1
|
Lin F, Yin S, Zhang Z, Yu Y, Fang H, Liang Z, Zhu R, Zhou H, Li J, Cao K, Guo W, Qin S, Zhang Y, Lu C, Li H, Liu S, Zhang H, Ye B, Lin J, Li Y, Kang X, Xi JJ, Chen PR. Multimodal targeting chimeras enable integrated immunotherapy leveraging tumor-immune microenvironment. Cell 2024:S0092-8674(24)01198-X. [PMID: 39504957 DOI: 10.1016/j.cell.2024.10.016] [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: 07/13/2022] [Revised: 08/02/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Although immunotherapy has revolutionized cancer treatment, its efficacy is affected by multiple factors, particularly those derived from the complexity and heterogeneity of the tumor-immune microenvironment (TIME). Strategies that simultaneously and synergistically engage multiple immune cells in TIME remain highly desirable but challenging. Herein, we report a multimodal and programmable platform that enables the integration of multiple therapeutic modules into single agents for tumor-targeted co-engagement of multiple immune cells within TIME. We developed the triple orthogonal linker (T-Linker) technology to integrate various therapeutic small molecules and biomolecules as multimodal targeting chimeras (Multi-TACs). The EGFR-CD3-PDL1 Multi-TAC facilitated T-dendritic cell co-engagement to target solid tumors with excellent efficacy, as demonstrated in vitro, in several humanized mouse models and in patient-derived tumor models. Furthermore, Multi-TACs were constructed to coordinate T cells with other immune cell types. The highly modular and programmable feature of our Multi-TACs may find broad applications in immunotherapy and beyond.
Collapse
Affiliation(s)
- Feng Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Shenyi Yin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zijian Zhang
- National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China
| | - Ying Yu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Haoming Fang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhen Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Rujie Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Haitao Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Kunxia Cao
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Weiming Guo
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Shan Qin
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yuxuan Zhang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chenghao Lu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Han Li
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Shibo Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Heng Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Buqing Ye
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| | - Yan Li
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210061, China; National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China.
| | - Xiaozheng Kang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
2
|
Søgaard AB, Hansson RF, Tvilum AS, Zelikin AN. Artificial Internalizing Receptors: Intracellular Delivery of Cargo Through Bio-Orthogonal Recognition. Adv Healthc Mater 2024:e2402472. [PMID: 39434478 DOI: 10.1002/adhm.202402472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/11/2024] [Indexed: 10/23/2024]
Abstract
Drug targeting is a methodology that helps to overcome the side effects of therapeutic molecules. However, insufficient targeting specificity and the on-target/off-site delivery leave much room for improvement in the targeting endeavors. One approach to enhance the specificity of drug targeting is to engineer artificial receptors with recognition ligands not observed in nature. To this end, artificial internalizing receptors that feature cholesterylamine as the artificial pull-in mechanism, and an anti-fluorescein antibody as the exofacial recognition and capture tool are developed. Fluorescein labeling is among the most routine techniques in biochemistry and can readily provide a way to make cognate derivatives for receptor-mediated endocytosis using these artificial receptors. Herein, the synthesis and the structure-activity relationship for these artificial receptors are detailed, their potency and efficacy in mediating drug delivery for the antibody-drug conjugates are illustrated, and the scope and limitations of targeting the chemically engineered cells via artificial receptors are investigated. Taken together, the presented data explore an innovative approach to drug targeting and contribute to the development of techniques in cell engineering using the tools of chemistry.
