional chemotherapeutic approaches in cancer therapy such as surgery, chemotherapy, and radiotherapy have several disadvantages due to their nontargeted distributions in the whole body. On the other hand, nanoparticles (NPs) based therapies are remarkably progressing to solve several limitations of conventional drug delivery systems (DDSs) including nonspecific biodistribution and targeting, poor water solubility, weak bioavailability and biodegradability, low pharmacokinetic properties, and so forth. The enhanced permeability and retention effect escape from P-glycoprotein trap in cancer cells as a passive targeting mechanism, and active targeting strategies are also other most important advantages of NPs in cancer diagnosis and therapy. Folic acid (FA) is one of the biologic molecules which has been targeted overexpressed-folic acid receptor (FR) on the surface of cancer cells. Therefore, conjugation of FA to NPs most easily enhances the FR-mediated targeting delivery of therapeutic agents. Here, the recent works in FA which have been decorated NPs-based DDSs are discussed and cancer therapy potency of these NPs in clinical trials are presented.
Collapse
Affiliation(s)
- Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Melina Rezvani
- Department of Biology, Faculty of Sciences, Payame Noor University, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Parvaneh Darkhor
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Bahram Amini
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Soheila Refahi
- Department of Medical Physics, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nouraddin Abdi Goushbolagh
- Department of Medical Physics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
8
|
Liu T, Chen S, Wu X, Han H, Zhang S, Wu P, Su X, Wu T, Yu S, Cai X. Folate-Targeted pH and Redox Dual Stimulation-Responsive Nanocarrier for Codelivering of Docetaxel and TFPI-2 for Nasopharyngeal Carcinoma Therapy. ACS APPLIED BIO MATERIALS 2019; 2:1830-1841. [PMID: 35030673 DOI: 10.1021/acsabm.8b00675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Due to the increasing incidence of tumor metastasis and multidrug resistance, even though a combined use of chemotherapy and radiotherapy is introduced, the 5-year average survival rate of an advanced nasopharyngeal carcinoma (NPC) patient still remains low. Hence, targeted slow-release anticancer drugs represent a potential therapy for advanced NPC. In this study, pH and redox dual stimulation-responsive folate-targeted folic acid - β-cyclodextrin - hyperbranched poly(amido amine)s (FA-DS-PAAs) nanocarriers for codelivery of docetaxel (DOC) and tissue factor pathway inhibitor 2 (TFPI-2) for NPC therapy are discussed. Physical and chemical properties, in vitro DOC-release properties, folic acid (FA)-targeting, transfection, Western blotting, DOC and TFPI-2 codelivery, therapeutic properties, targeted inhibition, and biocompatibility, in vivo FA-targeting, toxicity, and therapeutic properties of FA-DS-PAAs/DOC/TFPI2 nanoparticles are evaluated. The results indicate that the 200 nm low-toxicity FA-DS-PAAs/DOC/TFPI2 nanoparticles could enhance TFPI2 gene expression, make cancer cells more sensitive to DOC, induce cell apoptosis, and reduce cell invasion more effectively compared with monochemotherapy. With respect to the targeted release of drugs (DOC and TFPI2) in tumor cells, FA-DS-PAAs/DOC/TFPI2 is associated with the slowest growth rate of tumor and the smallest volume of tumor, so this study demonstrates the best synergetic antitumor effect. We anticipate that this study is important because it not only provides a potential new therapy approach for NPC but also paves the preclinical way for potential application of FA-DS-PAAs/DOC/TFPI2.
Collapse
Affiliation(s)
- Tao Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Shaohua Chen
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Xidong Wu
- Department of Pharmacology, Jiangxi Testing Center of Medical Instruments, No. 181, Nanjing East Road, 330029, Nanchang, P. R. China
| | - Hong Han
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Siyi Zhang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Peina Wu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Xiaomei Su
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Second Road, 510080, Guangzhou, P.R. China
| | - Ting Wu
- Department of Light Chemical Engineering, Guangdong Polytechnic, No. 20, Lanshi 2th Road, 528041, Chancheng District, Foshan, P.R. China
| | - Shaobin Yu
- The No. 1 Surgery Department, No. 5 People's Hospital of Foshan, No. 63, Xiqiao Zhen Jiang Pu Dong Road, 528211, Nanhai District, Foshan, Guangdong Province, P.R. China
| | - Xiang Cai
- Department of Light Chemical Engineering, Guangdong Polytechnic, No. 20, Lanshi 2th Road, 528041, Chancheng District, Foshan, P.R. China
| |
Collapse
|
9
|
Tahmasbi Rad A, Chen CW, Aresh W, Xia Y, Lai PS, Nieh MP. Combinational Effects of Active Targeting, Shape, and Enhanced Permeability and Retention for Cancer Theranostic Nanocarriers. ACS APPLIED MATERIALS & INTERFACES 2019; 11:10505-10519. [PMID: 30793580 DOI: 10.1021/acsami.8b21609] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Combinatory modulation of the physical and biochemical characteristics of nanocarrier delivery systems is an emergent topic in the field of nanomedicine. Here, we studied the combined effects of incorporation of active targeting moieties into nanocarriers and their morphology affecting the enhanced permeation and retention effect for nanomedicine cancer therapy. Self-assembled lipid discoidal and vesicular nanoparticles with low-polydispersity sub-50 nm size range and identical chemical compositions were synthesized, characterized, and correlated with in vitro cancer cellular internalization, in vivo tumor accumulation and cancer treatments. The fact that folate-associated bicelle yields the best outcome is indicative of the preference for discoidal carriers over spherical carriers and the improved targeting efficacy due to the targeting ligand/receptor binding. The approach is successfully adopted to design the nanocarriers for photodynamic therapy, which yields a consistent trend in in vitro and in vivo efficacy: folate nanodiscs > folate vesicles > nonfolate nanodiscs > nonfolate vesicles. Folate discs not only have shown a higher tumor uptake and photothermal therapeutic efficiency, but also minimize skin photosensitivity side effects. The advantages of nanodiscoidal bicelles as nanocarriers, including well-defined size, robust formation, easy encapsulation of hydrophobic molecules (therapeutics and/or diagnostics), easy incorporation of targeting molecules, and low toxicity, enable the scalable manufacturing of a generalized in vivo multimodal delivery platform.
Collapse
Affiliation(s)
- Armin Tahmasbi Rad
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| | - Ching-Wen Chen
- Department of Chemistry , National Chung Hsing University , Taichung 402 , Taiwan , ROC
| | - Wafa Aresh
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| | | | - Ping-Shan Lai
- Department of Chemistry , National Chung Hsing University , Taichung 402 , Taiwan , ROC
| | - Mu-Ping Nieh
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| |
Collapse
|
10
|
Szigetvari NM, Dhawan D, Ramos-Vara JA, Leamon CP, Klein PJ, Ruple AA, Heng HG, Pugh MR, Rao S, Vlahov IR, Deshuillers PL, Low PS, Fourez LM, Cournoyer AM, Knapp DW. Phase I/II clinical trial of the targeted chemotherapeutic drug, folate-tubulysin, in dogs with naturally-occurring invasive urothelial carcinoma. Oncotarget 2018; 9:37042-37053. [PMID: 30651934 PMCID: PMC6319348 DOI: 10.18632/oncotarget.26455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/26/2018] [Indexed: 01/01/2023] Open
Abstract
Purpose The purpose was to determine the safety and antitumor activity of a folate-tubulysin conjugate (EC0531) in a relevant preclinical animal model, dogs with naturally-occurring invasive urothelial carcinoma (iUC). Canine iUC is an aggressive cancer with high folate receptor (FR) expression similar to that in certain forms of human cancer. Experimental Design A 3+3 dose escalation study of EC0531 (starting dose 0.2 mg/kg given intravenously at two-week intervals) was performed in dogs with iUC expressing high levels of FRs (>50% positive tumor cells). Pharmacokinetic (PK) analysis was performed, and the maximum tolerated dose (MTD) was determined. The dose cohort at the MTD was expanded to determine antitumor activity. Results The MTD of EC0531 was 0.26 mg/kg every two weeks, with grade 3-4 neutropenia and gastrointestinal toxicity observed at higher doses. Treatment at the MTD was well tolerated. Clinical benefit was found in 20 of 28 dogs (71%), including three dogs with partial remission and 17 dogs with stable disease. Plasma EC0531 concentrations in the dogs far exceeded those required to inhibit proliferation of FR-expressing cell in vitro. Unlike human neutrophils, canine neutrophils were found to express FRs, which contributes to the neutropenia at higher doses of EC0531 in dogs. Conclusion EC0531 was well tolerated and had good antitumor activity in dogs with iUC. It is likely that humans will tolerate higher, potentially more effective doses of folate-tubulysin without myelotoxicity because of the absence of FRs on human neutrophils. The results clearly justify the evaluation of folate-tubulysin in human clinical trials.
