1
|
Akram F, Ali AM, Akhtar MT, Fatima T, Shabbir I, Ul Haq I. The journey of antibody-drug conjugates for revolutionizing cancer therapy: A review. Bioorg Med Chem 2025; 117:118010. [PMID: 39586174 DOI: 10.1016/j.bmc.2024.118010] [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: 09/07/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a powerful class of targeted cancer therapies that harness the specificity of monoclonal antibodies to deliver cytotoxic payloads directly to tumor cells, minimizing off-target effects. This review explores the advancements in ADC technologies, focusing on advancing next-generation ADCs with novel payloads, conjugation strategies, and enhanced pharmacokinetic profiles. In particular, we highlight innovative payloads, including microtubule inhibitors, spliceosome modulators, and RNA polymerase inhibitors, that offer new mechanisms of cytotoxicity beyond traditional apoptosis induction. Additionally, the introduction of sophisticated conjugation techniques, such as site-specific conjugation using engineered cysteines, enzymatic methods, and integration of non-natural amino acids, has greatly improved the homogeneity, efficacy, and safety of ADCs. Furthermore, the review delves into the mechanistic insights into ADC action, detailing the intracellular pathways that facilitate drug release and cell death, and discussing the significance of bioconjugation methods in optimizing drug-antibody ratios (DARs). The establishment of comprehensive databases like ADCdb, which catalog vital pharmacological and biological data for ADCs, is also explored as a critical resource for advancing ADC research and clinical application. Finally, the clinical landscape of ADCs is examined, with a focus on the evolution of FDA-approved ADCs, such as Gemtuzumab Ozogamicin and Trastuzumab Emtansine, as well as emerging candidates in ongoing trials. As ADCs continue to evolve, their potential to revolutionize cancer therapy remains immense, offering new hope for more effective and personalized treatment options. ADCs also offer a significant advancement in targeted cancer therapy by merging the specificity of monoclonal antibodies with cytotoxic potency of chemotherapeutic agents. Hence, this dual mechanism intensifies tumor selectivity while minimizing systemic toxicity, paving the way for more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Fatima Akram
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Department of Biology, Saint Louis University, St. Louis, MO, USA.
| | - Amna Murrawat Ali
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Muhammad Tayyab Akhtar
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Taseer Fatima
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ifrah Shabbir
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ikram Ul Haq
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Pakistan Academy of Sciences, Islamabad, Pakistan
| |
Collapse
|
2
|
Liolios C, Bouziotis D, Sihver W, Schäfer M, Lambrinidis G, Salvanou EA, Bauder-Wüst U, Benesova M, Kopka K, Kolocouris A, Bouziotis P. Synthesis and Preclinical Evaluation of a Bispecific PSMA-617/RM2 Heterodimer Targeting Prostate Cancer. ACS Med Chem Lett 2024; 15:1970-1978. [PMID: 39563828 PMCID: PMC11571012 DOI: 10.1021/acsmedchemlett.4c00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRPR) have been used for diagnostic molecular imaging/therapy of prostate cancer (PCa). To address tumor heterogeneity, we synthesized and evaluated a bispecific PSMA/GRPR ligand (3) combining PSMA-617 (1) and the GRPR antagonist RM2 (2) with the radiometal chelator DOTA. 3 was radiolabeled with 68Ga ([68Ga]Ga-3) and 177Lu ([177Lu]Lu-3). [68Ga]Ga-3 was tested in the following PCa cell lines for receptor affinity, time kinetic cell-binding/specificity, and cell-internalization: PC-3 and LNCaP. Compared to the monomers (1 and 2), ligand 3 showed specific cell binding, similar receptor affinities, and higher lipophilicity, while its internalization rates and cell-binding were superior. Docking calculations showed that 3 can have binding interactions of PSMA-617 (1) inside the PSMA receptor funnel and RM2 (2) inside the GRPR. In vivo biodistribution studies for [68Ga]Ga-3 showed dual targeting for PSMA(+) and GRPR(+) tumors and higher tumor uptake, faster pharmacokinetic, and lower kidney uptake compared to 1 and 2.
Collapse
Affiliation(s)
- Christos Liolios
- Division of Radiopharmaceutical Chemistry, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Radiochemical Studies Laboratory, INRASTES, N.C.S.R. "Demokritos", Agia Paraskevi Attikis, 15310 Athens, Greece
- Institute of Pharmaceutical Research & Technology (IFET), 18th km of Marathonos Avenue, 15351 Pallini, Attica, Greece
- Department of Nursing & Department of Physiotherapy, School of Health and Caring Sciences, University of West Attica, Agiou Spyridonos, 12243, Egaleo, Greece
| | - Danai Bouziotis
- Radiochemical Studies Laboratory, INRASTES, N.C.S.R. "Demokritos", Agia Paraskevi Attikis, 15310 Athens, Greece
| | - Wiebke Sihver
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - George Lambrinidis
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis-Zografou, 15771 Athens, Greece
| | | | - Ulrike Bauder-Wüst
- Division of Radiopharmaceutical Chemistry, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Martina Benesova
- Division of Radiopharmaceutical Chemistry, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328 Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technical University Dresden, Raum 413 Bergstr. 66, 01069 Dresden, Germany
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Penelope Bouziotis
- Radiochemical Studies Laboratory, INRASTES, N.C.S.R. "Demokritos", Agia Paraskevi Attikis, 15310 Athens, Greece
| |
Collapse
|
3
|
Mirzaei Y, Hussein Mer A, Fattah Maran B, Omidvar L, Misamogooe F, Amirkhani Z, Javaheri Haghighi N, Bagheri N, Keshtkaran Z, Rezaei B, Bargrizaneh F, Jahandideh S, Barpour N, Shahsavarani H, Bazyari A, Abdollahpour-Alitappeh M. Clinical and preclinical advances in PSMA-Directed Antibody-Drug conjugates (ADCs): Current status and hope for the future. Bioorg Chem 2024; 153:107803. [PMID: 39270526 DOI: 10.1016/j.bioorg.2024.107803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Prostate-specific membrane antigen (PSMA) is a type II membrane glycoprotein overexpressed in a variety of tumors, especially in nearly all prostate cancers, which makes it a potentially attractive antigen for targeted cancer therapies. More importantly, PSMA, due to no shedding into circulation and efficient internalization after antibody binding, becomes a potential target for antibody-drug conjugates (ADCs), a valid and emerging paradigm of cancer treatment. Four and eight PSMA-directed ADCs have been or are currently being investigated in clinical trials (three of which failed to confirm the promising results while one is currently being evaluated in an ongoing clinical study) and preclinical studies, respectively, for the treatment of PSMA-positive solid tumors, especially prostate cancer. The present study aims to completely review clinical- and preclinical-stage PSMA-directed ADCs.
Collapse
Affiliation(s)
- Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Ali Hussein Mer
- Department of Nursing, Mergasour Technical Institute, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Bahia Fattah Maran
- Department of business administration, Soran Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Leila Omidvar
- Department of Internal Medicine, School of Medicine, Clinical Research Development Unit (CRDU), Valiasr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Misamogooe
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Zahra Amirkhani
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | | | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord 8813733450, Iran
| | - Zahra Keshtkaran
- Community Based Psychiatric Care Research Center, Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behzad Rezaei
- Laparoscopy Research Center, Department of Surgery, School of Medicine, Shiraz University of Medical Sciences, Fars province, Iran
| | - Farshad Bargrizaneh
- Student Research Committee, School of Health Management and Information Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Jahandideh
- Department of Research and Development, Orchidgene co, Tehran 1387837584, Iran
| | - Nesa Barpour
- Department of Genetics, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983963113, Iran; Iranian Biological Resource Center, Academic Center for Education, Culture and Research (ACECR), Tehran 1533734716, Iran
| | - Ahmadreza Bazyari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
4
|
Yoon H, Savoy EA, Mesbahi N, Hendricksen AT, March GL, Fulton MD, Backer BS, Berkman CE. A PSMA-targeted doxorubicin small-molecule drug conjugate. Bioorg Med Chem Lett 2024; 104:129712. [PMID: 38521177 PMCID: PMC11062396 DOI: 10.1016/j.bmcl.2024.129712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
We developed a model small-molecule drug conjugate (SMDC) that employed doxorubicin as a representative chemotherapeutic targeted to the cell membrane biomarker PSMA (prostate-specific membrane antigen) expressed on prostate cancer cells. The strategy capitalized on the clatherin-mediated internalization of PSMA to facilitate the selective uptake and release of doxorubicin in the target cells. The SMDC was prepared and assessed for binding kinetics, plasma stability, cell toxicity, and specificity towards PSMA expressing prostate cancer cell lines. We observed high affinity of the SMDC for PSMA (IC50 5 nM) with irreversible binding, as well as specific effectiveness against PSMA(+) cells. These findings validated the strategy for a small molecule-based approach in targeted cancer therapy.
Collapse
Affiliation(s)
- Hosog Yoon
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Emily A Savoy
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Nooshin Mesbahi
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Aaron T Hendricksen
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Gabrielle L March
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Melody D Fulton
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Brian S Backer
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Clifford E Berkman
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States.
| |
Collapse
|
5
|
Savoy EA, Olatunji FP, Mesbahi N, Ballard RK, Lovingier CL, Hendricksen AT, Fulton MD, Berkman CE. PSMA-targeted SMART molecules outfitted with SN38. Bioorg Med Chem Lett 2024; 101:129657. [PMID: 38360419 DOI: 10.1016/j.bmcl.2024.129657] [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: 11/30/2023] [Revised: 01/18/2024] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Herein, we report the modular synthesis and evaluation of a prostate-specific membrane antigen (PSMA) targeted small molecule drug conjugate (SMDC) carrying the chemotherapeutic agent, SN38. Due to the fluorogenic properties of SN38, payload release kinetics from the platform was observed in buffers representing the pH conditions of systemic circulation and cellular internalization. It was found that this platform is stable with minimal payload release at physiological pH with most rapid payload release observed at pH values representing the endosome complex. We confirmed selective payload release and chemotherapeutic efficacy for PSMA(+) prostate cancer cells over PSMA(-) cells. These results demonstrate that chemotherapeutic agents with limited solubility can be conjugated to a water-soluble targeting and linker platform without attenuating efficacy.