Collapse
Affiliation(s)
- Ane Bretschneider Søgaard
- iNano Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, 8000, Denmark
- Department of Chemistry, Aarhus University, Aarhus, 8000, Denmark
| | | | | | - Alexander N Zelikin
- iNano Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, 8000, Denmark
- Department of Chemistry, Aarhus University, Aarhus, 8000, Denmark
| |
Collapse
|
3
|
Ge S, Dang M, Pires Dias AC, Zhang X. Engineered IgG Fc-conjugation prolongs the half-life of florfenicol and alleviates pneumonia in mice. Biochimie 2024:S0300-9084(24)00240-2. [PMID: 39427834 DOI: 10.1016/j.biochi.2024.10.014] [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: 08/06/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Small molecule drugs often exhibit short half-lives, requiring frequent administrations to maintain therapeutic concentrations over an extended period. To address this issue, the fragment crystallizable (Fc) region of IgG, known to prolong the half-life of antibodies via its interaction with the Fc neonatal receptor, was harnessed as a carrier protein to extend the half-life of a small molecule drug, florfenicol. Florfenicol, was chemically coupled to a recombinant Fc protein expressed using the eukaryotic expression system in HEK293 cells. The Fc-florfenicol conjugate exhibited a substantially prolonged half-life of from 3.8 to 9.1 h compared to unconjugated florfenicol and demonstrated excellent therapeutic properties in treating pneumonia in a mouse model. Our results, combined with the literature analysis on Fc-small molecule conjugates, show that Fc can substantially enhance the drug's half-life and suggest the potential for its use as a carrier in novel delivery systems.
Collapse
Affiliation(s)
- Shikun Ge
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Mei Dang
- China and Portugal Joint Research Center, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Department of Biological Sciences, Faculty of Science, National University of Singapore, 10 Keng Ridge Crescent, 119260, Singapore
| | - Alberto Carlos Pires Dias
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Xiaoying Zhang
- China and Portugal Joint Research Center, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, N1G 2W1, Guelph, Ontario, Canada.
| |
Collapse
|
4
|
Pellegrino C, Favalli N, Volta L, Benz R, Puglioli S, Bassi G, Zitzmann K, Auernhammer CJ, Nölting S, Magnani CF, Neri D, Beuschlein F, Manz MG. Peptide-guided adaptor-CAR T-Cell therapy for the treatment of SSTR2-expressing neuroendocrine tumors. Oncoimmunology 2024; 13:2412371. [PMID: 39376579 PMCID: PMC11457607 DOI: 10.1080/2162402x.2024.2412371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Somatostatin receptor type 2 (SSTR2) is one of the five subtypes of somatostatin receptors and is overexpressed on the surface of most gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), pituitary tumors, paraganglioma, and meningioma, as well as hepatocellular carcinoma and breast cancer. Chimeric antigen receptor (CAR) T-cells are genetically engineered to express an artificial, T-cell activating binder, leading upon ligation to biocidal activity against target-antigen expressing cells. Adaptor-CAR T-cells recognize, via the CAR, a tag on an antigen-binding molecule, building an activating bridge between the CAR and the target cell. We hypothesized that a novel fluorescent-peptide antagonist of SSTR2, called Octo-Fluo, in combination with anti-FITC adaptor CAR (AdFITC(E2)-CAR) T-cells, may function as an on-off tunable activating bridge between the CAR and SSTR2 expressing target cells. In vitro studies confirmed the binding of Octo-Fluo to Bon1-SSTR2 mCherry-Luc cells without evidence of internalization. AdFITC(E2)-CAR T-cells were activated and efficiently induced Bon1-SSTR2 cell death in vitro, in an Octo-Fluo concentration-dependent manner. Similarly, AdFITC(E2)-CAR T-cells in combination with Octo-Fluo efficiently infiltrated the tumor and eliminated Bon1-SSTR2 tumors in immunodeficient mice in therapeutic settings. Both, AdFITC(E2)-CAR T-cell tumor infiltration and biocidal activity were Octo-Fluo concentration-dependent, with high doses of Octo-Fluo, saturating both the CAR and the SSTR2 antigen independently, leading to the loss of tumor infiltration and biocidal activity due to the loss of bridge formation. Our findings demonstrate the potential of using AdFITC(E2)-CAR T-cells with Octo-Fluo as a versatile, on-off tunable bispecific adaptor for targeted CAR T-cell immunotherapy against SSTR2-positive NETs.