Collapse
Affiliation(s)
- Nicholas M Szigetvari
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Deepika Dhawan
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - José A Ramos-Vara
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | | | | | - A Audrey Ruple
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Hock Gan Heng
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | | | | | | | - Pierre L Deshuillers
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Lindsey M Fourez
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Ashleigh M Cournoyer
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Deborah W Knapp
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, West Lafayette, IN, USA
| |
Collapse
|
11
|
Puskas JE, Castano M, Mulay P, Dudipala V, Wesdemiotis C. Method for the Synthesis of γ-PEGylated Folic Acid and Its Fluorescein-Labeled Derivative. Macromolecules 2018. [DOI: 10.1021/acs.macromol.8b01888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
12
|
Merk D, Schubert-Zsilavecz M. The Linker Approach. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527674381.ch8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Daniel Merk
- Goethe University Frankfurt; Institute of Pharmaceutical Chemistry; Max-von-Laue-Str. 9 60438 Frankfurt Germany
| | - Manfred Schubert-Zsilavecz
- Goethe University Frankfurt; Institute of Pharmaceutical Chemistry; Max-von-Laue-Str. 9 60438 Frankfurt Germany
| |
Collapse
|
13
|
Affiliation(s)
- Madduri Srinivasarao
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S. Low
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
14
|
Fraczyk J, Walczak M, Szymanski L, Kolacinski Z, Wrzosek H, Majsterek I, Przybylowska-Sygut K, Kaminski ZJ. Carbon nanotubes functionalized with folic acid attached via biomimetic peptide linker. Nanomedicine (Lond) 2017; 12:2161-2182. [PMID: 28814127 DOI: 10.2217/nnm-2017-0120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
AIM Anchoring folic acid (FA) with a biomimetic peptidic linker resistant to proteolytic degradation to act as a homing device on functionalized carbon nanotubes. MATERIALS & METHODS Ethylenediamine was attached to oxidized multiwalled carbon nanotubes (MWNTs) using 4-(4,6-dimethoxy-[1,3,5]triazin-2-yl)-4-methylmorpholinium tetrafluoroborate. FA was coupled with 6-aminohexanoic acid and derivatives of β-alanine, affording four intermediates, which connected to the MWNTs via peptidic linkers of various lengths. RESULTS Biomimetic nanomaterials were produced with FA as a homing molecule. The structure and properties of the nanomaterials were analyzed, confirming the versatility of the peptides used as linkers. CONCLUSION Conjugates of FA attached to MWNTs via peptide linkers prepared from β-alanine residues are resistant to proteolytic degradation. Viability in colon cancer cells and normal colonocytes confirmed their lack of cytotoxicity.
Collapse
Affiliation(s)
- Justyna Fraczyk
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90924 Lodz, Poland
| | - Malgorzata Walczak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90924 Lodz, Poland
| | - Lukasz Szymanski
- Institute of Mechatronics & Information Systems, Lodz University of Technology, Stefanowskiego 18/22, 90924 Lodz, Poland
| | - Zbigniew Kolacinski
- Institute of Mechatronics & Information Systems, Lodz University of Technology, Stefanowskiego 18/22, 90924 Lodz, Poland
| | - Henryk Wrzosek
- Department of Material & Commodity Sciences & Textile Metrology, Lodz University of Technology, Zeromskiego 116, 90924 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry & Biochemistry, Medical University of Lodz, Plac Hallera 1, 90647 Lodz, Poland
| | - Karolina Przybylowska-Sygut
- Department of Clinical Chemistry & Biochemistry, Medical University of Lodz, Plac Hallera 1, 90647 Lodz, Poland
| | - Zbigniew J Kaminski
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90924 Lodz, Poland
| |
Collapse
|
15
|
Siwowska K, Schmid RM, Cohrs S, Schibli R, Müller C. Folate Receptor-Positive Gynecological Cancer Cells: In Vitro and In Vivo Characterization. Pharmaceuticals (Basel) 2017; 10:ph10030072. [PMID: 28809784 PMCID: PMC5620616 DOI: 10.3390/ph10030072] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 01/10/2023] Open
Abstract
The folate receptor alpha (FR) is expressed in a variety of gynecological cancer types. It has been widely used for tumor targeting with folic acid conjugates of diagnostic and therapeutic probes. The cervical KB tumor cells have evolved as the standard model for preclinical investigations of folate-based (radio) conjugates. In this study, a panel of FR-expressing human cancer cell lines—including cervical (HeLa, KB, KB-V1), ovarian (IGROV-1, SKOV-3, SKOV-3.ip), choriocarcinoma (JAR, BeWo) and endometrial (EFE-184) tumor cells—was investigated in vitro and for their ability to grow as xenografts in mice. FR-expression levels were compared in vitro and in vivo and the cell lines were characterized by determination of the sensitivity towards commonly-used chemotherapeutics and the expression of two additional, relevant tumor markers, HER2 and L1-CAM. It was found that, besides KB cells, its multiresistant KB-V1 subclone as well as the ovarian cancer cell lines, IGROV-1 and SKOV-3.ip, could be used as potentially more relevant preclinical models. They would allow addressing specific questions such as the therapeutic efficacy of FR-targeting agents in tumor (mouse) models of multi-resistance and in mouse models of metastases formation.
Collapse
Affiliation(s)
- Klaudia Siwowska
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, Villigen-PSI 5232, Switzerland.
| | - Raffaella M Schmid
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, Villigen-PSI 5232, Switzerland.
| | - Susan Cohrs
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, Villigen-PSI 5232, Switzerland.
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, Villigen-PSI 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland.
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, Villigen-PSI 5232, Switzerland.
| |
Collapse
|
16
|
König SG, Krämer R. Accessing Structurally Diverse Near-Infrared Cyanine Dyes for Folate Receptor-Targeted Cancer Cell Staining. Chemistry 2017; 23:9306-9312. [PMID: 28339120 DOI: 10.1002/chem.201700026] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Indexed: 11/09/2022]
Abstract
Folate receptor (FR) targeting is one of the most promising strategies for the development of small-molecule-based cancer imaging agents considering that the FR is highly overexpressed on the surface of many cancer cell types. FR-targeted conjugates of near-infrared (NIR) emissive cyanine dyes are in advanced clinical trials for fluorescence-guided surgery and are valuable research tools for optical molecular imaging in animal models. Only a small number of promising conjugates has been evaluated so far. Analysis of structure-performance relations to identify critical factors modulating the performance of targeted conjugates is essential for successful further optimization. This contribution addresses the need for convenient synthetic access to structurally diverse NIR-emissive cyanine dyes for conjugation with folic acid. Structural variations were introduced to readily available cyanine precursors in particular via C-C-coupling reactions including Suzuki and (for the first time with these types of dyes) Sonogashira cross-couplings. Photophysical properties such as absorbance maxima, brightness, and photostability are highly dependent on the molecular structure. Selected modified cyanines were conjugated to folic acid for cancer cell targeting. Several conjugates display a favorable combination of high fluorescence brightness and photostability with high affinity to FR-positive cancer cells, and enable the selective imaging of these cells with low background.