Collapse
Affiliation(s)
- Emily A Savoy
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Feyisola P Olatunji
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Nooshin Mesbahi
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Ryanne K Ballard
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Christine L Lovingier
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Aaron T Hendricksen
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Melody D Fulton
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Clifford E Berkman
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States.
| |
Collapse
|
6
|
Luo D, Wang X, Ramamurthy G, Walker E, Zhang L, Shirke A, Naidu NG, Burda C, Shakya R, Hostnik E, Joseph M, Ponsky L, Ponomarev V, Rosol TJ, Tweedle MF, Basilion JP. Evaluation of a photodynamic therapy agent using a canine prostate cancer model. Prostate 2023; 83:1176-1185. [PMID: 37211857 PMCID: PMC11135201 DOI: 10.1002/pros.24560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/30/2023] [Accepted: 05/04/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Male dogs can develop spontaneous prostate cancer, which is similar physiologically to human disease. Recently, Tweedle and coworkers have developed an orthotopic canine prostate model allowing implanted tumors and therapeutic agents to be tested in a more translational large animal model. We used the canine model to evaluate prostate-specific membrane antigen (PSMA)-targeted gold nanoparticles as a theranostic approach for fluorescence (FL) imaging and photodynamic therapy (PDT) of early stage prostate cancer. METHODS Dogs (four in total) were immunosuppressed with a cyclosporine-based immunosuppressant regimen and their prostate glands were injected with Ace-1-hPSMA cells using transabdominal ultrasound (US) guidance. Intraprostatic tumors grew in 4-5 weeks and were monitored by ultrasound (US). When tumors reached an appropriate size, dogs were injected intravenously (iv) with PSMA-targeted nano agents (AuNPs-Pc158) and underwent surgery 24 h later to expose the prostate tumors for FL imaging and PDT. Ex vivo FL imaging and histopathological studies were performed to confirm PDT efficacy. RESULTS All dogs had tumor growth in the prostate gland as revealed by US. Twenty-four hours after injection of PSMA-targeted nano agents (AuNPs-Pc158), the tumors were imaged using a Curadel FL imaging device. While normal prostate tissue had minimal fluorescent signal, the prostate tumors had significantly increased FL. PDT was activated by irradiating specific fluorescent tumor areas with laser light (672 nm). PDT bleached the FL signal, while fluorescent signals from the other unexposed tumor tissues were unaffected. Histological analysis of tumors and adjacent prostate revealed that PDT damaged the irradiated areas to a depth of 1-2 mms with the presence of necrosis, hemorrhage, secondary inflammation, and occasional focal thrombosis. The nonirradiated areas showed no visible damages by PDT. CONCLUSION We have successfully established a PSMA-expressing canine orthotopic prostate tumor model and used the model to evaluate the PSMA-targeted nano agents (AuNPs-Pc158) in the application of FL imaging and PDT. It was demonstrated that the nano agents allowed visualization of the cancer cells and enabled their destruction when they were irradiated with a specific wavelength of light.
Collapse
Affiliation(s)
- Dong Luo
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xinning Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Lifang Zhang
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Aditi Shirke
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Naraen G. Naidu
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Clemens Burda
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Reena Shakya
- Target Validation Shared Resource, James Comprehensive Cancer Center, The Ohio State University, Columbus Ohio, USA
| | - Eric Hostnik
- College of Veterinary Medicine- Veterinary Medical Center, The Ohio State University, Columbus, OH, USA
| | - Mathew Joseph
- Interventional Cardiology Cath Core Lab, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Lee Ponsky
- Department of Urology, University Hospitals, Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, USA
| | | | - Thomas J. Rosol
- Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Michael F. Tweedle
- Deptartment of Radiology, The Wright Center for Innovation in Biomolecular Imaging, The Ohio State University, Columbus, OH, USA
| | - James P. Basilion
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
7
|
Olatunji FP, Pun M, Herman JW, Romero O, Maniatopoulos M, Latoche JD, Parise RA, Guo J, Beumer JH, Anderson CJ, Berkman CE. Modular Smart Molecules for PSMA-Targeted Chemotherapy. Mol Cancer Ther 2022; 21:1701-1709. [PMID: 35999662 PMCID: PMC9842478 DOI: 10.1158/1535-7163.mct-22-0160] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/26/2022] [Accepted: 08/10/2022] [Indexed: 01/19/2023]
Abstract
New targeted chemotherapeutics are urgently needed to minimize off-target toxicity and reduce the high-mortality rate associated with metastatic prostate cancer. Herein, we report on the modular synthesis, pharmacokinetics, and efficacy of two small-molecule-drug conjugates (SMDC) targeted to prostate-specific membrane antigen (PSMA) incorporating either: (i) a cathepsin-B-cleavable valine-citrulline (Val-Cit), or (ii) an acid-cleavable phosphoramidate linker. Crucial components used in the design of the conjugates include: (i) CTT1298, a nanomolar affinity ligand that binds irreversibly to PSMA and has proven in past studies to rapidly internalize and shuttle payloads into PSMA-expressing prostate cancer cells, (ii) MMAE, a known potent cytotoxic payload, and (iii) an albumin-binder, proven to improve residence time of drug conjugates. At dose of 0.8 mg/kg (∼250 nmol/kg), the two SMDCs showed significant efficacy in a PSMA(+) PC3-PIP mouse model of human prostate cancer compared with controls, without inducing systemic toxicity. Though localization of the SMDCs was observed in tissues apart from the tumor, release of MMAE was observed predominantly in tumor tissue, at levels that were 2-3 orders of magnitude higher than non-target tissues. Furthermore, SMDC2, which incorporated a novel pH-responsive phosporamidate linker, demonstrated significantly improved efficacy over SMDC1 that has a Val-Cit linker, with a 100% survival over 90 days and 4 out of 8 mice showing complete tumor growth inhibition after 6 weekly doses of 0.8 mg/kg (244 nmol/kg). Our findings demonstrate the potential of irreversible PSMA inhibitors combined with pH-responsive linkers as a way to specifically deliver chemotherapeutic drugs to prostate cancer tumors with minimal toxicity.
Collapse
Affiliation(s)
| | - Michael Pun
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630,Department of Chemistry, University of Missouri, Columbia, MO 65211,Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO 65211
| | - Jacob W. Herman
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630
| | - Oscar Romero
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630
| | | | - Joseph D. Latoche
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213
| | - Robert A. Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213
| | - Jianxia Guo
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213
| | - Jan H. Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, 15261.,Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Carolyn J. Anderson
- Department of Chemistry, University of Missouri, Columbia, MO 65211,Department of Radiology, University of Missouri, Columbia, MO 65211,Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO 65211
| | - Clifford E. Berkman
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630
| |
Collapse
|
8
|
Zhao N, Chopra S, Trepka K, Wang YH, Sakhamuri S, Hooshdaran N, Kim H, Zhuo J, Lim SA, Leung KK, Egusa EA, Zhu J, Zhang L, Foye A, Sriram R, Chan E, Seo Y, Feng FY, Small EJ, Chou J, Wells JA, Aggarwal R, Evans MJ. CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease. Clin Cancer Res 2022; 28:3066-3075. [PMID: 35604681 PMCID: PMC9288514 DOI: 10.1158/1078-0432.ccr-21-3858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/07/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
PURPOSE With the improvement in overall survival with 177Lu-PSMA 617, radioligand therapy (RLT) is now a viable option for patients with metastatic castration-resistant prostate cancer (mCRPC). However, responses are variable, in part due to low PSMA expression in 30% of patients. Herein, we evaluated whether the cell surface protein CUB domain-containing protein 1 (CDCP1) can be exploited to treat mCRPC with RLT, including in PSMA-low subsets. EXPERIMENTAL DESIGN CDCP1 levels were evaluated using RNA sequencing from 119 mCRPC biopsies. CDCP1 levels were assessed in 17 post-enzalutamide- or abiraterone-treated mCRPC biopsies, 12 patient-derived xenografts (PDX), and prostate cancer cell lines. 4A06, a recombinant human antibody that targets the CDCP1 ectodomain, was labeled with Zr-89 or Lu-177 and tested in tumor-bearing mice. RESULTS CDCP1 expression was observed in 90% of mCRPC biopsies, including small-cell neuroendocrine (SCNC) and adenocarcinomas with low FOLH1 (PSMA) levels. Fifteen of 17 evaluable mCRPC biopsies (85%) demonstrated membranous CDCP1 expression, and 4 of 17 (23%) had higher CDCP1 H-scores compared with PSMA. CDCP1 was expressed in 10 of 12 PDX samples. Bmax values of approximately 22,000, 6,200, and 2,800 fmol/mg were calculated for PC3, DU145, and C4-2B human prostate cancer cells, respectively. 89Zr-4A06 PET detected six human prostate cancer xenografts, including PSMA-low tumors. 177Lu-4A06 significantly suppressed growth of DU145 and C4-2B xenografts. CONCLUSIONS The data provide the first evidence supporting CDCP1-directed RLT to treat mCRPC. Expanded studies are warranted to determine whether CDCP1 is a viable drug target for patients with mCPRC.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Shalini Chopra
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Kai Trepka
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - Yung-hua Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Sasank Sakhamuri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Nima Hooshdaran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Hyunjung Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Jie Zhuo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Shion A. Lim
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Kevin K. Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Emily A. Egusa
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Jun Zhu
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Li Zhang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - Adam Foye
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Emily Chan
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Felix Y. Feng
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Eric J. Small
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Jonathan Chou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Rahul Aggarwal
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Michael J. Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
9
|
Stenberg VY, Tornes AJK, Nilsen HR, Revheim ME, Bruland ØS, Larsen RH, Juzeniene A. Factors Influencing the Therapeutic Efficacy of the PSMA Targeting Radioligand 212Pb-NG001. Cancers (Basel) 2022; 14:cancers14112784. [PMID: 35681766 PMCID: PMC9179904 DOI: 10.3390/cancers14112784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a protein overexpressed in metastatic castration-resistant prostate cancer and a promising target for targeted radionuclide therapy. PSMA-targeted alpha therapy is of growing interest due to the high-emission energy and short range of alpha particles, resulting in a prominent cytotoxic potency. This study assesses the influence of various factors on the in vitro and in vivo therapeutic efficacy of the alpha particle generating PSMA-targeting radioligand 212Pb-NG001. Abstract This study aimed to determine the influence of cellular PSMA expression, radioligand binding and internalization, and repeated administrations on the therapeutic effects of the PSMA-targeting radioligand 212Pb-NG001. Cellular binding and internalization, cytotoxicity, biodistribution, and the therapeutic efficacy of 212Pb-NG001 were investigated in two human prostate cancer cell lines with different PSMA levels: C4-2 (PSMA+) and PC-3 PIP (PSMA+++). Despite 10-fold higher PSMA expression on PC-3 PIP cells, cytotoxicity and therapeutic efficacy of the radioligand was only 1.8-fold better than for the C4-2 model, possibly explained by lower cellular internalization and less blood-rich stroma in PC-3 PIP xenografts. Mice bearing subcutaneous PC-3 PIP xenografts were treated with 0.2, 0.4, and 0.8 MBq of 212Pb-NG001 that resulted in therapeutic indexes of 2.7, 3.0, and 3.5, respectively. A significant increase in treatment response was observed in mice that received repeated injections compared to the corresponding single dose (therapeutic indexes of 3.6 for 2 × 0.2 MBq and 4.4 for 2 × 0.4 MBq). The results indicate that 212Pb-NG001 can induce therapeutic effects at clinically transferrable doses, both in the C4-2 model that resembles solid tumors and micrometastases with natural PSMA expression and in the PC-3 PIP model that mimics poorly vascularized metastases.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
- Nucligen AS, 0379 Oslo, Norway;
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Correspondence: ; Tel.: +47-9012-8434
| | - Anna Julie Kjøl Tornes
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
- Nucligen AS, 0379 Oslo, Norway;
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
| | - Hogne Røed Nilsen
- Department of Pathology, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway;
| | - Mona-Elisabeth Revheim
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| | - Øyvind Sverre Bruland
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | | | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
| |
Collapse
|
10
|
PSMA-Targeting Imaging and Theranostic Agents-Current Status and Future Perspective. Int J Mol Sci 2022; 23:ijms23031158. [PMID: 35163083 PMCID: PMC8835702 DOI: 10.3390/ijms23031158] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
In the past two decades, extensive efforts have been made to develop agents targeting prostate-specific membrane antigen (PSMA) for prostate cancer imaging and therapy. To date, represented by two recent approvals of [68Ga]Ga-PSMA-11 and [18F]F-DCFPyL by the United States Food and Drug Administration (US-FDA) for positron emission tomography (PET) imaging to identify suspected metastases or recurrence in patients with prostate cancer, PSMA-targeting imaging and theranostic agents derived from small molecule PSMA inhibitors have advanced to clinical practice and trials of prostate cancer. The focus of current development of new PSMA-targeting agents has thus shifted to the improvement of in vivo pharmacokinetics and higher specific binding affinity with the aims to further increase the detection sensitivity and specificity and minimize the toxicity to non-target tissues, particularly the kidneys. The main strategies involve systematic chemical modifications of the linkage between the targeting moiety and imaging/therapy payloads. In addition to a summary of the development history of PSMA-targeting agents, this review provides an overview of current advances and future promise of PSMA-targeted imaging and theranostics with focuses on the structural determinants of the chemical modification towards the next generation of PSMA-targeting agents.