Collapse
Affiliation(s)
- Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | | | - Laura Volta
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Ramon Benz
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Sara Puglioli
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Gabriele Bassi
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Kathrin Zitzmann
- Department of Medicine II, University-Hospital Munich-Grosshadern, University of Munich, Munich, Germany
| | | | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich (USZ), University of Zürich (UZH), Zürich, Switzerland
| | - Chiara F. Magnani
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Dario Neri
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich (USZ), University of Zürich (UZH), Zürich, Switzerland
- Department of Internal Medicine IV and Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Ludwig Maximilian University, LMU Klinikum, Munich, Germany
- The LOOP Zurich - Medical Research Center, Zürich, Switzerland
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| |
Collapse
|
5
|
Galbiati A, Bocci M, Ravazza D, Mock J, Gilardoni E, Neri D, Cazzamalli S. Preclinical Evaluation of 177Lu-OncoFAP-23, a Multivalent FAP-Targeted Radiopharmaceutical Therapeutic for Solid Tumors. J Nucl Med 2024; 65:1604-1610. [PMID: 39266289 DOI: 10.2967/jnumed.124.268200] [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: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Fibroblast activation protein (FAP) is abundantly expressed in the stroma of most human solid tumors. Clinical-stage radiolabeled FAP ligands are increasingly used as tools for the detection of various cancer lesions. To unleash the full therapeutic potential of FAP-targeting agents, ligands need to remain at the tumor site for several days after administration. We recently described the discovery of OncoFAP, a high-affinity small organic ligand of FAP with a rapid accumulation in tumors and low uptake in healthy tissues in cancer patients. Trimerization of OncoFAP provided a derivative (named TriOncoFAP, or OncoFAP-23) with improved FAP affinity. In this work, we evaluated the tissue biodistribution profile and the therapeutic performance of OncoFAP-23 in tumor-bearing mice. Methods: OncoFAP-23 was radiolabeled with the theranostic radionuclide 177Lu. Preclinical experiments were conducted on mice bearing SK-RC-52.hFAP (BALB/c nude mice) or CT-26.hFAP (BALB/c mice) tumors. 177Lu-OncoFAP and 177Lu-FAP-2286 were included in the biodistribution study as controls. Toxicologic evaluation was performed on Wistar rats and CD1 mice by injecting high doses of OncoFAP-23 or its cold-labeled counterpart, respectively. Results: 177Lu-OncoFAP-23 emerged for its best-in-class biodistribution profile, high and prolonged tumor uptake (i.e., ∼16 percentage injected dose/g at 96 h), and low accumulation in healthy organs, which correlates well with its potent single-agent anticancer activity at low levels of administered radioactivity. Combination treatment with the tumor-targeted interleukin 2 (L19-IL2, a clinical-stage immunocytokine) further expands the therapeutic window of 177Lu-OncoFAP-23 by potentiating its in vivo antitumor activity. Proteomics studies revealed a potent tumor-directed immune response on treatment with the combination. OncoFAP-23 and natLu-OncoFAP-23 exhibited a favorable toxicologic profile, without showing any side effects or signs of toxicity. Conclusion: OncoFAP-23 presents enhanced tumor uptake and tumor retention and low accumulation in healthy organs, findings that correspond to a strongly improved in vivo antitumor efficacy. The data presented in this work support the clinical development of 177Lu-OncoFAP-23 for the treatment of FAP-positive solid tumors.