Collapse
Affiliation(s)
- Sandra G König
- Institute of Inorganic Chemistry, Heidelberg University, Im Neuenheimer Feld 274, 69120, Heidelberg, Germany
| | - Roland Krämer
- Institute of Inorganic Chemistry, Heidelberg University, Im Neuenheimer Feld 274, 69120, Heidelberg, Germany
| |
Collapse
|
17
|
Enhancing the therapeutic range of a targeted small-molecule tubulysin conjugate for folate receptor-based cancer therapy. Cancer Chemother Pharmacol 2017; 79:1151-1160. [DOI: 10.1007/s00280-017-3311-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/07/2016] [Indexed: 10/19/2022]
|
18
|
Guertin AD, O'Neil J, Stoeck A, Reddy JA, Cristescu R, Haines BB, Hinton MC, Dorton R, Bloomfield A, Nelson M, Vetzel M, Lejnine S, Nebozhyn M, Zhang T, Loboda A, Picard KL, Schmidt EV, Dussault I, Leamon CP. High Levels of Expression of P-glycoprotein/Multidrug Resistance Protein Result in Resistance to Vintafolide. Mol Cancer Ther 2016; 15:1998-2008. [PMID: 27256377 DOI: 10.1158/1535-7163.mct-15-0950] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/09/2016] [Indexed: 11/16/2022]
|
19
|
Kue CS, Kamkaew A, Burgess K, Kiew LV, Chung LY, Lee HB. Small Molecules for Active Targeting in Cancer. Med Res Rev 2016; 36:494-575. [PMID: 26992114 DOI: 10.1002/med.21387] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/29/2022]
Abstract
For the purpose of this review, active targeting in cancer research encompasses strategies wherein a ligand for a cell surface receptor expressed on tumor cells is used to deliver a cytotoxic or imaging cargo. This area of research is more than two decades old, but in those 20 and more years, how many receptors have been studied extensively? What kinds of the ligands are used for active targeting? Are they mostly naturally occurring molecules such as folic acid, or synthetic substances developed in campaigns for medicinal chemistry efforts? This review outlines the most important receptor or ligand combinations that have been used in active targeting to answer these questions, and therefore to address the most important one of all: is research in active targeting affording diminishing returns, or is this an area for which the potential far exceeds progress made so far?
Collapse
Affiliation(s)
- Chin S Kue
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Anyanee Kamkaew
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Lik V Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lip Y Chung
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hong B Lee
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Kurosaki A, Hasegawa K, Kato T, Abe K, Hanaoka T, Miyara A, O'Shannessy DJ, Somers EB, Yasuda M, Sekino T, Fujiwara K. Serum folate receptor alpha as a biomarker for ovarian cancer: Implications for diagnosis, prognosis and predicting its local tumor expression. Int J Cancer 2015; 138:1994-2002. [PMID: 26595060 DOI: 10.1002/ijc.29937] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/14/2015] [Accepted: 10/26/2015] [Indexed: 11/11/2022]
Abstract
Folate receptor alpha (FRA) is a GPI-anchored glycoprotein and encoded by the FOLR1 gene. High expression of FRA is observed in specific malignant tumors of epithelial origin, including ovarian cancer, but exhibits very limited normal tissue expression, making it as an attractive target for the ovarian cancer therapy. FRA is known to shed from the cell surface into the circulation which allows for its measurement in the serum of patients. Recently, methods to detect the soluble form of FRA have been developed and serum FRA (sFRA) is considered a highly promising biomarker for ovarian cancer. We prospectively investigated the levels of sFRA in patients clinically suspected of having malignant ovarian tumors. A total of 231 patients were enrolled in this study and analyzed for sFRA as well as tumor expression of FRA by immunohistochemistry. High sFRA was predominantly observed in epithelial ovarian cancer patients, but not in patients with benign or borderline gynecological disease or metastatic ovarian tumors from advanced colorectal cancers. Levels of sFRA were highly correlated to clinical stage, tumor grade and histological type and demonstrated superior accuracy for the detection of ovarian cancer than did serum CA125. High sFRA was significantly associated with shorter progression-free survival in both early and advanced ovarian cancer patients. Finally, tumor FRA expression status was strongly correlated with sFRA levels. Taken together, these data suggest that sFRA might be a useful noninvasive serum biomarkers for future clinical trials assessing FRA-targeted therapy.
Collapse
Affiliation(s)
- Akira Kurosaki
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Japan
| | - Tomomi Kato
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kenji Abe
- Department of Research and Development, EIDIA Co. Ltd, Inashiki, Japan
| | - Tatsuya Hanaoka
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Japan
| | - Akiko Miyara
- Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Japan
| | | | - Elizabeth B Somers
- Department of Translation Medicine and Diagnostics, Morphotek, Inc, Exton, PA
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Tetsuo Sekino
- Department of Research and Development, EIDIA Co. Ltd, Inashiki, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Japan
| |
Collapse
|
21
|
Ledermann J, Canevari S, Thigpen T. Targeting the folate receptor: diagnostic and therapeutic approaches to personalize cancer treatments. Ann Oncol 2015; 26:2034-43. [DOI: 10.1093/annonc/mdv250] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 05/22/2015] [Indexed: 11/13/2022] Open
|
22
|
Nogueira E, Mangialavori IC, Loureiro A, Azoia NG, Sárria MP, Nogueira P, Freitas J, Härmark J, Shimanovich U, Rollett A, Lacroix G, Bernardes GJL, Guebitz G, Hebert H, Moreira A, Carmo AM, Rossi JPFC, Gomes AC, Preto A, Cavaco-Paulo A. Peptide Anchor for Folate-Targeted Liposomal Delivery. Biomacromolecules 2015; 16:2904-10. [PMID: 26241560 DOI: 10.1021/acs.biomac.5b00823] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Specific folate receptors are abundantly overexpressed in chronically activated macrophages and in most cancer cells. Directed folate receptor targeting using liposomes is usually achieved using folate linked to a phospholipid or cholesterol anchor. This link is formed using a large spacer like polyethylene glycol. Here, we report an innovative strategy for targeted liposome delivery that uses a hydrophobic fragment of surfactant protein D linked to folate. Our proposed spacer is a small 4 amino acid residue linker. The peptide conjugate inserts deeply into the lipid bilayer without affecting liposomal integrity, with high stability and specificity. To compare the drug delivery potential of both liposomal targeting systems, we encapsulated the nuclear dye Hoechst 34580. The eventual increase in blue fluorescence would only be detectable upon liposome disruption, leading to specific binding of this dye to DNA. Our delivery system was proven to be more efficient (2-fold) in Caco-2 cells than classic systems where the folate moiety is linked to liposomes by polyethylene glycol.