Collapse
|
11
|
Jin Y, Schladetsch MA, Huang X, Balunas MJ, Wiemer AJ. Stepping forward in antibody-drug conjugate development. Pharmacol Ther 2022; 229:107917. [PMID: 34171334 PMCID: PMC8702582 DOI: 10.1016/j.pharmthera.2021.107917] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/03/2023]
Abstract
Antibody-drug conjugates (ADCs) are cancer therapeutic agents comprised of an antibody, a linker and a small-molecule payload. ADCs use the specificity of the antibody to target the toxic payload to tumor cells. After intravenous administration, ADCs enter circulation, distribute to tumor tissues and bind to the tumor surface antigen. The antigen then undergoes endocytosis to internalize the ADC into tumor cells, where it is transported to lysosomes to release the payload. The released toxic payloads can induce apoptosis through DNA damage or microtubule inhibition and can kill surrounding cancer cells through the bystander effect. The first ADC drug was approved by the United States Food and Drug Administration (FDA) in 2000, but the following decade saw no new approved ADC drugs. From 2011 to 2018, four ADC drugs were approved, while in 2019 and 2020 five more ADCs entered the market. This demonstrates an increasing trend for the clinical development of ADCs. This review summarizes the recent clinical research, with a specific focus on how the in vivo processing of ADCs influences their design. We aim to provide comprehensive information about current ADCs to facilitate future development.
Collapse
Affiliation(s)
- Yiming Jin
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Megan A Schladetsch
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Xueting Huang
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Marcy J Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Andrew J Wiemer
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
12
|
Ojha R, Chen IC, Hsieh CM, Nepali K, Lai RW, Hsu KC, Lin TE, Pan SL, Chen MC, Liou JP. Installation of Pargyline, a LSD1 Inhibitor, in the HDAC Inhibitory Template Culminated in the Identification of a Tractable Antiprostate Cancer Agent. J Med Chem 2021; 64:17824-17845. [PMID: 34908406 DOI: 10.1021/acs.jmedchem.1c00966] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pragmatic insertion of pargyline, a LSD1 inhibitor, as a surface recognition part in the HDAC inhibitory pharmacophore was planned in pursuit of furnishing potent antiprostate cancer agents. Resultantly, compound 14 elicited magnificent cell growth inhibitory effects against the PC-3 and DU-145 cell lines and led to remarkable suppression of tumor growth in human prostate PC-3 and DU-145 xenograft nude mouse models. The outcome of the enzymatic assays ascertained that the substantial antiproliferative effects of compound 14 were mediated through HDAC6 isoform inhibition as well as selective MAO-A and LSD1 inhibition. Moreover, the signatory feature of LSD1 inhibition by 14 in the context of H3K4ME2 accumulation was clearly evident from the results of western blot analysis. Gratifyingly, hydroxamic acid 14 demonstrates good human hepatocytic stability and good oral bioavailability in rats and exhibits enough promise to emerge as a therapeutic for the treatment of prostate cancer in the near future.
Collapse
Affiliation(s)
- Ritu Ojha
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - I-Chung Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Chien-Ming Hsieh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan
| | - Row-Wen Lai
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan.,Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Tony Eight Lin
- Master Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110031, Taiwan
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan.,Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mei-Chuan Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan.,Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan.,Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110031, Taiwan.,Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan.,TMU Research Center of Drug Discovery, Taipei Medical University, Taipei 110031, Taiwan
| |
Collapse
|
13
|
Machulkin AE, Uspenskaya AA, Zyk NY, Nimenko EA, Ber AP, Petrov SA, Shafikov RR, Skvortsov DA, Smirnova GB, Borisova YA, Pokrovsky VS, Kolmogorov VS, Vaneev AN, Ivanenkov YA, Khudyakov AD, Kovalev SV, Erofeev AS, Gorelkin PV, Beloglazkina EK, Zyk NV, Khazanova ES, Majouga AG. PSMA-targeted small-molecule docetaxel conjugate: Synthesis and preclinical evaluation. Eur J Med Chem 2021; 227:113936. [PMID: 34717125 DOI: 10.1016/j.ejmech.2021.113936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
Prostate cancer is one of the most commonly diagnosed men's cancers and remains one of the leading causes of cancer death. The development of approaches to the treatment of this oncological disease is an ongoing process. In this work, we have carried out the selection of ligands for the creation of conjugates based on the drug docetaxel and synthesized a series of three docetaxel conjugates. In vitro cytotoxicity of these molecules was evaluated using the MTT assay. Based on the assay results, we selected the conjugate which showed cytotoxic potential close to unmodified docetaxel. At the same time, the molar solubility of the resulting compound increased up to 20 times in comparison with the drug itself. In vivo evaluation on 22Rv1 (PSMA+) xenograft model demonstrated a good potency of the synthesized conjugate to inhibit tumor growth: the inhibition turned out to be more than 80% at a dose of 30 mg/kg. Pharmacokinetic parameters of conjugate distribution were analyzed. Also, it was found that PSMA-targeted docetaxel conjugate is less toxic than docetaxel itself, the decrease of molar acute toxicity in comparison with free docetaxel was up to 20%. Obtained conjugate PSMA-DOC is a good candidate for further expanded preclinical trials because of high antitumor activity, fewer side toxic effects and better solubility.
Collapse
Affiliation(s)
- Aleksei E Machulkin
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation.
| | - Anastasia A Uspenskaya
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay Y Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Ekaterina A Nimenko
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Anton P Ber
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Stanislav A Petrov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Radik R Shafikov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, Moscow, 117997, Russian Federation
| | - Dmitry A Skvortsov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Faculty of Biology and Biotechnologies, Higher School of Economics, Myasnitskaya 13, Moscow, 101000, Russia
| | - Galina B Smirnova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Yulia A Borisova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Vadim S Pokrovsky
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia; RUDN University, Miklukho-Maklaya Str.6, Moscow, 117198, Russian Federation
| | - Vasilii S Kolmogorov
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Alexander N Vaneev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Yan A Ivanenkov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region, 141700, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russia; Institute of Biochemistry and Genetics Ufa Science Centre Russian Academy of Sciences (IBG RAS), Oktyabrya Prospekt 71, Ufa, 450054, Russian Federation
| | - Alexander D Khudyakov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Sergei V Kovalev
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Alexander S Erofeev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Petr V Gorelkin
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Elena K Beloglazkina
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay V Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Elena S Khazanova
- LLC Izvarino-Pharma, V. Vnukovskoe, Vnukovskoe Sh., 5th Km., Building 1, Moscow, 108817, Russian Federation
| | - Alexander G Majouga
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, Moscow, 125047, Russian Federation
| |
Collapse
|
14
|
Al-Mansoori L, Elsinga P, Goda SK. Bio-vehicles of cytotoxic drugs for delivery to tumor specific targets for cancer precision therapy. Biomed Pharmacother 2021; 144:112260. [PMID: 34607105 DOI: 10.1016/j.biopha.2021.112260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/09/2023] Open
Abstract
Abnormal structural and molecular changes in malignant tissues were thoroughly investigated and utilized to target tumor cells, hence rescuing normal healthy tissues and lowering the unwanted side effects as non-specific cytotoxicity. Various ligands for cancer cell specific markers have been uncovered and inspected for directional delivery of the anti-cancer drug to the tumor site, in addition to diagnostic applications. Over the past few decades research related to the ligand targeted therapy (LTT) increased tremendously aiming to treat various pathologies, mainly cancers with well exclusive markers. Malignant tumors are known to induce elevated levels of a variety of proteins and peptides known as cancer "markers" as certain antigens (e.g., Prostate specific membrane antigen "PSMA", carcinoembryonic antigen "CEA"), receptors (folate receptor, somatostatin receptor), integrins (Integrin αvβ3) and cluster of differentiation molecules (CD13). The choice of an appropriate marker to be targeted and the design of effective ligand-drug conjugate all has to be carefully selected to generate the required therapeutic effect. Moreover, since some tumors express aberrantly high levels of more than one marker, some approaches investigated targeting cancer cells with more than one ligand (dual or multi targeting). We aim in this review to report an update on the cancer-specific receptors and the vehicles to deliver cytotoxic drugs, including recent advancements on nano delivery systems and their implementation in targeted cancer therapy. We will discuss the advantages and limitations facing this approach and possible solutions to mitigate these obstacles. To achieve the said aim a literature search in electronic data bases (PubMed and others) using keywords "Cancer specific receptors, cancer specific antibody, tumor specific peptide carriers, cancer overexpressed proteins, gold nanotechnology and gold nanoparticles in cancer treatment" was carried out.
Collapse
Affiliation(s)
- Layla Al-Mansoori
- Qatar University, Biomedical Research Centre, Qatar University, Doha 2713, Qatar.
| | - Philip Elsinga
- University of Groningen, University Medical Center Groningen (UMCG), Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sayed K Goda
- Cairo University, Faculty of Science, Giza, Egypt; University of Derby, College of Science and Engineering, Derby, UK.