Collapse
Affiliation(s)
| | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Switzerland
| | | | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland; and
- Philogen S.p.A., Siena, Italy
| | | |
Collapse
|
6
|
Liao ZX, Huang PH, Hsu SH, Chang HH, Chang CH, Tseng SJ. Clinical strategies with antibody-drug conjugates as potential modifications for virotherapy. Drug Discov Today 2024; 29:104165. [PMID: 39270970 DOI: 10.1016/j.drudis.2024.104165] [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: 06/08/2023] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
The ability to selectively target cancer cells makes antibody-drug conjugates (ADCs) promising therapeutic options. They have been tested in clinical trials as a vehicle for tumor-specific delivery of cytotoxic payloads for a range of cancers. However, systemic administration of oncolytic virotherapy is challenging, because only a small portion of injected viruses reach the target. Despite the approval of higher viral doses, most viruses still end up in the liver, potentially causing toxicity in that organ. Integrating ADCs with virotherapy in the form of antibody-virus conjugates or virus-drug conjugates can potentially overcome these challenges and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Zi-Xian Liao
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Po-Hsiang Huang
- Department of Oncology, National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10051, Taiwan
| | - Hsiung-Hao Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chi-Heng Chang
- School of Pharmacy, National Cheng Kung University, Tainan, 70101, Taiwan
| | - S-Ja Tseng
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; NCKU Center of Applied Nanomedicine, Tainan 70101, Taiwan.
| |
Collapse
|
7
|
Prodi E, Neri D, De Luca R. Tumor-Homing Antibody-Cytokine Fusions for Cancer Therapy. Onco Targets Ther 2024; 17:697-715. [PMID: 39224695 PMCID: PMC11368152 DOI: 10.2147/ott.s480787] [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: 07/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Recombinant cytokine products have emerged as a promising avenue in cancer therapy due to their capacity to modulate and enhance the immune response against tumors. However, their clinical application is significantly hindered by systemic toxicities already at low doses, thus preventing escalation to therapeutically active regimens. One promising approach to overcoming these limitations is using antibody-cytokine fusion proteins (also called immunocytokines). These biopharmaceuticals leverage the targeting specificity of antibodies to deliver cytokines directly to the tumor microenvironment, thereby reducing systemic exposure and enhancing the therapeutic index. This review comprehensively examines the development and potential of antibody-cytokine fusion proteins in cancer therapy. It explores the molecular characteristics that influence the performance of these fusion proteins, and it highlights key findings from preclinical and clinical studies, illustrating the potential of immunocytokines to improve treatment outcomes in cancer patients. Recent advancements in the field, such as novel engineering strategies and combination strategies to enhance the efficacy and safety of immunocytokines, are also discussed. These innovations offer new opportunities to optimize this class of biotherapeutics, making them a more viable and effective option for cancer treatment. As the field continues to evolve, understanding the critical factors that influence the performance of immunocytokines will be essential for successfully translating these therapies into clinical practice.
Collapse
Affiliation(s)
- Eleonora Prodi
- Philochem AG, Otelfingen, 8112, Switzerland
- University of Trento, Italy, CiBIO (Department of Cellular, Computational and Integrative Biology), Povo, 38123, Trento
| | - Dario Neri
- Philogen Spa, Siena, 53100, Italy
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
8
|
Lindeman SD, Booth OC, Tudi P, Schleinkofer TC, Moss JN, Kearney NB, Mukkamala R, Thompson LK, Modany MA, Srinivasarao M, Low PS. FAP Radioligand Linker Optimization Improves Tumor Dose and Tumor-to-Healthy Organ Ratios in 4T1 Syngeneic Model. J Med Chem 2024; 67:11827-11840. [PMID: 39013156 DOI: 10.1021/acs.jmedchem.4c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Fibroblast activation protein (FAP) has attracted considerable attention as a possible target for the radiotherapy of solid tumors. Unfortunately, initial efforts to treat solid tumors with FAP-targeted radionuclides have yielded only modest clinical responses, suggesting that further improvements in the molecular design of FAP-targeted radiopharmaceutical therapies (RPT) are warranted. In this study, we report several advances on the previously described FAP6 radioligand that increase tumor retention and accelerate healthy tissue clearance. Seven FAP6 derivatives with different linkers or albumin binders were synthesized, radiolabeled, and investigated for their effects on binding and cellular uptake. The radioligands were then characterized in 4T1 tumor-bearing Balb/c mice using both single-photon emission computed tomography (SPECT) and ex vivo biodistribution analyses to identify the conjugate with the best tumor retention and tumor-to-healthy organ ratios. The results reveal an optimized FAP6 radioligand that exhibits efficacy and safety properties that potentially justify its translation into the clinic.