Collapse
Affiliation(s)
- Eugénia Nogueira
- CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho , Braga 4710-057, Portugal.,CEB - Centre of Biological Engineering, University of Minho , Braga 4710-057, Portugal
| | - Irene C Mangialavori
- IQUIFIB - Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET , 1113 Buenos Aires, Argentina
| | - Ana Loureiro
- CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho , Braga 4710-057, Portugal.,CEB - Centre of Biological Engineering, University of Minho , Braga 4710-057, Portugal
| | - Nuno G Azoia
- CEB - Centre of Biological Engineering, University of Minho , Braga 4710-057, Portugal
| | - Marisa P Sárria
- CEB - Centre of Biological Engineering, University of Minho , Braga 4710-057, Portugal
| | - Patrícia Nogueira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular , 4150-180 Porto, Portugal
| | - Jaime Freitas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular , 4150-180 Porto, Portugal
| | - Johan Härmark
- Department of Biosciences and Nutrition, The Royal Institute of Technology, School of Technology and Health, Karolinska Institutet , S-14183 Huddinge, Sweden
| | - Ulyana Shimanovich
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Alexandra Rollett
- Institute of Environmental Biotechnology, University of Natural Resources and Life Sciences , 3430 Tulln, Austria
| | - Ghislaine Lacroix
- INERIS - Institut National de l'Environnement Industriel et des Risques , 60550 Verneuil en Halatte, France
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Georg Guebitz
- Institute of Environmental Biotechnology, University of Natural Resources and Life Sciences , 3430 Tulln, Austria
| | - Hans Hebert
- Department of Biosciences and Nutrition, The Royal Institute of Technology, School of Technology and Health, Karolinska Institutet , S-14183 Huddinge, Sweden
| | - Alexandra Moreira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular , 4150-180 Porto, Portugal
| | - Alexandre M Carmo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular , 4150-180 Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto , 4099-003 Porto, Portugal
| | - Juan Pablo F C Rossi
- IQUIFIB - Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET , 1113 Buenos Aires, Argentina
| | - Andreia C Gomes
- CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho , Braga 4710-057, Portugal
| | - Ana Preto
- CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho , Braga 4710-057, Portugal
| | - Artur Cavaco-Paulo
- CEB - Centre of Biological Engineering, University of Minho , Braga 4710-057, Portugal
| |
Collapse
|
23
|
Vergote I, Leamon CP. Vintafolide: a novel targeted therapy for the treatment of folate receptor expressing tumors. Ther Adv Med Oncol 2015; 7:206-18. [PMID: 26136852 DOI: 10.1177/1758834015584763] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite advances in the development of molecularly targeted therapies, limited improvements in overall survival have been noted among many cancer patients with solid tumors, primarily due to development of drug resistance. Accordingly, there is an unmet need for new targeted therapies and treatment approaches for cancer, especially for overcoming resistance. Expression of the folate receptor is upregulated in many tumor types and thus represents an ideal target for cancer treatment. Several folate receptor targeted therapies are in development, including the small molecule drug conjugate vintafolide, the monoclonal antibody farletuzumab, and the antibody-drug conjugate IMGN853. The role of the folate receptor as a target in cancer progression and resistance as well as emerging preclinical and clinical data from studies on those folate receptor targeted agents that are in development with a focus on vintafolide are reviewed. The folate receptor has several unique properties, such as high expression in several tumor types, that make it a rational target for cancer treatment, and allow for selective delivery of folate receptor targeted agents. Early-stage clinical data in lung and ovarian cancer suggest that vintafolide has the potential for combination with other standard approved agents.
Collapse
|
24
|
Wayua C, Roy J, Putt KS, Low PS. Selective Tumor Targeting of Desacetyl Vinblastine Hydrazide and Tubulysin B via Conjugation to a Cholecystokinin 2 Receptor (CCK2R) Ligand. Mol Pharm 2015; 12:2477-83. [PMID: 26043355 PMCID: PMC4674820 DOI: 10.1021/acs.molpharmaceut.5b00218] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
As the delivery of selectively targeted
cytotoxic agents via antibodies
or small molecule ligands to malignancies has begun to show promise
in the clinic, the need to identify and validate additional cellular
targets for specific therapeutic delivery is critical. Although a
multitude of cancers have been targeted using the folate receptor,
PSMA, bombesin receptor, somatostatin receptor, LHRH, and αvβ3, there is a notable lack of specific small
molecule ligand/receptor pairs to cellular targets found within cancers
of the GI tract. Because of the selective GI tract expression of the
cholecystokinin 2 receptor (CCK2R), we undertook the creation of conjugates
that would deliver microtubule-disrupting drugs to malignancies through
the specific targeting of CCK2R via a high affinity small molecule
ligand. The cytotoxic activity of these conjugates were shown to be
receptor mediated in vitro and in vivo with xenograft mouse models
exhibiting delayed growth or regression of tumors that expressed CCK2R.
Overall, this work demonstrates that ligands to CCK2R can be used
to create selectively targeted therapeutic conjugates.
Collapse
Affiliation(s)
- Charity Wayua
- †Department of Chemistry and ‡Center for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jyoti Roy
- †Department of Chemistry and ‡Center for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Karson S Putt
- †Department of Chemistry and ‡Center for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S Low
- †Department of Chemistry and ‡Center for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
25
|
Haller S, Reber J, Brandt S, Bernhardt P, Groehn V, Schibli R, Müller C. Folate receptor-targeted radionuclide therapy: preclinical investigation of anti-tumor effects and potential radionephropathy. Nucl Med Biol 2015; 42:770-9. [PMID: 26162583 DOI: 10.1016/j.nucmedbio.2015.06.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 05/23/2015] [Accepted: 06/08/2015] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Application of therapeutic folate radioconjugates is a promising option for the treatment of folate receptor (FR)-positive tumors, although high uptake of radiofolates in the kidneys remains a critical issue. Recently, it was shown that enhancing the blood circulation of radiofolates results in increased tumor uptake and reduced retention of radioactivity in the kidneys. In this study, we investigated and compared the anti-tumor effects and potential long-term damage to the kidneys after application of an albumin-binding ((177)Lu-cm09), and a conventional ((177)Lu-EC0800) folate radioconjugate. METHODS In vivo studies were performed with KB tumor-bearing nude mice. (177)Lu-EC0800 and (177)Lu-cm09 were applied at variable quantities (10-30 MBq/mouse), and the tumor growth was monitored over time. Mice without tumors were injected with the same radiofolates and investigated over eight months by determination of creatinine and blood urea nitrogen plasma levels and by measuring renal uptake of (99m)Tc-DMSA using SPECT. At the study end, the morphological changes were examined on renal tissue sections using variable staining methods. RESULTS Compared to untreated controls, dose-dependent tumor growth inhibition and prolonged survival was observed in all treated mice. In line with the resulting absorbed dose, the treatment was more effective with (177)Lu-cm09 than with (177)Lu-EC0800, enabling complete tumor remission after application of ≥20MBq (≥28Gy). Application of radiofolates with an absorbed renal dose ≥23 Gy showed increased levels of renal plasma parameters and reduced renal uptake of (99m)Tc-DSMA. Morphological changes observed on tissue sections confirmed radionephropathy of variable stages. CONCLUSIONS (177)Lu-cm09 showed more favorable anti-tumor effects and significantly less damage to the kidneys compared to (177)Lu-EC0800 as was expected based on improved tumor-to-kidney ratios. It was demonstrated that enhancing the blood circulation time of radiofolates was favorable regarding the risk-benefit profile of a therapeutic application. These results hold promise for future translation of the albumin-binder concept to the clinics, potentially enabling FR-targeted radionuclide therapy in patients.
Collapse
Affiliation(s)
- Stephanie Haller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Josefine Reber
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Simone Brandt
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska Universitetssjukhuset, 41345 Gothenburg, Sweden
| | - Viola Groehn
- Merck and Cie, Laternenacker 5, 8200 Schaffhausen, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.