| |
Collapse
|
15
|
Stenberg VY, Larsen RH, Ma LW, Peng Q, Juzenas P, Bruland ØS, Juzeniene A. Evaluation of the PSMA-Binding Ligand 212Pb-NG001 in Multicellular Tumour Spheroid and Mouse Models of Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094815. [PMID: 34062920 PMCID: PMC8124365 DOI: 10.3390/ijms22094815] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/12/2023] Open
Abstract
Radioligand therapy targeting the prostate-specific membrane antigen (PSMA) is rapidly evolving as a promising treatment for metastatic castration-resistant prostate cancer. The PSMA-targeting ligand p-SCN-Bn-TCMC-PSMA (NG001) labelled with 212Pb efficiently targets PSMA-positive cells in vitro and in vivo. The aim of this preclinical study was to evaluate the therapeutic potential of 212Pb-NG001 in multicellular tumour spheroid and mouse models of prostate cancer. The cytotoxic effect of 212Pb-NG001 was tested in human prostate C4-2 spheroids. Biodistribution at various time points and therapeutic effects of different activities of the radioligand were investigated in male athymic nude mice bearing C4-2 tumours, while long-term toxicity was studied in immunocompetent BALB/c mice. The radioligand induced a selective cytotoxic effect in spheroids at activity concentrations of 3–10 kBq/mL. In mice, the radioligand accumulated rapidly in tumours and was retained over 24 h, while it rapidly cleared from nontargeted tissues. Treatment with 0.25, 0.30 or 0.40 MBq of 212Pb-NG001 significantly inhibited tumour growth and improved median survival with therapeutic indexes of 1.5, 2.3 and 2.7, respectively. In BALB/c mice, no signs of long-term radiation toxicity were observed at activities of 0.05 and 0.33 MBq. The obtained results warrant clinical studies to evaluate the biodistribution, therapeutic efficacy and toxicity of 212Pb-NG001.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
- Department of Research and Development, Nucligen AS, 0379 Oslo, Norway;
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Correspondence: ; Tel.: +47-9012-8434
| | | | - Li-Wei Ma
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| | - Qian Peng
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Petras Juzenas
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| |
Collapse
|
16
|
Boinapally S, Ahn HH, Cheng B, Brummet M, Nam H, Gabrielson KL, Banerjee SR, Minn I, Pomper MG. A prostate-specific membrane antigen (PSMA)-targeted prodrug with a favorable in vivo toxicity profile. Sci Rep 2021; 11:7114. [PMID: 33782486 PMCID: PMC8007718 DOI: 10.1038/s41598-021-86551-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 11/09/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a promising target for the treatment of advanced prostate cancer (PC) and various solid tumors. Although PSMA-targeted radiopharmaceutical therapy (RPT) has enabled significant imaging and prostate-specific antigen (PSA) responses, accumulating clinical data are beginning to reveal certain limitations, including a subgroup of non-responders, relapse, radiation-induced toxicity, and the need for specialized facilities for its administration. To date non-radioactive attempts to leverage PSMA to treat PC with antibodies, nanomedicines or cell-based therapies have met with modest success. We developed a non-radioactive prodrug, SBPD-1, composed of a small-molecule PSMA-targeting moiety, a cancer-selective cleavable linker, and the microtubule inhibitor monomethyl auristatin E (MMAE). SBPD-1 demonstrated high binding affinity to PSMA (Ki = 8.84 nM) and selective cytotoxicity to PSMA-expressing PC cell lines (IC50 = 3.90 nM). SBPD-1 demonstrated a significant survival benefit in two murine models of human PC relative to controls. The highest dose tested did not induce toxicity in immunocompetent mice. The high specific targeting ability of SBPD-1 to PSMA-expressing tumors and its favorable toxicity profile warrant its further development.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Hye-Hyun Ahn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Bei Cheng
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Mary Brummet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Hwanhee Nam
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Kathleen L Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Sangeeta R Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
17
|
Juzeniene A, Stenberg VY, Bruland ØS, Larsen RH. Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:779. [PMID: 33668474 PMCID: PMC7918517 DOI: 10.3390/cancers13040779] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Bone, lymph node, and visceral metastases are frequent in castrate-resistant prostate cancer patients. Since such patients have only a few months' survival benefit from standard therapies, there is an urgent need for new personalized therapies. The prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer and is a molecular target for imaging diagnostics and targeted radionuclide therapy (theragnostics). PSMA-targeted α therapies (PSMA-TAT) may deliver potent and local radiation more selectively to cancer cells than PSMA-targeted β- therapies. In this review, we summarize both the recent preclinical and clinical advances made in the development of PSMA-TAT, as well as the availability of therapeutic α-emitting radionuclides, the development of small molecules and antibodies targeting PSMA. Lastly, we discuss the potentials, limitations, and future perspectives of PSMA-TAT.
Collapse
Affiliation(s)
- Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway;
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway;
- Nucligen, Ullernchausséen 64, 0379 Oslo, Norway;
- Institute for Clinical Medicine, University of Oslo, Box 1171 Blindern, 0318 Oslo, Norway;
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, Box 1171 Blindern, 0318 Oslo, Norway;
- Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | | |
Collapse
|
18
|
Small Molecule-Based Prodrug Targeting Prostate Specific Membrane Antigen for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:cancers13030417. [PMID: 33499427 PMCID: PMC7865627 DOI: 10.3390/cancers13030417] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Metastatic castration-resistant prostate cancer poses a serious clinical problem with poor outcomes and remains a deadly disease. New targeted treatment options are urgently needed. PSMA is highly expressed in prostate cancer and has been an attractive biomarker for the treatment of prostate cancer. In this study, we explored the feasibility of targeted delivery of an antimitotic drug, monomethyl auristatin E (MMAE), to tumor tissue using a small-molecule based PSMA lig-and. With the aid of Cy5.5, we found that a cleavable linker is vital for the antitumor activity of the ligand-drug conjugate and have developed a new PSMA-targeting prodrug, PSMA-1-VcMMAE. In in vitro studies, PSMA-1-VcMMAE was 48-fold more potent in killing PSMA-positive PC3pip cells than killing PSMA-negative PC3flu cells. In in vivo studies, PSMA-1-VcMMAE significantly inhibited tumor growth leading to prolonged animal survival in different animal models, including metastatic prostate cancer models. Compared to anti-PSMA antibody-MMAE conjugate (PSMA-ADC) and MMAE, PSMA-1-VcMMAE had over a 10-fold improved maximum tolerated dose, resulting in improved therapeutic index. The small molecule-drug conjugates reported here can be easily synthesized and are more cost efficient than anti-body-drug conjugates. The therapeutic profile of the PSMA-1-VcMMAE encourages further clin-ical development for the treatment of advanced prostate cancer.
Collapse
|
19
|
Walker E, Turaga SM, Wang X, Gopalakrishnan R, Shukla S, Basilion JP, Lathia JD. Development of near-infrared imaging agents for detection of junction adhesion molecule-A protein. Transl Oncol 2021; 14:101007. [PMID: 33421750 PMCID: PMC7804988 DOI: 10.1016/j.tranon.2020.101007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/29/2022] Open
Abstract
Anti-junctional adhesion molecule-A (JAM-A) monoclonal antibodies (mAb) conjugated with near infra-red fluorescent dye, IR700 – as a JAM-A mAb/IR700 agent was developed. An in vivo JAM-A mAb/IR700-specific near infra-red imaging of human-derived prostate and breast cancer xenograft is presented. A single injection of the agent is diminished number of mitotic cells in cancerous tissue of mice bearing heterotopic tumors. Since, our agent depicts the specific accumulation within the targeted tumors, this agent may be adapted to solid tumor targeted photoimmunotherapy.
Introduction Prostate and breast cancer are the most prevalent primary malignant human tumors globally. Prostatectomy and breast conservative surgery remain the most common definitive treatment option for the >500,000 men and women newly diagnosed with localized prostate and breast cancer each year only in the US. Morphological examination is the mainstay of diagnosis but margin under-sampling of the excised cancer tissue may lead to local recurrence. In despite of the progress of non-invasive optical imaging, there is still a clinical need for targeted optical imaging probes that could rapidly and globally visualize cancerous tissues. Methods Elevated expression of junctional adhesion molecule-A (JAM-A) on tumor cells and its multiple pro-tumorigenic activity make the JAM-A a candidate for molecular imaging. Near-infrared imaging probe, which employed anti-JAM-A monoclonal antibody (mAb) phthalocyanine dye IR700 conjugates (JAM-A mAb/IR700), was synthesized and used to identify and visualize heterotopic human prostate and breast tumor mouse xenografts in vivo. Results The intravenously injected JAM-A mAb/IR700 conjugates enabled the non-invasive detection of prostate and breast cancerous tissue by fluorescence imaging. A single dose of JAM-A mAb/IR700 reduced number of mitotic cancer cells in vivo, indicating theranostic ability of this imaging agent. The JAM-A mAb/IR700 conjugates allowed us to image a specific receptor expression in prostate and breast tumors without post-image processing. Conclusion This agent demonstrates promise as a method to image the extent of prostate and breast cancer in vivo and could assist with real-time visualization of extracapsular extension of cancerous tissue.
Collapse
Affiliation(s)
- E Walker
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| | - S M Turaga
- Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - X Wang
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA
| | - R Gopalakrishnan
- Department of Radiology, Case Center for Imaging Research, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106-7207, USA
| | - S Shukla
- Department of Urology at the University of Florida College of Medicine, Faculty Clinic, 653 West 8th Street, FC12, Jacksonville, FL 32209, USA
| | - J P Basilion
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA; Department of Radiology, Case Center for Imaging Research, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106-7207, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - J D Lathia
- Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
20
|
Huang Y, Del Nagro CJ, Balic K, Mylott WR, Ismaiel OA, Ma E, Faria M, Wheeler AM, Yuan M, Waldron MP, Peay MG, Cortes DF, Roskos L, Liang M, Rosenbaum AI. Multifaceted Bioanalytical Methods for the Comprehensive Pharmacokinetic and Catabolic Assessment of MEDI3726, an Anti-Prostate-Specific Membrane Antigen Pyrrolobenzodiazepine Antibody–Drug Conjugate. Anal Chem 2020; 92:11135-11144. [DOI: 10.1021/acs.analchem.0c01187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | - William R. Mylott
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Omnia A. Ismaiel
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia 44519, Egypt
| | - Eric Ma
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Morse Faria
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Aaron M. Wheeler
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Moucun Yuan
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Michael P. Waldron
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Marlking G. Peay
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | - Diego F. Cortes
- PPD Laboratories 2244 Dabney Road, Richmond, Virginia 23230, United States
| | | | | | | |
Collapse
|
21
|
Winter G, Koch ABF, Löffler J, Lindén M, Solbach C, Abaei A, Li H, Glatting G, Beer AJ, Rasche V. Multi-Modal PET and MR Imaging in the Hen's Egg Test-Chorioallantoic Membrane (HET-CAM) Model for Initial in Vivo Testing of Target-Specific Radioligands. Cancers (Basel) 2020; 12:cancers12051248. [PMID: 32429233 PMCID: PMC7281765 DOI: 10.3390/cancers12051248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/17/2022] Open
Abstract
The validation of novel target-specific radioligands requires animal experiments mostly using mice with xenografts. A pre-selection based on a simpler in vivo model would allow to reduce the number of animal experiments, in accordance with the 3Rs principles (reduction, replacement, refinement). In this respect, the chick embryo or hen’s egg test–chorioallantoic membrane (HET-CAM) model is of special interest, as it is not considered an animal until day 17. Thus, we evaluated the feasibility of quantitative analysis of target-specific radiotracer accumulation in xenografts using the HET-CAM model and combined positron emission tomography (PET) and magnetic resonance imaging (MRI). For proof-of-principle we used established prostate-specific membrane antigen (PSMA)-positive and PSMA-negative prostate cancer xenografts and the clinically widely used PSMA-specific PET-tracer [68Ga]Ga-PSMA-11. Tracer accumulation was quantified by PET and tumor volumes measured with MRI (n = 42). Moreover, gamma-counter analysis of radiotracer accumulation was done ex-vivo. A three- to five-fold higher ligand accumulation in the PSMA-positive tumors compared to the PSMA-negative tumors was demonstrated. This proof-of-principle study shows the general feasibility of the HET-CAM xenograft model for target-specific imaging with PET and MRI. The ultimate value for characterization of novel target-specific radioligands now has to be validated in comparison to mouse xenograft experiments.
Collapse
Affiliation(s)
- Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany; (A.B.F.K.); (J.L.); (C.S.); (A.J.B.)
- Correspondence: (G.W.); (V.R.); Tel.: +49-731-500-61364 (G.W.); +49-731-500-45014 (V.R.)
| | - Andrea B. F. Koch
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany; (A.B.F.K.); (J.L.); (C.S.); (A.J.B.)
| | - Jessica Löffler
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany; (A.B.F.K.); (J.L.); (C.S.); (A.J.B.)
- Core Facility Small Animal Imaging, Ulm University Medical Center, 89081 Ulm, Germany; (A.A.); (H.L.)
| | - Mika Lindén
- Department of Inorganic Chemistry II, Ulm University, 89081 Ulm, Germany;
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany; (A.B.F.K.); (J.L.); (C.S.); (A.J.B.)
| | - Alireza Abaei
- Core Facility Small Animal Imaging, Ulm University Medical Center, 89081 Ulm, Germany; (A.A.); (H.L.)
| | - Hao Li
- Core Facility Small Animal Imaging, Ulm University Medical Center, 89081 Ulm, Germany; (A.A.); (H.L.)
| | - Gerhard Glatting
- Department of Nuclear Medicine, Medical Radiation Physics, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany; (A.B.F.K.); (J.L.); (C.S.); (A.J.B.)
| | - Volker Rasche
- Core Facility Small Animal Imaging, Ulm University Medical Center, 89081 Ulm, Germany; (A.A.); (H.L.)