Collapse
Affiliation(s)
- Spencer D Lindeman
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| | - Owen C Booth
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pooja Tudi
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Taylor C Schleinkofer
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jackson N Moss
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nicholas B Kearney
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ramesh Mukkamala
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lauren K Thompson
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mollie A Modany
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| |
Collapse
|
9
|
Paulus J, Sewald N. Small molecule- and peptide-drug conjugates addressing integrins: A story of targeted cancer treatment. J Pept Sci 2024; 30:e3561. [PMID: 38382900 DOI: 10.1002/psc.3561] [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: 09/20/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 02/23/2024]
Abstract
Targeted cancer treatment should avoid side effects and damage to healthy cells commonly encountered during traditional chemotherapy. By combining small molecule or peptidic ligands as homing devices with cytotoxic drugs connected by a cleavable or non-cleavable linker in peptide-drug conjugates (PDCs) or small molecule-drug conjugates (SMDCs), cancer cells and tumours can be selectively targeted. The development of highly affine, selective peptides and small molecules in recent years has allowed PDCs and SMDCs to increasingly compete with antibody-drug conjugates (ADCs). Integrins represent an excellent target for conjugates because they are overexpressed by most cancer cells and because of the broad knowledge about native binding partners as well as the multitude of small-molecule and peptidic ligands that have been developed over the last 30 years. In particular, integrin αVβ3 has been addressed using a variety of different PDCs and SMDCs over the last two decades, following various strategies. This review summarises and describes integrin-addressing PDCs and SMDCs while highlighting points of great interest.
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
10
|
Obara T, Kawano N, Tatsumi K, Katsuyama A, Nakajima K, Ogawa M, Ichikawa S. Development of small molecule-drug conjugates based on derivatives of natural proteasome inhibitors that exhibit selectivity for PSMA-expressing cancer cells. Bioorg Med Chem 2024; 108:117773. [PMID: 38850999 DOI: 10.1016/j.bmc.2024.117773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/10/2024]
Abstract
In this study, we have developedsmall molecule drug conjugates (SMDCs)consisting ofa prostate specific membrane antigen (PSMA) ligandand syringolin derivatives, which are potent proteasome inhibitors, to selectively deliver syringolin derivatives to prostate cancer cells. Two parent compounds were used for syringolin derivatives with different linkage sites. These SMDCs exhibited PSMA-expressing cell-selective cytotoxicity and they could potentially be used for safer treatment of cancer.
Collapse
Affiliation(s)
- Takahiro Obara
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Nanami Kawano
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Kengo Tatsumi
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Akira Katsuyama
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Kohei Nakajima
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mikako Ogawa
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Satoshi Ichikawa
- Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
11
|
Keller M, Petrov D, Gloger A, Dietschi B, Jobin K, Gradinger T, Martinelli A, Plais L, Onda Y, Neri D, Scheuermann J. Highly pure DNA-encoded chemical libraries by dual-linker solid-phase synthesis. Science 2024; 384:1259-1265. [PMID: 38870307 DOI: 10.1126/science.adn3412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/07/2024] [Indexed: 06/15/2024]
Abstract
The first drugs discovered using DNA-encoded chemical library (DEL) screens have entered late-stage clinical development. However, DEL technology as a whole still suffers from poor chemical purity resulting in suboptimal performance. In this work, we report a technique to overcome this issue through self-purifying release of the DEL after magnetic bead-based synthesis. Both the first and last building blocks of each assembled library member were linked to the beads by tethers that could be cleaved by mutually orthogonal chemistry. Sequential cleavage of the first and last tether, with washing in between, ensured that the final library comprises only the fully complete compounds. The outstanding purity attained by this approach enables a direct correlation of chemical display and encoding, allows for an increased chemical reaction scope, and facilitates the use of more diversity elements while achieving greatly improved signal-to-noise ratios in selections.