| |
Collapse
|
26
|
Samain F, Casi G. Small targeted cytotoxics from DNA-encoded chemical libraries. Curr Opin Chem Biol 2015; 26:72-9. [DOI: 10.1016/j.cbpa.2015.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/23/2015] [Accepted: 02/09/2015] [Indexed: 01/09/2023]
|
27
|
Necela BM, Crozier JA, Andorfer CA, Lewis-Tuffin L, Kachergus JM, Geiger XJ, Kalari KR, Serie DJ, Sun Z, Aspita AM, O’Shannessy DJ, Maltzman JD, McCullough AE, Pockaj BA, Cunliffe HE, Ballman KV, Thompson EA, Perez EA. Folate receptor-α (FOLR1) expression and function in triple negative tumors. PLoS One 2015; 10:e0122209. [PMID: 25816016 PMCID: PMC4376802 DOI: 10.1371/journal.pone.0122209] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022] Open
Abstract
Folate receptor alpha (FOLR1) has been identified as a potential prognostic and therapeutic target in a number of cancers. A correlation has been shown between intense overexpression of FOLR1 in breast tumors and poor prognosis, yet there is limited examination of the distribution of FOLR1 across clinically relevant breast cancer subtypes. To explore this further, we used RNA-seq data from multiple patient cohorts to analyze the distribution of FOLR1 mRNA across breast cancer subtypes comprised of estrogen receptor positive (ER+), human epidermal growth factor receptor positive (HER2+), and triple negative (TNBC) tumors. FOLR1 expression varied within breast tumor subtypes; triple negative/basal tumors were significantly associated with increased expression of FOLR1 mRNA, compared to ER+ and HER2+ tumors. However, subsets of high level FOLR1 expressing tumors were observed in all clinical subtypes. These observations were supported by immunohistochemical analysis of tissue microarrays, with the largest number of 3+ positive tumors and highest H-scores of any subtype represented by triple negatives, and lowest by ER+ tumors. FOLR1 expression did not correlate to common clinicopathological parameters such as tumor stage and nodal status. To delineate the importance of FOLR1 overexpression in triple negative cancers, RNA-interference was used to deplete FOLR1 in overexpressing triple negative cell breast lines. Loss of FOLR1 resulted in growth inhibition, whereas FOLR1 overexpression promoted folate uptake and growth advantage in low folate conditions. Taken together, our data suggests patients with triple negative cancers expressing high FOLR1 expression represent an important population of patients that may benefit from targeted anti-FOLR1 therapy. This may prove particularly helpful for a large number of patients who would typically be classified as triple negative and who to this point have been left without any targeted treatment options.
Collapse
Affiliation(s)
- Brian M. Necela
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Jennifer A. Crozier
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Cathy A. Andorfer
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Laura Lewis-Tuffin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Jennifer M. Kachergus
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Xochiquetzal J. Geiger
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Krishna R. Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Daniel J. Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida United States of America
| | - Zhifu Sun
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alvaro Moreno Aspita
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Daniel J. O’Shannessy
- Department of Translational Medicine and Diagnostics, Morphotek, Exton, Pennsylvania, United States of America
| | - Julia D. Maltzman
- Department of Clinical Development, Morphotek, Exton, Pennsylvania, United States of America
| | - Ann E. McCullough
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Barbara A. Pockaj
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Heather E. Cunliffe
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Karla V. Ballman
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - E. Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
- * E-mail:
| | - Edith A. Perez
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| |
Collapse
|
28
|
Nkepang G, Bio M, Rajaputra P, Awuah SG, You Y. Folate receptor-mediated enhanced and specific delivery of far-red light-activatable prodrugs of combretastatin A-4 to FR-positive tumor. Bioconjug Chem 2014; 25:2175-88. [PMID: 25351441 PMCID: PMC4275160 DOI: 10.1021/bc500376j] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
We examined the concept of a novel
prodrug strategy in which anticancer
drug can be locally released by visible/near IR light, taking advantage
of the photodynamic process and photo-unclick chemistry. Our most
recently formulated prodrug of combretastatin A-4, Pc-(L-CA4)2, showed multifunctionality for fluorescence imaging, light-activated
drug release, and the combined effects of PDT and local chemotherapy.
In this formulation, L is a singlet oxygen cleavable linker. Here,
we advanced this multifunctional prodrug by adding a tumor-targeting
group, folic acid (FA). We designed and prepared four FA-conjugated
prodrugs 1–4 (CA4-L-Pc-PEGn-FA: n = 0, 2, 18, ∼45) and one non-FA-conjugated
prodrug 5 (CA4-L-Pc-PEG18-boc). Prodrugs 3 and 4 had a longer PEG spacer and showed higher
hydrophilicity, enhanced uptake to colon 26 cells via FR-mediated
mechanisms, and more specific localization to SC colon 26 tumors in
Balb/c mice than prodrugs 1 and 2. Prodrug 4 also showed higher and more specific uptake to tumors, resulting
in selective tumor damage and more effective antitumor efficacy than
non-FA-conjugated prodrug 5. FR-mediated targeting seemed
to be an effective strategy to spare normal tissues surrounding tumors
in the illuminated area during treatment with this prodrug.
Collapse
Affiliation(s)
- Gregory Nkepang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma 73117, United States
| | | | | | | | | |
Collapse
|
29
|
Ambrosio AJ, Suzin D, Palmer EL, Penson RT. Vintafolide (EC145) for the treatment of folate-receptor-α positive platinum-resistant ovarian cancer. Expert Rev Clin Pharmacol 2014; 7:443-50. [PMID: 24742319 DOI: 10.1586/17512433.2014.909723] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Seminal advances in the treatment of cancer have been achieved because of drug development in ovarian cancer; notably the developments of platinums and taxanes. However, no new drug has been FDA approved for ovarian cancer since 2006, and the approval of an antiangiogenic agent for ovarian cancer in the US has stalled. Predicting the next breakthrough is a high risk and highly expensive venture. One of the most promising prospects is folate-receptor (FR)-targeted therapy, given the high expression in FR ovarian cancer. We review the development of vintafolide (EC145), a folic acid-desacetylvinblastine conjugate, the predictive utility of a FR-targeted imaging agent, technetium-(99)m-etarfolatide (EC20), the challenges in proving survival advantage, and other approaches to exploiting FR as a target in ovarian cancer.
Collapse
Affiliation(s)
- Allison J Ambrosio
- Division of Hematology Oncology, Yawkey 9-064, Massachusetts General Hospital, 32 Fruit Street, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
30
|
Trindade AF, Frade RFM, Maçôas EMS, Graça C, Rodrigues CAB, Martinho JMG, Afonso CAM. "Click and go": simple and fast folic acid conjugation. Org Biomol Chem 2014; 12:3181-90. [PMID: 24723199 DOI: 10.1039/c4ob00150h] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Folic acid targeting by functionalization of the terminal γ-carboxylic acid is one of the most important strategies to selectively deliver chemotherapeutics and dyes to cancer cells which overexpress folate receptors. However, conjugation of folic acid is limited by its unique solubility and by selectivity issues imposing the need for expensive preparative reverse-phase chromatographic purification to isolate γ-folate conjugates. Herein is provided a novel synthetic tool for the synthesis of new folic acid conjugates with excellent γ-purity based on strain-promoted alkyne-azide cycloadditions with a γ-folate-cyclooctyne conjugate 3. To demonstrate the potential of this methodology several new folate conjugates were synthesized with high γ-purity and without using any type of chromatographic purification by reacting conjugate 3 with several fluorescent probes, polymers and siliceous materials bearing azide. In addition, the cycloaddition reaction between conjugate 3 and an azido-derived fluorescent dye was successfully performed in cellular media leading to an increase of fluorescence in the cells which overexpress folate receptors (NCI-H460).
Collapse
Affiliation(s)
- Alexandre F Trindade
- CQFM, Centro de Química-Física Molecular, IN-Institute of Nanosciences and Nanotechnology, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | | | | | | | | | | | | |
Collapse
|
31
|
Morris RT, Joyrich RN, Naumann RW, Shah NP, Maurer AH, Strauss HW, Uszler JM, Symanowski JT, Ellis PR, Harb WA. Phase II study of treatment of advanced ovarian cancer with folate-receptor-targeted therapeutic (vintafolide) and companion SPECT-based imaging agent (99mTc-etarfolatide). Ann Oncol 2014; 25:852-858. [PMID: 24667717 DOI: 10.1093/annonc/mdu024] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND This report examines (99m)Tc-etarfolatide imaging to identify the presence of folate receptor (FR) on tumors of women with recurrent/refractory ovarian or endometrial cancer and correlates expression with response to FR-targeted therapy (vintafolide). PATIENTS AND METHODS In this phase II, single-arm, multicenter study, patients with advanced ovarian cancer were imaged with (99m)Tc-etarfolatide before vintafolide treatment. Up to 10 target lesions (TLs) were selected based on Response Evaluation Criteria In Solid Tumors criteria using computed tomography scans. Single-photon emission computed tomography images of TLs were assessed for (99m)Tc-etarfolatide uptake as either FR positive or negative. Patients were categorized by percentage of TLs positive and grouped as FR(100%), FR(10%-90%), and FR(0%). Lesion and patient response were correlated with etarfolatide uptake. RESULTS Forty-nine patients were enrolled; 43 were available for analysis. One hundred thirty-nine lesions were (99m)Tc-etarfolatide evaluable: 110 FR positive and 29 FR negative. Lesion disease control rate (DCR = stable or response) was observed in 56.4% of FR-positive lesions versus 20.7% of FR-negative lesions (P < 0.001). Patient DCR was 57%, 36%, and 33% in FR(100%), FR(10%-90%), and FR(0%) patients, respectively. Median overall survival was 14.6, 9.6, and 3.0 months in FR(100%), FR(10%-90%), and FR(0%) patients, respectively. CONCLUSIONS Overall response to FR-targeted therapy and DCR correlate with FR positivity demonstrated by (99m)Tc-etarfolatide imaging. CLINICAL TRIAL NUMBER NCT00507741.