- Internal Medicine II, Ulm University Medical Center, 89081 Ulm, Germany
- Correspondence: (G.W.); (V.R.); Tel.: +49-731-500-61364 (G.W.); +49-731-500-45014 (V.R.)
| |
Collapse
|
22
|
Czerwińska M, Bilewicz A, Kruszewski M, Wegierek-Ciuk A, Lankoff A. Targeted Radionuclide Therapy of Prostate Cancer-From Basic Research to Clinical Perspectives. Molecules 2020; 25:E1743. [PMID: 32290196 PMCID: PMC7181060 DOI: 10.3390/molecules25071743] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and the second leading cause of cancer-related deaths in Western civilization. Although localized prostate cancer can be treated effectively in different ways, almost all patients progress to the incurable metastatic castration-resistant prostate cancer. Due to the significant mortality and morbidity rate associated with the progression of this disease, there is an urgent need for new and targeted treatments. In this review, we summarize the recent advances in research on identification of prostate tissue-specific antigens for targeted therapy, generation of highly specific and selective molecules targeting these antigens, availability of therapeutic radionuclides for widespread medical applications, and recent achievements in the development of new-generation small-molecule inhibitors and antibody-based strategies for targeted prostate cancer therapy with alpha-, beta-, and Auger electron-emitting radionuclides.
Collapse
Affiliation(s)
- Malwina Czerwińska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
| | - Aleksander Bilewicz
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland;
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Aneta Wegierek-Ciuk
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| | - Anna Lankoff
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| |
Collapse
|
23
|
Kacar S, Unver H, Sahinturk V. A mononuclear copper(II) complex containing benzimidazole and pyridyl ligands: Synthesis, characterization, and antiproliferative activity against human cancer cells. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2019.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
24
|
Loiseau A, Boudon J, Oudot A, Moreau M, Boidot R, Chassagnon R, Mohamed Saïd N, Roux S, Mirjolet C, Millot N. Titanate Nanotubes Engineered with Gold Nanoparticles and Docetaxel to Enhance Radiotherapy on Xenografted Prostate Tumors. Cancers (Basel) 2019; 11:cancers11121962. [PMID: 31817706 PMCID: PMC6966691 DOI: 10.3390/cancers11121962] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022] Open
Abstract
Nanohybrids based on titanate nanotubes (TiONts) were developed to fight prostate cancer by intratumoral (IT) injection, and particular attention was paid to their step-by-step synthesis. TiONts were synthesized by a hydrothermal process. To develop the custom-engineered nanohybrids, the surface of TiONts was coated beforehand with a siloxane (APTES), and coupled with both dithiolated diethylenetriaminepentaacetic acid-modified gold nanoparticles (Au@DTDTPA NPs) and a heterobifunctional polymer (PEG3000) to significantly improve suspension stability and biocompatibility of TiONts for targeted biomedical applications. The pre-functionalized surface of this scaffold had reactive sites to graft therapeutic agents, such as docetaxel (DTX). This novel combination, aimed at retaining the AuNPs inside the tumor via TiONts, was able to enhance the radiation effect. Nanohybrids have been extensively characterized and were detectable by SPECT/CT imaging through grafted Au@DTDTPA NPs, radiolabeled with 111In. In vitro results showed that TiONts-AuNPs-PEG3000-DTX had a substantial cytotoxic activity on human PC-3 prostate adenocarcinoma cells, unlike initial nanohybrids without DTX (Au@DTDTPA NPs and TiONts-AuNPs-PEG3000). Biodistribution studies demonstrated that these novel nanocarriers, consisting of AuNP- and DTX-grafted TiONts, were retained within the tumor for at least 20 days on mice PC-3 xenografted tumors after IT injection, delaying tumor growth upon irradiation.
Collapse
Affiliation(s)
- Alexis Loiseau
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303, CNRS-Université Bourgogne Franche Comté, BP 47870, 21078 Dijon Cedex, France; (A.L.); (R.C.)
| | - Julien Boudon
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303, CNRS-Université Bourgogne Franche Comté, BP 47870, 21078 Dijon Cedex, France; (A.L.); (R.C.)
- Correspondence: (J.B.); (C.M.); (N.M.)
| | - Alexandra Oudot
- Preclinical Imaging Platform, Nuclear Medicine Department, Georges-Francois Leclerc Cancer Center, 21079 Dijon Cedex, France;
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l’Université Bourgogne, UMR 6302, CNRS-Université Bourgogne Franche Comté, 21078 Dijon Cedex, France;
| | - Romain Boidot
- Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center–UNICANCER, 21079 Dijon Cedex, France;
| | - Rémi Chassagnon
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303, CNRS-Université Bourgogne Franche Comté, BP 47870, 21078 Dijon Cedex, France; (A.L.); (R.C.)
| | - Nasser Mohamed Saïd
- Institut UTINAM, UMR 6213, CNRS-Université Bourgogne Franche-Comté, 25030 Besançon Cedex, France; (N.M.S.); (S.R.)
| | - Stéphane Roux
- Institut UTINAM, UMR 6213, CNRS-Université Bourgogne Franche-Comté, 25030 Besançon Cedex, France; (N.M.S.); (S.R.)
| | - Céline Mirjolet
- INSERM LNC UMR 1231, 21078 Dijon Cedex, France
- Radiotherapy Department, Georges-Francois Leclerc Cancer Center, 21079 Dijon Cedex, France
- Correspondence: (J.B.); (C.M.); (N.M.)
| | - Nadine Millot
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303, CNRS-Université Bourgogne Franche Comté, BP 47870, 21078 Dijon Cedex, France; (A.L.); (R.C.)
- Correspondence: (J.B.); (C.M.); (N.M.)
| |
Collapse
|
25
|
Holzapfel M, Mutas M, Chandralingam S, von Salisch C, Peric N, Segelke T, Fischer M, Chakraborty I, Parak WJ, Frangioni JV, Maison W. Nonradioactive Cell Assay for the Evaluation of Modular Prostate-Specific Membrane Antigen Targeting Ligands via Inductively Coupled Plasma Mass Spectrometry. J Med Chem 2019; 62:10912-10918. [PMID: 31714783 DOI: 10.1021/acs.jmedchem.9b01606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The development of novel prostate-specific membrane antigen (PSMA)-targeted radioactive theranostic agents is currently limited to facilities capable of working with high-energy radioisotopes. Even preselection of lead structures in vitro relies mostly on radioactive assays with PSMA(+) LNCaP and PSMA(-) PC-3 cells. Assays utilizing radioisotopes are time consuming, costly, and limit discovery to a small group of scientists with special facilities. Nonradioactive alternatives are therefore needed in the field. In this paper, we describe an inductively coupled plasma mass spectrometry (ICP-MS)-based method for the evaluation of PSMA-targeting ligands conjugated to DOTA-chelates of Europium. This method is based on LNCaP and PC-3 cells and has been validated with the well-established targeting ligand PSMA-617.
Collapse
Affiliation(s)
- Malte Holzapfel
- Department of Chemistry , Universität Hamburg , Bundesstrasse 45 , 20146 Hamburg , Germany
| | | | - Sharah Chandralingam
- Department of Chemistry , Universität Hamburg , Bundesstrasse 45 , 20146 Hamburg , Germany
| | - Carla von Salisch
- Department of Chemistry , Universität Hamburg , Bundesstrasse 45 , 20146 Hamburg , Germany
| | - Natalija Peric
- Department of Chemistry , Universität Hamburg , Bundesstrasse 45 , 20146 Hamburg , Germany
| | | | | | - Indranath Chakraborty
- Center for Hybrid Nanostructure (CHyN) , Universität Hamburg , Luruper Chausee 149 , 22761 Hamburg , Germany
| | - Wolfgang J Parak
- Center for Hybrid Nanostructure (CHyN) , Universität Hamburg , Luruper Chausee 149 , 22761 Hamburg , Germany
| | - John V Frangioni
- Curadel, LLC , 11 Erie Drive , Natick , Massachusetts 01760 , United States
| | - Wolfgang Maison
- Department of Chemistry , Universität Hamburg , Bundesstrasse 45 , 20146 Hamburg , Germany
| |
Collapse
|
26
|
18F-Labeled Cyclized α-Melanocyte-Stimulating Hormone Derivatives for Imaging Human Melanoma Xenograft with Positron Emission Tomography. Sci Rep 2019; 9:13575. [PMID: 31537869 PMCID: PMC6753210 DOI: 10.1038/s41598-019-50014-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022] Open
Abstract
Since metastatic melanoma is deadly, early diagnosis thereof is crucial for managing the disease. We recently developed α-melanocyte-stimulating hormone (αMSH) derivatives, [68Ga]Ga-CCZ01048 and [18F]CCZ01064, that target the melanocortin 1 receptor (MC1R) for mouse melanoma imaging. In this study, we aim to evaluate [18F]CCZ01064 as well as a novel dual-ammoniomethyl-trifluoroborate (AmBF3) derivative, [18F]CCZ01096, for targeting human melanoma xenograft using μPET imaging. The peptides were synthesized on solid phase using Fmoc chemistry. Radiolabeling was achieved in a one-step 18F-19F isotope-exchange reaction. μPET imaging and biodistribution studies were performed in NSG mice bearing SK-MEL-1 melanoma xenografts. The MC1R density on the SK-MEL-1 cell line was determined to be 972 ± 154 receptors/cell (n = 4) via saturation assays. Using [18F]CCZ01064, moderate tumor uptake (3.05 ± 0.47%ID/g) and image contrast were observed at 2 h post-injection. Molar activity was determined to play a key role. CCZ01096 with two AmBF3 motifs showed comparable sub-nanomolar binding affinity to MC1R and much higher molar activity. This resulted in improved tumor uptake (6.46 ± 1.42%ID/g) and image contrast (tumor-to-blood and tumor-to-muscle ratios were 30.6 ± 5.7 and 85.7 ± 11.3, respectively) at 2 h post-injection. [18F]CCZ01096 represents a promising αMSH-based μPET imaging agent for human melanoma and warrants further investigation for potential clinical translation.
Collapse
|
27
|
Wang S, Blaha C, Santos R, Huynh T, Hayes TR, Beckford-Vera DR, Blecha JE, Hong AS, Fogarty M, Hope TA, Raleigh DR, Wilson DM, Evans MJ, VanBrocklin HF, Ozawa T, Flavell RR. Synthesis and Initial Biological Evaluation of Boron-Containing Prostate-Specific Membrane Antigen Ligands for Treatment of Prostate Cancer Using Boron Neutron Capture Therapy. Mol Pharm 2019; 16:3831-3841. [PMID: 31381351 DOI: 10.1021/acs.molpharmaceut.9b00464] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Boron neutron capture therapy (BNCT) is a therapeutic modality which has been used for the treatment of cancers, including brain and head and neck tumors. For effective treatment via BNCT, efficient and selective delivery of a high boron dose to cancer cells is needed. Prostate-specific membrane antigen (PSMA) is a target for prostate cancer imaging and drug delivery. In this study, we conjugated boronic acid or carborane functional groups to a well-established PSMA inhibitor scaffold to deliver boron to prostate cancer cells and prostate tumor xenograft models. Eight boron-containing PSMA inhibitors were synthesized. All of these compounds showed a strong binding affinity to PSMA in a competition radioligand binding assay (IC50 from 555.7 to 20.3 nM). Three selected compounds 1a, 1d, and 1f were administered to mice, and their in vivo blocking of 68Ga-PSMA-11 uptake was demonstrated through a positron emission tomography (PET) imaging and biodistribution experiment. Biodistribution analysis demonstrated boron uptake of 4-7 μg/g in 22Rv1 prostate xenograft tumors and similar tumor/muscle ratios compared to the ratio for the most commonly used BNCT compound, 4-borono-l-phenylalanine (BPA). Taken together, these data suggest a potential role for PSMA targeted BNCT agents in prostate cancer therapy following suitable optimization.