Collapse
Affiliation(s)
- Michelle Keller
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Dimitar Petrov
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas Gloger
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Bastien Dietschi
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Kilian Jobin
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Timon Gradinger
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Louise Plais
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Yuichi Onda
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jörg Scheuermann
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
12
|
Zhang J, Hu F, Aras O, Chai Y, An F. Small Molecule-Drug Conjugates: Opportunities for the Development of Targeted Anticancer Drugs. ChemMedChem 2024; 19:e202300720. [PMID: 38396351 DOI: 10.1002/cmdc.202300720] [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: 12/21/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
Conventional chemotherapy is insufficient for precise cancer treatment due to its lack of selectivity and inevitable side effects. Targeted drugs have emerged as a promising solution for precise cancer treatment. A common strategy is to conjugate therapeutic agents with ligands that can specifically bind to tumor cells, providing targeted therapy. Similar to the more successful antibody drug conjugates (ADCs), small molecule drug conjugates (SMDCs) are another promising class of targeted drugs, consisting of three parts: targeting ligand, cleavable linker and payload. Compared to ADCs, SMDCs have the advantages of smaller size, better permeability, simpler preparation process and non-immunogenicity, making them a promising alternative to ADCs. This review describes the characteristics of the targeting ligand, linker and payload of SMDCs and the criteria for selecting a suitable one. We also discuss recently reported SMDCs and list some successful SMDCs that have entered clinical trials.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Fanchun Hu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yichao Chai
- Department of Oncology, The Second Affiliated Hospital of Xi'an, Jiaotong University, No.157 Xiwu Road, Xincheng District, Xi'an, Shaanxi, 710004, China
| | - Feifei An
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
13
|
Ning D, Xue J, Lou X, Shao R, Liu Y, Chen G. Transforming toxins into treatments: the revolutionary role of α-amanitin in cancer therapy. Arch Toxicol 2024; 98:1705-1716. [PMID: 38555326 DOI: 10.1007/s00204-024-03727-0] [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: 01/17/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Amanita phalloides is the primary species responsible for fatal mushroom poisoning, as its main toxin, α-amanitin, irreversibly and potently inhibits eukaryotic RNA polymerase II (RNAP II), leading to cell death. There is no specific antidote for α-amanitin, which hinders its clinical application. However, with the advancement of precision medicine in oncology, including the development of antibody-drug conjugates (ADCs), the potential value of various toxic small molecules has been explored. These ADCs ingeniously combine the targeting precision of antibodies with the cytotoxicity of small-molecule payloads to precisely kill tumor cells. We searched PubMed for studies in this area using these MeSH terms "Amanitins, Alpha-Amanitin, Therapeutic use, Immunotherapy, Immunoconjugates, Antibodies" and did not limit the time interval. Recent studies have conducted preclinical experiments on ADCs based on α-amanitin, showing promising therapeutic effects and good tolerance in primates. The current challenges include the not fully understood toxicological mechanism of α-amanitin and the lack of clinical studies to evaluate the therapeutic efficacy of ADCs developed based on α-amanitin. In this article, we will discuss the role and therapeutic efficacy of α-amanitin as an effective payload in ADCs for the treatment of various cancers, providing background information for the research and application strategies of current and future drugs.