Collapse
Affiliation(s)
| | - R N Joyrich
- Detroit Medical Center, Wayne State University, Detroit
| | | | - N P Shah
- Charlotte Radiology, Carolinas Medical Center, Charlotte
| | - A H Maurer
- Temple University Hospital, Philadelphia
| | - H W Strauss
- Memorial Sloan-Kettering Cancer Center, New York
| | - J M Uszler
- Saint John's Health Center, Santa Monica
| | | | | | - W A Harb
- Horizon Oncology Research, Lafayette, USA
| |
Collapse
|
32
|
Leamon CP, Vlahov IR, Reddy JA, Vetzel M, Santhapuram HKR, You F, Bloomfield A, Dorton R, Nelson M, Kleindl P, Vaughn JF, Westrick E. Folate-vinca alkaloid conjugates for cancer therapy: a structure-activity relationship. Bioconjug Chem 2014; 25:560-8. [PMID: 24564229 DOI: 10.1021/bc400441s] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Vintafolide is a potent folate-targeted vinca alkaloid small molecule drug conjugate (SMDC) that has shown promising results in multiple clinical oncology studies. Structurally, vintafolide consists of 4 essential modules: (1) folic acid, (2) a hydrophilic peptide spacer, (3) a disulfide-containing, self-immolative linker, and (4) the cytotoxic drug, desacetylvinblastine hydrazide (DAVLBH). Here, we report a structure-activity study evaluating the biological impact of (i) substituting DAVLBH within the vintafolide molecule with other vinca alkaloid analogues such as vincristine, vindesine, vinflunine, or vinorelbine; (ii) substituting the naturally (S)-configured Asp-Arg-Asp-Asp-Cys peptide with alternative hydrophilic spacers of varied composition; and (iii) varying the composition of the linker module. A series of vinca alkaloid-containing SMDCs were synthesized and purified by HPLC and LCMS. The SMDCs were screened in vitro against folate receptor (FR)-positive cells, and anti-tumor activity was tested against well-established subcutaneous FR-positive tumor xenografts. The cytotoxic and anti-tumor activity was directly compared to that produced by vintafolide. Among all the folate vinca alkaloid SMDCs tested, DAVLBH-containing SMDCs were active, while those constructed with vincristine, vindesine, or vinorelbine analogues failed to produce meaningful biological activity. Within the DAVLBH series, having a bioreleasable, self-immolative linker system was found to be critical for activity since multiple analogues constructed with thioether-based linkers all failed to produce meaningful activity both in vitro and in vivo. Substitutions of some or all of the natural amino acids within vintafolide's hydrophilic spacer module did not significantly change the in vitro or in vivo potency of the SMDCs. Vintafolide remains one of the most potent folate-vinca alkaloid SMDCs produced to date, and continued clinical development is warranted.
Collapse
|
33
|
Serpe L, Gallicchio M, Canaparo R, Dosio F. Targeted treatment of folate receptor-positive platinum-resistant ovarian cancer and companion diagnostics, with specific focus on vintafolide and etarfolatide. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:31-42. [PMID: 24516337 PMCID: PMC3917542 DOI: 10.2147/pgpm.s58374] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Among the gynecological malignancies, ovarian cancer is the leading cause of mortality in developed countries. Treatment of ovarian cancer is based on surgery integrated with chemotherapy. Platinum-based drugs (cisplatin and carboplatin) comprise the core of first-line chemotherapy for patients with advanced ovarian cancer. Platinum-resistant ovarian cancer can be treated with cytotoxic chemotherapeutics such as paclitaxel, topotecan, PEGylated liposomal doxorubicin, or gemcitabine, but many patients eventually relapse on treatment. Targeted therapies based on agents specifically directed to overexpressed receptors, or to selected molecular targets, may be the future of clinical treatment. In this regard, overexpression of folate receptor-α on the surface of almost all epithelial ovarian cancers makes this receptor an excellent "tumor-associated antigen". With appropriate use of spacers/linkers, folate-targeted drugs can be distributed within the body, where they preferentially bind to ovarian cancer cells and are released inside their target cells. Here they can exert their desired cytotoxic function. Based on this strategy, 12 years after it was first described, a folate-targeted vinblastine derivative has now reached Phase III clinical trials in ovarian cancer. This review examines the importance of folate targeting, the state of the art of a vinblastine folate-targeted agent (vintafolide) for treating platinum-resistant ovarian cancer, and its diagnostic companion (etarfolatide) as a prognostic agent. Etarfolatide is a valuable noninvasive diagnostic imaging agent with which to select ovarian cancer patient populations that may benefit from this specific targeted therapy.
Collapse
Affiliation(s)
- Loredana Serpe
- Department of Drug Science and Technology, University of Turin, Italy
| | | | - Roberto Canaparo
- Department of Drug Science and Technology, University of Turin, Italy
| | - Franco Dosio
- Department of Drug Science and Technology, University of Turin, Italy
| |
Collapse
|
34
|
Reddy JA, Dorton R, Bloomfield A, Nelson M, Vetzel M, Guan J, Leamon CP. Rational combination therapy of vintafolide (EC145) with commonly used chemotherapeutic drugs. Clin Cancer Res 2014; 20:2104-14. [PMID: 24429878 DOI: 10.1158/1078-0432.ccr-13-2423] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE When evaluated in patients with ovarian and other cancer, vintafolide (EC145), a potent folate-targeted vinca alkaloid conjugate, displayed a toxicity profile that seemed to be nonoverlapping with many standard-of-care cancer therapeutics. It was, therefore, hypothesized that combining vintafolide with certain approved anticancer drugs may afford greater therapeutic efficacy compared with single-agent therapy. To explore this concept, vintafolide was evaluated in combination with pegylated liposomal doxorubicin (PLD; DOXIL), cisplatin, carboplatin, paclitaxel, docetaxel, topotecan, and irinotecan against folate receptor (FR)-positive models. EXPERIMENTAL DESIGN FR-expressing KB, M109, IGROV, and L1210 cells were first exposed to graded concentrations of vintafolide, either alone or in combination with doxorubicin (active ingredient in PLD), and isobologram plots and combination index values generated. The vintafolide combinations were also studied in mice bearing various FR-expressing tumors. RESULTS Vintafolide displayed strong synergistic activity against KB cells when combined with doxorubicin, and no less-than-additive effects resulted when tested against M109, IGROV, and L1210 cells. In contrast, when either desacetylvinblastine hydrazide (DAVLBH; the vinca alkaloid moiety in vintafolide) or vindesine (the vinca alkaloid most structurally similar to DAVLBH) were tested in combination with doxorubicin, less-than-additive antitumor effects were observed. In vivo, all vintafolide drug combinations produced far greater antitumor effect (complete responses and cures) compared with the single agents alone, without significant increase in overall toxicity. Importantly, these benefits were not observed with combinations of PLD and DAVLBH or vindesine. CONCLUSIONS On the basis of these encouraging preclinical results, clinical studies to evaluate vintafolide drug combination therapies are now under way.