Collapse
Affiliation(s)
- Sinan Wang
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Charles Blaha
- Department of Bioengineering and Therapeutic Sciences , University of California , San Francisco , California , United States
| | - Raquel Santos
- Department of Neurological Surgery , University of California , San Francisco , California , United States
| | - Tony Huynh
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Thomas R Hayes
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Joseph E Blecha
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Andrew S Hong
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Miko Fogarty
- Department of Neurological Surgery , University of California , San Francisco , California , United States
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - David R Raleigh
- Department of Neurological Surgery , University of California , San Francisco , California , United States.,Departments of Radiation Oncology , University of California , San Francisco , California , United States
| | - David M Wilson
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States
| | - Tomoko Ozawa
- Department of Neurological Surgery , University of California , San Francisco , California , United States
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging , University of California , San Francisco , California , United States.,Department of Pharmaceutical Chemistry , University of California , San Francisco , California , United States
| |
Collapse
|
28
|
Liolios C, Sachpekidis C, Schäfer M, Kopka K. Bispecific radioligands targeting prostate-specific membrane antigen and gastrin-releasing peptide receptors on the surface of prostate cancer cells. J Labelled Comp Radiopharm 2019; 62:510-522. [DOI: 10.1002/jlcr.3749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Christos Liolios
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
- German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| |
Collapse
|
29
|
Pal A, Albusairi W, Liu F, Tuhin MTH, Miller M, Liang D, Joo H, Amin TU, Wilson EA, Faridi JS, Park M, Alhamadsheh MM. Hydrophilic Small Molecules That Harness Transthyretin To Enhance the Safety and Efficacy of Targeted Chemotherapeutic Agents. Mol Pharm 2019; 16:3237-3252. [PMID: 31136717 PMCID: PMC6607395 DOI: 10.1021/acs.molpharmaceut.9b00432] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
hydrophobicity of many chemotherapeutic agents usually results
in their nonselective passive distribution into healthy cells and
organs causing collateral toxicity. Ligand-targeted drugs (LTDs) are
a promising class of targeted anticancer agents. The hydrophilicity
of the targeting ligands in LTDs limits its nonselective passive tissue
distribution and toxicity to healthy cells. In addition, the small
size of LTDs allows for better tumor penetration, especially in the
case of solid tumors. However, the short circulation half-life of
LTDs, due to their hydrophilicity and small size, remains a significant
challenge for achieving their full therapeutic potential. Therefore,
extending the circulation half-life of targeted chemotherapeutic agents
while maintaining their hydrophilicity and small size will represent
a significant advance toward effective and safe cancer treatment.
Here, we present a new approach for enhancing the safety and efficacy
of targeted chemotherapeutic agents. By endowing hydrophobic chemotherapeutic
agents with a targeting moiety and a hydrophilic small molecule that
binds reversibly to the serum protein transthyretin, we generated
small hydrophilic drug conjugates that displayed enhanced circulation
half-life in rodents and selectivity to cancer cells. To the best
of our knowledge, this is the first demonstration of a successful
approach that maintains the small size and hydrophilicity of targeted
anticancer agents containing hydrophobic payloads while at the same
time extending their circulation half-life. This was demonstrated
by the superior in vivo efficacy and lower toxicity of our conjugates
in xenograft mouse models of metastatic prostate cancer.
Collapse
|
30
|
Winter G, Vogt A, Jiménez-Franco LD, Rinscheid A, Yousefzadeh-Nowshahr E, Solbach C, Beer AJ, Glatting G, Kletting P. Modelling the internalisation process of prostate cancer cells for PSMA-specific ligands. Nucl Med Biol 2019; 72-73:20-25. [PMID: 31260881 DOI: 10.1016/j.nucmedbio.2019.05.003] [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] [Received: 02/05/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION In prostate-specific membrane antigen (PSMA)-targeting radioligand therapy, small molecules are regularly internalised by the tumour cells. To determine the effectiveness of these ligands, the internalised fraction over time is derived from cell studies. Parameters, such as the ligand concentration and the number of cells, are experiment-specific and therefore a comparison between ligands is difficult. A more objective approach that allows better comparison is desirable. Therefore, the aim of this work was to develop a compartmental model that fully describes all relevant pharmacokinetic interactions of PSMA-specific ligands with prostate cancer cells. METHODS Internalisation studies were performed using the lymph node carcinoma of the prostate cell line LNCaP C4-2 and the prostatic carcinoma cell line PC-3. A new protocol was established for the determination of the PSMA-binding specificity by surface plasmon resonance (SPR). The experimental data in combination with parameters from literature were used for the modelling approach. RESULTS A compartmental model which includes the relevant physiological mechanisms was developed. The basic model structure and some parameters originate from the literature. The PSMA-specific association and dissociation rates of Ga-PSMA-11 were measured using surface plasmon resonance technology. The ligand-induced internalisation and PSMA synthesis rates were estimated by fitting the developed model to experimental data obtained using LNCaP C4-2 cells. For all [68Ga]Ga-PSMA-11 concentrations and including four various incubation times, the ligand-induced internalisation was determined to be (3.6 ± 0.1) % min-1. CONCLUSIONS The presented approach is a prerequisite for better estimation and thus comparison of important ligand-cell interaction parameters, by combining SPR measurements, cell experiments and mathematical modelling. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT A compartmental model was developed for evaluation and comparison of PSMA-binding small molecules. A SPR protocol was established for the determination of PSMA-binding specificity.
Collapse
Affiliation(s)
- Gordon Winter
- Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | - Anja Vogt
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Luis David Jiménez-Franco
- Department of Radiation Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Andreas Rinscheid
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | - Elham Yousefzadeh-Nowshahr
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | | | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| |
Collapse
|
31
|
Wu J, Han D, Shi S, Zhang Q, Zheng G, Wei M, Han Y, Li G, Yang F, Jiao D, Xie P, Zhang L, Yang AG, Zhao A, Qin W, Wen W. A Novel Fully Human Antibody targeting Extracellular Domain of PSMA Inhibits Tumor Growth in Prostate Cancer. Mol Cancer Ther 2019; 18:1289-1301. [PMID: 31048359 DOI: 10.1158/1535-7163.mct-18-1078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/07/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and the second leading cause of cancer-related death. It is of vital importance to develop new strategies for prostate cancer therapy. PSMA (prostate-specific membrane antigen) is specifically expressed in prostate cancer and the neovasculature of certain cancer types, thus is considered to be an ideal target for cancer therapy. In our previous study, we have obtained a PSMA-specific single-chain variable fragment (scFv), named gy1, from a large yeast display naïve human scFv library. In this study, we reconstructed the PSMA scFv into a fully human antibody (named PSMAb) and evaluated its characterization both in vitro and in vivo We showed that PSMAb can specifically bind with and internalize into PSMA+ cells. The binding affinity of PSMAb is measured to be at nanomolar level, and PSMAb has very good thermostability. In vivo study showed that near IR dye-labeled PSMAb can specifically localize at PSMA+ tumors, and the application of PSMAb in vivo significantly inhibited the growth of PSMA+ tumors, but not PSMA- tumors. At the studied doses, no obvious toxicity was observed when applied in vivo, as shown by the relative normal liver and kidney function and normal structure of important organs, shown by hematoxylin and eosin staining. In addition, PSMAb may inhibit tumor growth through antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity mechanisms. Our results indicated that the novel fully human antibody, PSMAb, deserve further study for PSMA-targeted diagnosis and therapy for prostate cancer and other cancer types with vascular PSMA expression.
Collapse
Affiliation(s)
- Jieheng Wu
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shengjia Shi
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qiang Zhang
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Guoxu Zheng
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Ming Wei
- Department of Urology, 150th Central Hospital of PLA, Luoyang, China
| | | | | | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dian Jiao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Pin Xie
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lingling Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | | | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Weihong Wen
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
32
|
Petrylak DP, Kantoff P, Vogelzang NJ, Mega A, Fleming MT, Stephenson JJ, Frank R, Shore ND, Dreicer R, McClay EF, Berry WR, Agarwal M, DiPippo VA, Rotshteyn Y, Stambler N, Olson WC, Morris SA, Israel RJ. Phase 1 study of PSMA ADC, an antibody-drug conjugate targeting prostate-specific membrane antigen, in chemotherapy-refractory prostate cancer. Prostate 2019; 79:604-613. [PMID: 30663074 DOI: 10.1002/pros.23765] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/27/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) is a well-characterized target that is overexpressed selectively on prostate cancer cells. PSMA antibody-drug conjugate (ADC) is a fully human IgG1 monoclonal antibody conjugated to the microtubule disrupting agent monomethyl auristatin E (MMAE), which is designed to specifically bind PSMA-positive cells, internalize, and then release its cytotoxic payload into the cells. PSMA ADC has demonstrated potent and selective antitumor activity in preclinical models of advanced prostate cancer. A Phase 1 study was conducted to assess the safety, pharmacokinetics, and preliminary antitumor effects of PSMA ADC in subjects with treatment-refractory prostate cancer. METHODS In this first-in-man dose-escalation study, PSMA ADC was administered by intravenous infusion every three weeks to subjects with progressive metastatic castration-resistant prostate cancer (mCRPC) who were previously treated with docetaxel chemotherapy. The primary endpoint was to establish a maximum tolerated dose (MTD). The study also examined the pharmacokinetics of the study drug, total antibody, and free MMAE. Antitumor effects were assessed by measuring changes in serum prostate-specific antigen (PSA), circulating tumor cells (CTCs), and radiologic imaging. RESULTS Fifty-two subjects were administered doses ranging from 0.4 to 2.8 mg/kg. Subjects had a median of two prior chemotherapy regimens and prior treatment with abiraterone and/or enzalutamide. Neutropenia and peripheral neuropathy were identified as important first-cycle and late dose-limiting toxicities, respectively. The dose of 2.5 mg/kg was determined to be the MTD. Pharmacokinetics were approximately dose-proportional with minimal drug accumulation. Reductions in PSA and CTCs in subjects treated with doses of ≥1.8 mg/kg were durable and often concurrent. CONCLUSIONS In an extensively pretreated mCRPC population, PSMA ADC demonstrated acceptable toxicity. Antitumor activity was observed over dose ranges up to and including 2.5 mg/kg. The observed anti-tumor activity supported further evaluation of this novel agent for the treatment of advanced metastatic prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, South Carolina
| | - Robert Dreicer
- University of Virginia Cancer Center, Charlottesville, Virginia
| | - Edward F McClay
- California Cancer Associates for Research & Excellence, Encinitas, California
| | | | - Manish Agarwal
- Associates in Oncology and Hematology, Rockville, Maryland
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Targeted therapies hold great promise for cancer treatment and may exhibit even greater efficacy when combined with patient selection tools. The clinical impact of identifying likely responders includes reducing the number of unnecessary and ineffective therapies as well as more accurately determining drug effects. Positron emission tomography (PET) imaging using zirconium-89 radiolabeled monoclonal antibodies (mAbs), also referred to as zirconium-89 (89Zr)-immuno-PET, provides a potential biomarker to measure target expression and verify optimal delivery of targeted agents to tumors. Antibody-drug conjugates (ADCs) combine the high affinity and specificity of mAbs with the potency of cytotoxic drugs to target tumor-expressing antigen and destroy cancer cells. Thus, 89Zr-immuno-PET of whole-body biodistribution, pharmacokinetics, and tumor targeting of antibodies and ADCs to predict toxicity and efficacy could help guide individualized treatment. Here, we review how 89Zr-immuno-PET is being used as a companion diagnostic with the development of ADCs. Furthermore, we discuss how 89Zr-immuno-PET may be utilized in future clinical trials as an adjunct tool with novel ADCs to select cancer patients who have the greatest potential to benefit from treatment and improve ADC dosing regimens.