Collapse
Affiliation(s)
- Deyuan Ning
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinfang Xue
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Liu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, No 157 Jinbi Road, Xishan District, Kunming, 650032, China.
| |
Collapse
|
14
|
Zheng Y, Xu R, Cheng H, Tai W. Mono-amino acid linkers enable highly potent small molecule-drug conjugates by conditional release. Mol Ther 2024; 32:1048-1060. [PMID: 38369752 PMCID: PMC11163218 DOI: 10.1016/j.ymthe.2024.02.020] [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: 09/15/2023] [Revised: 01/18/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
The endosome cleavable linkers have been widely employed by antibody-drug conjugates and small molecule-drug conjugates (SMDCs) to control the accurate release of payloads. An effective linker should provide stability in systemic circulation but efficient payload release at its targeted tumor sites. This conflicting requirement always leads to linker design with increasing structural complexity. Balance of the effectiveness and structural complexity presents a linker design challenge. Here, we explored the possibility of mono-amino acid as so far the simplest cleavable linker (X-linker) for SMDC-based auristatin delivery. Within a diverse set of X-linkers, the SMDCs differed widely in bioactivity, with one (Asn-linker) having significantly improved potency (IC50 = 0.1 nM) and fast response to endosomal cathepsin B cleavage. Notably, this SMDC, once grafted with effector protein fragment crystallizable (Fc), demonstrated a profound in vivo therapeutic effect in aspects of targetability, circulation half-life (t1/2 = 73 h), stability, and anti-tumor efficacy. On the basis of these results, we believe that this mono-amino acid linker, together with the new SMDC-Fc scaffold, has significant potential in targeted delivery application.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Ruolin Xu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
15
|
Chen Z, Liu M, Wang N, Xiao W, Shi J. Unleashing the Potential of Camptothecin: Exploring Innovative Strategies for Structural Modification and Therapeutic Advancements. J Med Chem 2024; 67:3244-3273. [PMID: 38421819 DOI: 10.1021/acs.jmedchem.3c02115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Camptothecin (CPT) is a potent anti-cancer agent targeting topoisomerase I (TOP1). However, CPT has poor pharmacokinetic properties, causes toxicities, and leads to drug resistance, which limit its clinical use. In this paper, to review the current state of CPT research. We first briefly explain CPT's TOP1 inhibition mechanism and the key hurdles in CPT drug development. Then we examine strategies to overcome CPT's limitations through structural modifications and advanced delivery systems. Though modifications alone seem insufficient to fully enhance CPT's therapeutic potential, structure-activity relationship analysis provides insights to guide optimization of CPT analogs. In comparison, advanced delivery systems integrating controlled release, imaging capabilities, and combination therapies via stimulus-responsive linkers and targeting moieties show great promise for improving CPT's pharmacological profile. Looking forward, multifaceted approaches combining selective CPT derivatives with advanced delivery systems, informed by emerging biological insights, hold promise for fully unleashing CPT's anti-cancer potential.
Collapse
Affiliation(s)
- Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Maoyu Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu 610083, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
16
|
Bocci M, Zana A, Principi L, Lucaroni L, Prati L, Gilardoni E, Neri D, Cazzamalli S, Galbiati A. In vivo activation of FAP-cleavable small molecule-drug conjugates for the targeted delivery of camptothecins and tubulin poisons to the tumor microenvironment. J Control Release 2024; 367:779-790. [PMID: 38346501 DOI: 10.1016/j.jconrel.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Small molecule-drug conjugates (SMDCs) are increasingly considered as a therapeutic alternative to antibody-drug conjugates (ADCs) for cancer therapy. OncoFAP is an ultra-high affinity ligand of Fibroblast Activation Protein (FAP), a stromal tumor-associated antigen overexpressed in a wide variety of solid human malignancies. We have recently reported the development of non-internalizing OncoFAP-based SMDCs, which are activated by FAP thanks to selective proteolytic cleavage of the -GlyPro- linker with consequent release of monomethyl auristatin E (MMAE) in the tumor microenvironment. In this article, we describe the generation and the in vivo characterization of FAP-cleavable OncoFAP-drug conjugates based on potent topoisomerase I inhibitors (DXd, SN-38, and exatecan) and an anti-tubulin payload (MMAE), which are already exploited in clinical-stage and approved ADCs. The Glycine-Proline FAP-cleavable technology was directly benchmarked against linkers found in Adcetris™, Enhertu™, and Trodelvy™ structures by means of in vivo therapeutic experiments in mice bearing tumors with cellular or stromal FAP expression. OncoFAP-GlyPro-Exatecan and OncoFAP-GlyPro-MMAE emerged as the most efficacious anti-cancer therapeutics against FAP-positive cellular models. OncoFAP-GlyPro-MMAE exhibited a potent antitumor activity also against stromal models, and was therefore selected for clinical development.