Collapse
Affiliation(s)
- Joseph A Reddy
- Authors' Affiliation: Endocyte Inc., West Lafayette, Indiana
| | | | | | | | | | | | | |
Collapse
|
35
|
Reber J, Haller S, Leamon CP, Müller C. 177Lu-EC0800 combined with the antifolate pemetrexed: preclinical pilot study of folate receptor targeted radionuclide tumor therapy. Mol Cancer Ther 2013; 12:2436-45. [PMID: 24030631 DOI: 10.1158/1535-7163.mct-13-0422-t] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
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 (177)Lu-EC0800. Determination of the combination index (CI) revealed a synergistic inhibitory effect of (177)Lu-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 (177)Lu-EC0800 (20 MBq) and a subtherapeutic (0.4 mg) or therapeutic amount (1.6 mg) of PMX. Application of (177)Lu-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. PMXsubther protected the kidneys from undesired side effects of (177)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 (99m)Tc-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.
Collapse
Affiliation(s)
- Josefine Reber
- Corresponding Author: Cristina Müller, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.
| | | | | | | |
Collapse
|
36
|
Kelderhouse LE, Chelvam V, Wayua C, Mahalingam S, Poh S, Kularatne SA, Low PS. Development of tumor-targeted near infrared probes for fluorescence guided surgery. Bioconjug Chem 2013; 24:1075-80. [PMID: 23642154 DOI: 10.1021/bc400131a] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Complete surgical resection of malignant disease is the only reliable method to cure cancer. Unfortunately, quantitative tumor resection is often limited by a surgeon's ability to locate all malignant disease and distinguish it from healthy tissue. Fluorescence-guided surgery has emerged as a tool to aid surgeons in the identification and removal of malignant lesions. While nontargeted fluorescent dyes have been shown to passively accumulate in some tumors, the resulting tumor-to-background ratios are often poor, and the boundaries between malignant and healthy tissues can be difficult to define. To circumvent these problems, our laboratory has developed high affinity tumor targeting ligands that bind to receptors that are overexpressed on cancer cells and deliver attached molecules selectively into these cells. In this study, we explore the use of two tumor-specific targeting ligands (i.e., folic acid that targets the folate receptor (FR) and DUPA that targets prostate specific membrane antigen (PSMA)) to deliver near-infrared (NIR) fluorescent dyes specifically to FR and PSMA expressing cancers, thereby rendering only the malignant cells highly fluorescent. We report here that all FR- and PSMA-targeted NIR probes examined bind cultured cancer cells in the low nanomolar range. Moreover, upon intravenous injection into tumor-bearing mice with metastatic disease, these same ligand-NIR dye conjugates render receptor-expressing tumor tissues fluorescent, enabling their facile resection with minimal contamination from healthy tissues.
Collapse
Affiliation(s)
- Lindsay E Kelderhouse
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | | | | | | | | | | | | |
Collapse
|
37
|
Du C, Deng D, Shan L, Wan S, Cao J, Tian J, Achilefu S, Gu Y. A pH-sensitive doxorubicin prodrug based on folate-conjugated BSA for tumor-targeted drug delivery. Biomaterials 2013; 34:3087-97. [DOI: 10.1016/j.biomaterials.2013.01.041] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/05/2013] [Indexed: 10/27/2022]
|
38
|
Okamatsu A, Motoyama K, Onodera R, Higashi T, Koshigoe T, Shimada Y, Hattori K, Takeuchi T, Arima H. Folate-Appended β-Cyclodextrin as a Promising Tumor Targeting Carrier for Antitumor Drugs in Vitro and in Vivo. Bioconjug Chem 2013; 24:724-33. [DOI: 10.1021/bc400015r] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ayaka Okamatsu
- Graduate School of Pharmaceutical
Sciences, Kumamoto University, 5-1 Oe-honmachi,
Chuo-ku, Kumamoto 862-0973, Japan
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical
Sciences, Kumamoto University, 5-1 Oe-honmachi,
Chuo-ku, Kumamoto 862-0973, Japan
| | - Risako Onodera
- Graduate School of Pharmaceutical
Sciences, Kumamoto University, 5-1 Oe-honmachi,
Chuo-ku, Kumamoto 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical
Sciences, Kumamoto University, 5-1 Oe-honmachi,
Chuo-ku, Kumamoto 862-0973, Japan
| | - Takahiro Koshigoe
- Faculty of Engineering, Tokyo Polytechnic University, 1583 Iiyama, Atsugi 243-0297,
Japan
| | - Yasutaka Shimada
- R&D Lab, NanoDex Inc., 705-1 Shimoimaizumi, Ebina 243-0435, Japan
| | - Kenjiro Hattori
- Faculty of Engineering, Tokyo Polytechnic University, 1583 Iiyama, Atsugi 243-0297,
Japan
- R&D Lab, NanoDex Inc., 705-1 Shimoimaizumi, Ebina 243-0435, Japan
| | - Tomoko Takeuchi
- Faculty of Engineering, Tokyo Polytechnic University, 1583 Iiyama, Atsugi 243-0297,
Japan
| | - Hidetoshi Arima
- Graduate School of Pharmaceutical
Sciences, Kumamoto University, 5-1 Oe-honmachi,
Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|
39
|
Zhao F, Yin H, Zhang Z, Li J. Folic acid modified cationic γ-cyclodextrin-oligoethylenimine star polymer with bioreducible disulfide linker for efficient targeted gene delivery. Biomacromolecules 2013; 14:476-84. [PMID: 23323627 DOI: 10.1021/bm301718f] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
For an efficient folate-targeted delivery, while the interaction between the folate on the carriers and the folate receptor (FR) on the cells is necessary, the recovering and recycling of FR to maintain a high density level of FR on the cellular membrane is also important. Herein, we demonstrate a design and synthesis of a new star-shaped cationic polymer containing a γ-cyclodextrin (γ-CD) core and multiple oligoethylenimine (OEI) arms with folic acid (FA) linked by a bioreducible disulfide bond for efficient targeted gene delivery. The newly synthesized cationic polymer, named γ-CD-OEI-SS-FA, could be cleaved efficiently, and FA was readily released under reductive condition similar to intracellular environment. The γ-CD-OEI-SS-FA polymer was well-characterized and studied in terms of its gene delivery properties in FR-positive KB cells and FR-negative A549 cells under various conditions, in comparison with cationic polymers such as high molecular weight branched polyethylenimine (PEI), γ-CD-OEI star-shaped cationic polymer, γ-CD-OEI-FA polymer where FA was directed linked to the star polymer without disulfide linker. Our data have demonstrated that the new γ-CD-OEI-SS-FA gene carrier had low cytotoxicity and possessed capacity to target and deliver DNA to specific tumor cells that overexpress FRs, as well as functions to recover and recycle FRs onto cellular membranes to facilitate continuous FR-mediated endocytosis to achieve very high levels of gene expression. This study has expanded the strategy of FA-targeted delivery by combining the smart FR-recycling function to achieve the significant enhancement of gene expression. The new FA-targeted and bioreducible carrier may be a promising efficient gene delivery system for potential cancer gene therapy.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Bioengineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore
| | | | | | | |
Collapse
|
40
|
Krall N, Scheuermann J, Neri D. Small Targeted Cytotoxics: Current State and Promises from DNA-Encoded Chemical Libraries. Angew Chem Int Ed Engl 2013; 52:1384-402. [DOI: 10.1002/anie.201204631] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Indexed: 01/06/2023]
|
41
|
Krall N, Scheuermann J, Neri D. Entwicklung zielgerichteter niedermolekularer zytotoxischer Wirkstoffverbindungen mit DNA-codierten chemischen Bibliotheken. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201204631] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
42
|
Liang X, Sun Y, Zeng W, Liu L, Ma X, Zhao Y, Fan J. Synthesis and biological evaluation of a folate-targeted rhaponticin conjugate. Bioorg Med Chem 2013; 21:178-185. [PMID: 23177726 DOI: 10.1016/j.bmc.2012.10.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 10/11/2012] [Accepted: 10/25/2012] [Indexed: 12/21/2022]
Abstract
To improve the therapeutic effect of rhaponticin (RHA), a folate receptor (FR) targeted RHA conjugate was synthesized by utilizing a hydrophilic peptide spacer linked to folic acid (FA) via a releasable disulfide linker. This water-soluble conjugate was found to retain high affinity for FR-positive cells, and it produced specific, dose-responsive activity in vitro. Treatment of FRHA with a reducing agent indicated that the amino-reactive derivative of RHA would be released spontaneously following disulfide bond reduction within the endosomes. FRHA also proved to be active predominantly specific against FR-positive syngeneic and xenograft models in vivo, and possible curative activity resulted with minimal to moderate toxicity. The FRHA conjugate greatly enhanced the therapeutic effects and reduced the toxicity of RHA. In conclusion, FRHA represents a folate-targeted chemotherapeutic that can produce potent activity against established sc tumors. Hence, this report has a great significance in pharmacology and clinical medicine as well as methodology.