Collapse
Affiliation(s)
- Kendra S Carmon
- 1 Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ali Azhdarinia
- 1 Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
34
|
Raposo Moreira Dias A, Pina A, Dean A, Lerchen H, Caruso M, Gasparri F, Fraietta I, Troiani S, Arosio D, Belvisi L, Pignataro L, Dal Corso A, Gennari C. Neutrophil Elastase Promotes Linker Cleavage and Paclitaxel Release from an Integrin-Targeted Conjugate. Chemistry 2019; 25:1696-1700. [PMID: 30452790 PMCID: PMC6471013 DOI: 10.1002/chem.201805447] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/16/2018] [Indexed: 12/16/2022]
Abstract
This work takes advantage of one of the hallmarks of cancer, that is, the presence of tumor infiltrating cells of the immune system and leukocyte-secreted enzymes, to promote the activation of an anticancer drug at the tumor site. The peptidomimetic integrin ligand cyclo(DKP-RGD) was found to accumulate on the surface of αv β3 integrin-expressing human renal cell carcinoma 786-O cells. The ligand was conjugated to the anticancer drug paclitaxel through a Asn-Pro-Val (NPV) tripeptide linker, which is a substrate of neutrophil-secreted elastase. In vitro linker cleavage assays and cell antiproliferative experiments demonstrate the efficacy of this tumor-targeting conjugate, opening the way to potential therapeutic applications.
Collapse
Affiliation(s)
| | - Arianna Pina
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Amelia Dean
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | | | - Michele Caruso
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Fabio Gasparri
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Ivan Fraietta
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Sonia Troiani
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Daniela Arosio
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| | - Laura Belvisi
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| | - Luca Pignataro
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Alberto Dal Corso
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Cesare Gennari
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| |
Collapse
|
35
|
Cimadamore A, Cheng M, Santoni M, Lopez-Beltran A, Battelli N, Massari F, Galosi AB, Scarpelli M, Montironi R. New Prostate Cancer Targets for Diagnosis, Imaging, and Therapy: Focus on Prostate-Specific Membrane Antigen. Front Oncol 2018; 8:653. [PMID: 30622933 PMCID: PMC6308151 DOI: 10.3389/fonc.2018.00653] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022] Open
Abstract
The rising incidence rate of the cancer in the prostate gland has increased the demand for improved diagnostic, imaging, and therapeutic approaches. Prostate-specific membrane antigen (PSMA), with folate hydrolase and carboxypeptidase and, internalization activities, is highly expressed in the epithelial cells of the prostate gland and is strongly upregulated in prostatic adenocarcinoma, with elevated expression correlating with, metastasis, progression, and androgen independence. Recently, PSMA has been an active target of investigation by several approaches, including the successful utilization of small molecule inhibitors, RNA aptamer conjugates, PSMA-based immunotherapy, and PSMA-targeted prodrug therapy. Future investigations of PSMA in prostate cancer (PCa) should focus in particular on its intracellular activities and functions. The objective of this contribution is to review the current role of PSMA as a marker for PCa diagnosis, imaging, and therapy.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | - Monica Cheng
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | | | | | | | - Andrea B Galosi
- Institute of Urology, School of Medicine, United Hospitals, Marche Polytechnic University, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
36
|
Kumar A, Kinneer K, Masterson L, Ezeadi E, Howard P, Wu H, Gao C, Dimasi N. Synthesis of a heterotrifunctional linker for the site-specific preparation of antibody-drug conjugates with two distinct warheads. Bioorg Med Chem Lett 2018; 28:3617-3621. [DOI: 10.1016/j.bmcl.2018.10.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 01/08/2023]
|
37
|
Kularatne SA, Thomas M, Myers CH, Gagare P, Kanduluru AK, Crian CJ, Cichocki BN. Evaluation of Novel Prostate-Specific Membrane Antigen-Targeted Near-Infrared Imaging Agent for Fluorescence-Guided Surgery of Prostate Cancer. Clin Cancer Res 2018; 25:177-187. [PMID: 30201762 DOI: 10.1158/1078-0432.ccr-18-0803] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/13/2018] [Accepted: 09/05/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The ability to locate and remove all malignant lesions during radical prostatectomy leads not only to prevent biochemical recurrence (BCR) and possible side effects but also to improve the life expectancy of patients with prostate cancer. Fluorescence-guided surgery (FGS) has emerged as a technique that uses fluorescence to highlight cancerous cells and guide surgeons to resect tumors in real time. Thus, development of tumor-specific near-infrared (NIR) agents that target biomarkers solely expressed on prostate cancer cells will enable to assess negative tumor margins and affected lymph nodes. EXPERIMENTAL DESIGN Because PSMA is overexpressed in prostate cancer cells in >90% of the prostate cancer patient population, a prostate-specific membrane antigen (PSMA)-targeted NIR agent (OTL78) was designed and synthesized. Optical properties, in vitro and in vivo specificity, tumor-to-background ratio (TBR), accomplishment of negative surgical tumor margins using FGS, pharmacokinetics (PKs) properties, and preclinical toxicology of OTL78 were then evaluated in requisite models. RESULTS OTL78 binds to PSMA-expressing cells with high affinity, concentrates selectively to PSMA-positive cancer tissues, and clears rapidly from healthy tissues with a half-time of 17 minutes. It also exhibits an excellent TBR (5:1) as well as safety profile in animals. CONCLUSIONS OTL78 is an excellent tumor-specific NIR agent for use in fluorescence-guided radical prostatectomy and FGS of other cancers.
Collapse
Affiliation(s)
| | - Mini Thomas
- On Target Laboratories, West Lafayette, Indiana
| | | | | | | | - Christa J Crian
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana
| | - Brandy N Cichocki
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana
| |
Collapse
|
38
|
Yue Y, Xu Y, Huang L, Pan D, Bai Z, Wang L, Yang R, Yan J, Song H, Li X, Yang M. Evaluation of A Novel GLP-1R Ligand for PET Imaging of Prostate Cancer. Anticancer Agents Med Chem 2018; 19:509-514. [PMID: 30068284 DOI: 10.2174/1871520618666180801101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 07/20/2018] [Accepted: 07/21/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glucagon-like peptide 1 receptor (GLP-1R) is an important biomarker for diagnosis and therapy of the endocrine cancers due to overexpression. Recently, in human prostate cancer cell lines the receptor was also observed, therefore it may be a potential target for the disease. 18F-Al-NOTA-MAL-Cys39- exendin-4 holds great promise for GLP-1R. Therefore, the feasibility of the 18F-labeled exendin-4 analog for prostate cancer imaging was investigated. METHODS New probe 18F-Al-NOTA-MAL-Cys39-exendin-4 was made through one-step fluorination. Prostate cancer PC3 cell xenograft model mice were established to primarily evaluate the imaging properties of the tracer via small animal PET studies in vivo. Pathological studies and Western Blots were also performed. RESULTS PC-3 prostate xenografts were clearly imaged under baseline conditions. At 30 and 60 min postinjection, the tumor uptakes were 2.90±0.41%ID/g and 2.26±0.32 %ID/g respectively. The presence of cys39-exendin-4 significantly reduced the tumor uptake to 0.82±0.10 %ID/g at 60 min p.i. Findings of ex vivo biodistribution studies were similar to those of in vivo PET imaging. The tumors to blood and muscles were significantly improved with the increase of time due to rapid clearance of the tracer from normal organs. Low levels of radioactivity were also detected in the GLP-1R positive tumor and normal organs after coinjection with excessive unlabeled peptides. Immunohistochemistry and Western Blots results confirmed that GLP-1R was widely expressed in PC-3 prostate cancers. CONCLUSION 18F-Al labeled exendin-4 analog might be a promising tracer for in vivo detecting GLP-1R positive prostate cancer with the advantage of facile synthesis and favorable pharmacokinetics. It may be useful in differential diagnosis, molecularly targeted therapy and prognosis of the cancers.
Collapse
Affiliation(s)
- Yuanyuan Yue
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yuping Xu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Lirong Huang
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Zhicheng Bai
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Runlin Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Junjie Yan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Huizhu Song
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiaotian Li
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Min Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| |
Collapse
|
39
|
Yao HP, Feng L, Weng TH, Hu CY, Suthe SR, Mostofa AGM, Chen LH, Wu ZG, Wang WL, Wang MH. Preclinical Efficacy of Anti-RON Antibody-Drug Conjugate Zt/g4-MMAE for Targeted Therapy of Pancreatic Cancer Overexpressing RON Receptor Tyrosine Kinase. Mol Pharm 2018; 15:3260-3271. [PMID: 29944378 DOI: 10.1021/acs.molpharmaceut.8b00298] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant expression of the RON receptor tyrosine kinase, a cell surface protein, is a pathogenic feature in pancreatic cancer, which renders it a drug target for targeted therapy. Nevertheless, development of therapeutics targeting RON for pancreatic cancer therapy is hampered due to the lack of full addiction by pancreatic cancer cells to RON signaling for growth and survival. Here we describe a novel strategy using anti-RON antibody-directed drug delivery in the form of an antibody-drug conjugate for inhibition and/or eradication of pancreatic cancers. Monoclonal antibody Zt/g4 specific to the RON Sema domain was selected as the drug carrier based on its ability to induce robust RON internalization. Conjugation of Zt/g4 with monomethyl auristatin E, designated as Zt/g4-MMAE, was achieved through a protease-sensitive dipeptide linker to reach a drug to antibody ratio of 3.29:1. Zt/g4-MMAE was stable in human plasma with a dissociation rate less than 4% within a 10 day period. In vitro, Zt/g4-MMAE rapidly induced RON internalization, resulting in cell cycle arrest followed by massive cell death. The maximal effect was seen in pancreatic cancer cells with more than 10 000 receptor molecules per cell. Zt/g4-MMAE also synergized in vitro with chemotherapeutics including gemcitabine, 5-fluorouracil, and oxaliplatin to further reduce PDAC cell viability. In vivo, Zt/g4-MMAE exerts a long-lasting activity, which not only inhibited but also eradicated pancreatic xenograft tumors. These finding indicate that Zt/g4-directed drug delivery is highly effective for eradicating pancreatic tumors. Thus, Zt/g4-MMAE is a novel biotherapeutic with potential for therapy of RON-expressing pancreatic malignancies.