Collapse
Affiliation(s)
- Xuhua Liang
- School of Chemical Engineering, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Kornilova AY, Algayer B, Breslin M, Addona GH, Uebele V. Development of a fluorescence polarization binding assay for folate receptor. Anal Biochem 2013; 432:59-62. [DOI: 10.1016/j.ab.2012.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 09/11/2012] [Accepted: 09/16/2012] [Indexed: 10/27/2022]
|
44
|
Lorusso PM, Edelman MJ, Bever SL, Forman KM, Pilat M, Quinn MF, Li J, Heath EI, Malburg LM, Klein PJ, Leamon CP, Messmann RA, Sausville EA. Phase I study of folate conjugate EC145 (Vintafolide) in patients with refractory solid tumors. J Clin Oncol 2012; 30:4011-6. [PMID: 23032618 DOI: 10.1200/jco.2011.41.4946] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE EC145 (vintafolide), a conjugate of folic acid and the vinca alkaloid desacetylvinblastine hydrazide (DAVLBH), is a ligand for the folate receptor (FR), with activity against FR-positive tumor xenografts in vivo. This phase I study determined the maximum-tolerated dose (MTD) of EC145 administered as a bolus intravenous injection or 1-hour infusion in patients with refractory solid tumors. PATIENTS AND METHODS EC145 was administered as a bolus injection or 1-hour infusion on days 1, 3, and 5 and days 15, 17, and 19 of each 28-day cycle with dose escalation in cohorts of three to six patients until the MTD was identified. Plasma pharmacokinetics were determined on days 1 and 3 of the first cycle. RESULTS The MTD of EC145 was 2.5 mg when administered as either a bolus injection or 1-hour infusion. Constipation was the dose-limiting toxicity with both routes. Constipation, nausea, fatigue, and vomiting were the most commonly reported adverse events. One partial response to therapy was observed in a patient with metastatic ovarian cancer. CONCLUSION EC145 administered by bolus injection or as a 1-hour infusion at a dose of 2.5 mg on days 1, 3, and 5 and days 15, 17, and 19 of a 28-day cycle has an acceptable safety profile in patients with advanced cancer. On the basis of these findings, phase II studies of EC145 have been initiated in patients with advanced epithelial ovarian cancer and non-small-cell lung cancer.
Collapse
Affiliation(s)
- Patricia M Lorusso
- Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene St, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abu Ajaj K, El-Abadla N, Welker P, Azab S, Zeisig R, Fichtner I, Kratz F. Comparative evaluation of the biological properties of reducible and acid-sensitive folate prodrugs of a highly potent doxorubicin derivative. Eur J Cancer 2012; 48:2054-65. [DOI: 10.1016/j.ejca.2011.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/24/2011] [Accepted: 08/15/2011] [Indexed: 11/24/2022]
|
46
|
Vlahov IR, Leamon CP. Engineering folate-drug conjugates to target cancer: from chemistry to clinic. Bioconjug Chem 2012; 23:1357-69. [PMID: 22667324 DOI: 10.1021/bc2005522] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The folate receptor (FR) is a potentially useful biological target for the management of many human cancers. This membrane protein binds extracellular folates with very high affinity and, through an endocytic process, physically delivers them inside the cell for biological consumption. There are now many examples of how this physiological system can be exploited for the targeted delivery of biologically active molecules to cancer. In fact, strong preclinical as well as emerging clinical evidence exists showing how FR-positive cancers can be (i) anatomically identified using folate conjugates of radiodiagnostic imaging agents and (ii) effectively treated with companion folate-targeted chemotherapies. While the biological results are compelling, it is of equal importance to understand the conjugation chemistries that were developed to produce these active molecules. Therefore, this review will focus on the methods utilized to construct folate-based small-molecule drug conjugates (SMDCs), with particular attention focused on modular design, hydrophilic spacers, and self-immolative linkers.
Collapse
|
47
|
|
48
|
Pribble P, Edelman MJ. EC145: a novel targeted agent for adenocarcinoma of the lung. Expert Opin Investig Drugs 2012; 21:755-61. [DOI: 10.1517/13543784.2012.671294] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Pamela Pribble
- University of Maryland School of Medicine, Department of Medicine, Baltimore, MD, USA
| | - Martin J Edelman
- University of Maryland Greenebaum Cancer Center, 22 S. Greene Street, Baltimore, MD 21201, USA ;
| |
Collapse
|
49
|
Yang JJ, Kularatne SA, Chen X, Low PS, Wang E. Characterization of in vivo disulfide-reduction mediated drug release in mouse kidneys. Mol Pharm 2012; 9:310-7. [PMID: 22171616 DOI: 10.1021/mp200483t] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Due to the overexpression of a folate receptor (FR) on many malignant cells, folate-targeted drugs have been developed to improve the cancer specificity of chemotherapeutic agents. Therapeutic index is further enhanced with the use of self-immolative linkers that efficiently release the attached drug upon cellular internalization of the folate-drug conjugate. Because FR is also abundant in normal kidney proximal tubule (PT) cells, we sought to examine in real time the trafficking and release of folate-targeted drugs in the kidney in vivo. Thus, we conducted two-photon kidney imaging studies in mice utilizing a Förster resonance energy transfer (FRET) based folate conjugate that undergoes a color shift from red to green upon reduction of the disulfide bond linking folate to a surrogate drug molecule. Following infusion via intravenous injection, folate-FRET reached the kidney in its intact unreduced form. The folate-FRET conjugate was then filtered into the lumen of PT, where it was efficiently captured by FR. As FR transcytosed across PT, some disulfide reduction occurred, with reduced folate-FRET detectable in PT vesicles 30 min postinjection. Prolonged monitoring of folate-FRET in mice showed modest progression of reduction in PT cells over time. Moreover, inhibition of FR trafficking in PT cells by colchicine did not significantly affect the rate or extent of folate-FRET reduction. Finally, the lack of cytosolic accumulation of released drug surrogate in the PT suggests that drug release via disulfide bond reduction should cause little kidney toxicity.
Collapse
Affiliation(s)
- Jun J Yang
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, USA
| | | | | | | | | |
Collapse
|
50
|
Kim SH, de Mas N, Parlanti L, Lyngberg OK, Ströhlein G, Guo Z, Dambalas K, Rosso VW, Yang BS, Girard KP, Manaloto ZA, D’Arasmo G, Frigerio RE, Wang W, Lu X, Bolgar MS, Gokhale M, Thakur AB. Synthesis, Chromatographic Purification, and Isolation of Epothilone–Folic Acid Conjugate BMS-753493. Org Process Res Dev 2011. [DOI: 10.1021/op200023g] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Germano D’Arasmo
- NerPharMa DS, Nerviano Medical Sciences Group, Via Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Riccardo E. Frigerio
- NerPharMa DS, Nerviano Medical Sciences Group, Via Pasteur 10, 20014 Nerviano, Milan, Italy
| | | | | | | | | | | |
Collapse
|