Collapse
|
40
|
Lee BC, Chalouni C, Doll S, Nalle SC, Darwish M, Tsai SP, Kozak KR, Del-Rosario G, Yu SF, Erickson H, Vandlen R. FRET Reagent Reveals the Intracellular Processing of Peptide-Linked Antibody–Drug Conjugates. Bioconjug Chem 2018; 29:2468-2477. [DOI: 10.1021/acs.bioconjchem.8b00362] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Byoung-Chul Lee
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Cecile Chalouni
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sophia Doll
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sam C. Nalle
- Department of Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Siao Ping Tsai
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Katherine R. Kozak
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Geoffrey Del-Rosario
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shang-Fan Yu
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hans Erickson
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Vandlen
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
41
|
Tunable cytotoxic aptamer-drug conjugates for the treatment of prostate cancer. Proc Natl Acad Sci U S A 2018; 115:4761-4766. [PMID: 29666232 DOI: 10.1073/pnas.1717705115] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Therapies that can eliminate both local and metastatic prostate tumor lesions while sparing normal organ tissue are desperately needed. With the goal of developing an improved drug-targeting strategy, we turned to a new class of targeted anticancer therapeutics: aptamers conjugated to highly toxic chemotherapeutics. Cell selection for aptamers with prostate cancer specificity yielded the E3 aptamer, which internalizes into prostate cancer cells without targeting normal prostate cells. Chemical conjugation of E3 to the drugs monomethyl auristatin E (MMAE) and monomethyl auristatin F (MMAF) yields a potent cytotoxic agent that efficiently kills prostate cancer cells in vitro but does not affect normal prostate epithelial cells. Importantly, the E3 aptamer targets tumors in vivo and treatment with the MMAF-E3 conjugate significantly inhibits prostate cancer growth in mice, demonstrating the in vivo utility of aptamer-drug conjugates. Additionally, we report the use of antidotes to block E3 aptamer-drug conjugate cytotoxicity, providing a safety switch in the unexpected event of normal cell killing in vivo.
Collapse
|
42
|
Lowe DB, Bivens CK, Mobley AS, Herrera CE, McCormick AL, Wichner T, Sabnani MK, Wood LM, Weidanz JA. TCR-like antibody drug conjugates mediate killing of tumor cells with low peptide/HLA targets. MAbs 2018; 9:603-614. [PMID: 28273004 DOI: 10.1080/19420862.2017.1302630] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
The currently marketed antibody-drug conjugates (ADC) destabilize microtubule assembly in cancer cells and initiate apoptosis in patients. However, few tumor antigens (TA) are expressed at high densities on cancer lesions, potentially minimizing the therapeutic index of current ADC regimens. The peptide/human leukocyte antigen (HLA) complex can be specifically targeted by therapeutic antibodies (designated T cell receptor [TCR]-like antibodies) and adequately distinguish malignant cells, but has not been the focus of ADC development. We analyzed the killing potential of TCR-like ADCs when cross-linked to the DNA alkylating compound duocarmycin. Our data comprise proof-of-principle results that TCR-like ADCs mediate potent tumor cytotoxicity, particularly under common scenarios of low TA/HLA density, and support their continued development alongside agents that disrupt DNA replication. Additionally, TCR-like antibody ligand binding appears to play an important role in ADC functionality and should be addressed during therapy development to avoid binding patterns that negate ADC killing efficacy.
Collapse
Affiliation(s)
- Devin B Lowe
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Camille K Bivens
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Alexis S Mobley
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Christian E Herrera
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Amanda L McCormick
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Timea Wichner
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Manoj K Sabnani
- b Department of Biology , College of Science, University of Texas at Arlington , Arlington , TX , USA
| | - Laurence M Wood
- a Department of Immunotherapeutics and Biotechnology , School of Pharmacy, Texas Tech University Health Sciences Center , Abilene , TX , USA
| | - Jon A Weidanz
- b Department of Biology , College of Science, University of Texas at Arlington , Arlington , TX , USA
| |
Collapse
|
43
|
Challenges of Antibody Drug Conjugates in Cancer Therapy: Current Understanding of Mechanisms and Future Strategies. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40495-018-0122-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
44
|
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
|
45
|
Hoffmann RM, Coumbe BGT, Josephs DH, Mele S, Ilieva KM, Cheung A, Tutt AN, Spicer JF, Thurston DE, Crescioli S, Karagiannis SN. Antibody structure and engineering considerations for the design and function of Antibody Drug Conjugates (ADCs). Oncoimmunology 2017; 7:e1395127. [PMID: 29375935 PMCID: PMC5769674 DOI: 10.1080/2162402x.2017.1395127] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 01/29/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are emerging as effective tools in cancer therapy, combining the antibody's exquisite specificity for the target antigen-expressing cancer cell together with the cytotoxic potency of the payload. Much success stems from the rational design of "toxic warheads", chemically linked to antibodies, and from fine-tuning the intricate properties of chemical linkers. Here, we focus on the antibody moiety of ADCs, dissecting the impact of Fab, linkers, isotype and Fc structure on the anti-tumoral and immune-activating functions of ADCs. Novel design approaches informed by antibody structural attributes present opportunities that may contribute to the success of next generation ADCs.
Collapse
Affiliation(s)
- Ricarda M Hoffmann
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London, King's College London, London, United Kingdom
| | - Ben G T Coumbe
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,School of Clinical Medicine, University College London Medical School, London, United Kingdom
| | - Debra H Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom
| | - Kristina M Ilieva
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Andrew N Tutt
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - James F Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - David E Thurston
- Institute of Pharmaceutical Science, King's College London, Britannia House, London, United Kingdom.,Femtogenix Ltd, BioPark, Welwyn Garden City, Hertfordshire, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London, King's College London, London, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Tower Wing, Guy's Hospital, London, United Kingdom.,NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London, King's College London, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| |
Collapse
|
46
|
Lütje S, Gerrits D, Molkenboer-Kuenen JD, Herrmann K, Fracasso G, Colombatti M, Boerman OC, Heskamp S. Characterization of Site-Specifically Conjugated Monomethyl Auristatin E– and Duocarmycin-Based Anti-PSMA Antibody–Drug Conjugates for Treatment of PSMA-Expressing Tumors. J Nucl Med 2017; 59:494-501. [DOI: 10.2967/jnumed.117.196279] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/11/2017] [Indexed: 01/06/2023] Open
|
47
|
Hu H, Zhang Y, Shukla S, Gu Y, Yu X, Steinmetz NF. Dysprosium-Modified Tobacco Mosaic Virus Nanoparticles for Ultra-High-Field Magnetic Resonance and Near-Infrared Fluorescence Imaging of Prostate Cancer. ACS NANO 2017; 11:9249-9258. [PMID: 28858475 PMCID: PMC5747565 DOI: 10.1021/acsnano.7b04472] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The increasing prevalence of ultra-high-field magnetic resonance imaging (UHFMRI) in biomedical research and clinical settings will improve the resolution and diagnostic accuracy of MRI scans. However, better contrast agents are needed to achieve a satisfactory signal-to-noise ratio. Here, we report the synthesis of a bimodal contrast agent prepared by loading the internal cavity of tobacco mosaic virus (TMV) nanoparticles with a dysprosium (Dy3+) complex and the near-infrared fluorescence (NIRF) dye Cy7.5. The external surface of TMV was conjugated with an Asp-Gly-Glu-Ala (DGEA) peptide via a polyethylene glycol linker to target integrin α2β1. The resulting nanoparticle (Dy-Cy7.5-TMV-DGEA) was stable and achieved a high transverse relaxivity in ultra-high-strength magnetic fields (326 and 399 mM-1 s-1 at 7 and 9.4 T, respectively). The contrast agent was also biocompatible (low cytotoxicity) and targeted PC-3 prostate cancer cells and tumors in vitro and in vivo as confirmed by bimodal NIRF imaging and T2-mapping UHFMRI. Our results show that Dy-Cy7.5-TMV-DGEA is suitable for multiscale MRI scanning from the cellular level to the whole body, particularly in the context of UHFMRI applications.
Collapse
Affiliation(s)
- He Hu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Yifan Zhang
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Yuning Gu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Xin Yu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Radiology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Materials Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Macromolecular Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Division of General Medical Sciences-Oncology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Corresponding Author:
| |
Collapse
|
48
|
Kalim M, Chen J, Wang S, Lin C, Ullah S, Liang K, Ding Q, Chen S, Zhan J. Intracellular trafficking of new anticancer therapeutics: antibody-drug conjugates. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2265-2276. [PMID: 28814834 PMCID: PMC5546728 DOI: 10.2147/dddt.s135571] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antibody-drug conjugate (ADC) is a milestone in targeted cancer therapy that comprises of monoclonal antibodies chemically linked to cytotoxic drugs. Internalization of ADC takes place via clathrin-mediated endocytosis, caveolae-mediated endocytosis, and pinocytosis. Conjugation strategies, endocytosis and intracellular trafficking optimization, linkers, and drugs chemistry present a great challenge for researchers to eradicate tumor cells successfully. This inventiveness of endocytosis and intracellular trafficking has given considerable momentum recently to develop specific antibodies and ADCs to treat cancer cells. It is significantly advantageous to emphasize the endocytosis and intracellular trafficking pathways efficiently and to design potent engineered conjugates and biological entities to boost efficient therapies enormously for cancer treatment. Current studies illustrate endocytosis and intracellular trafficking of ADC, protein, and linker strategies in unloading and also concisely evaluate practically applicable ADCs.
Collapse
Affiliation(s)
- Muhammad Kalim
- Department of Biochemistry and Genetics, School of Medicine
| | - Jie Chen
- Department of Biochemistry and Genetics, School of Medicine
| | - Shenghao Wang
- Department of Biochemistry and Genetics, School of Medicine
| | - Caiyao Lin
- Department of Biochemistry and Genetics, School of Medicine
| | - Saif Ullah
- Department of Biochemistry and Genetics, School of Medicine
| | - Keying Liang
- Department of Biochemistry and Genetics, School of Medicine
| | - Qian Ding
- Department of Biochemistry and Genetics, School of Medicine
| | - Shuqing Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Jinbiao Zhan
- Department of Biochemistry and Genetics, School of Medicine
| |
Collapse
|
49
|
Pina A, Dal Corso A, Caruso M, Belvisi L, Arosio D, Zanella S, Gasparri F, Albanese C, Cucchi U, Fraietta I, Marsiglio A, Pignataro L, Donati D, Gennari C. Targeting Integrin αV
β3
with Theranostic RGD-Camptothecin Conjugates Bearing a Disulfide Linker: Biological Evaluation Reveals a Complex Scenario. ChemistrySelect 2017. [DOI: 10.1002/slct.201701052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Arianna Pina
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| | - A. Dal Corso
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| | - Michele Caruso
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Laura Belvisi
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM); CNR; Via C. Golgi 19 20133 Milano Italy
| | - Simone Zanella
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| | - Fabio Gasparri
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Clara Albanese
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Ulisse Cucchi
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Ivan Fraietta
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Aurelio Marsiglio
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Luca Pignataro
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| | - Daniele Donati
- Nerviano Medical Sciences (NMS); Via Pasteur 10 20014 Nerviano Italy
| | - Cesare Gennari
- Dipartimento di Chimica; Università degli Studi di Milano; Via C. Golgi 19 20133 Milano Italy
| |
Collapse
|
50
|
Newman DJ, Cragg GM. Current Status of Marine-Derived Compounds as Warheads in Anti-Tumor Drug Candidates. Mar Drugs 2017; 15:md15040099. [PMID: 28353637 PMCID: PMC5408245 DOI: 10.3390/md15040099] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 01/09/2023] Open
Abstract
In this review, we have attempted to describe all of the antibody–drug conjugates using a marine-derived compound as the “warhead”, that are currently in clinical trials as listed in the current version of the NIH clinical trials database (clinicaltrials.gov). In searching this database, we used the beta-test version currently available, as it permitted more specific search parameters, since the regular version did not always find trials that had been completed in the past with some agents. We also added small discussion sections on candidates that are still at the preclinical stage, including a derivative of diazonamide that has an unusual interaction with tubulin (DZ-23840), which may also be a potential warhead in the future.
Collapse
|