Collapse
Affiliation(s)
- Matilde Bocci
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| | - Aureliano Zana
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Laura Lucaroni
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | - Luca Prati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Dario Neri
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, Zurich CH-8093, Switzerland; Philogen S.p.A., Siena 53100, Italy
| | | | - Andrea Galbiati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| |
Collapse
|
17
|
Rebstock AS, Wiedmann M, Stelte-Ludwig B, Wong H, Johnson AJ, Izumi R, Hamdy A, Lerchen HG. Neutrophil elastase as a versatile cleavage enzyme for activation of αvβ3 integrin-targeted small molecule drug conjugates with different payload classes in the tumor microenvironment. Front Pharmacol 2024; 15:1358393. [PMID: 38495100 PMCID: PMC10943695 DOI: 10.3389/fphar.2024.1358393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/06/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction: The development of bioconjugates for the targeted delivery of anticancer agents is gaining momentum after recent success of antibody drug conjugates (ADCs) in the clinic. Smaller format conjugates may have several advantages including better tumor penetration; however, cellular uptake and trafficking may be substantially different from ADCs. To fully leverage the potential of small molecule drug conjugates (SMDCs) with potent binding molecules mediating tumor homing, novel linker chemistries susceptible for efficient extracellular activation and payload release in the tumor microenvironment (TME) need to be explored. Methods: We designed a novel class of SMDCs, which target αvβ3 integrins for tumor homing and are cleaved by neutrophil elastase (NE), a serine protease active in the TME. A peptidomimetic αvβ3 ligand was attached via optimized linkers composed of substrate peptide sequences of NE connected to different functional groups of various payload classes, such as camptothecins, monomethyl auristatin E, kinesin spindle protein inhibitors (KSPi) and cyclin-dependent kinase 9 inhibitors (CDK-9i). Results: NE-mediated cleavage was found compatible with the diverse linker attachments via hindered ester bonds, amide bonds and sulfoximide bonds. Efficient and traceless release of the respective payloads was demonstrated in biochemical assays. The newly designed SMDCs were highly stable in buffer as well as in rat and human plasma. Cytotoxicity of the SMDCs in cancer cell lines was clearly dependent on NE. IC50 values were in the nanomolar or sub-nanomolar range across several cancer cell lines reaching similar potencies as compared to the respective payloads only in the presence of NE. In vivo pharmacokinetics evaluating SMDC and free payload exposures in rat and particularly the robust efficacy with good tolerability in triple negative breast and small cell lung cancer murine models demonstrate the utility of this approach for selective delivery of payloads to the tumor. Discussion: These results highlight the broad scope of potential payloads and suitable conjugation chemistries paving the way for future SMDCs harnessing the safety features of targeted delivery approaches in combination with NE cleavage in the TME.
Collapse
Affiliation(s)
| | | | | | - Harvey Wong
- Vincerx Pharma, Inc., Palo Alto, CA, United States
| | | | - Raquel Izumi
- Vincerx Pharma, Inc., Palo Alto, CA, United States
| | - Ahmed Hamdy
- Vincerx Pharma, Inc., Palo Alto, CA, United States
| | | |
Collapse
|
18
|
Ahmadi M, Ritter CA, von Woedtke T, Bekeschus S, Wende K. Package delivered: folate receptor-mediated transporters in cancer therapy and diagnosis. Chem Sci 2024; 15:1966-2006. [PMID: 38332833 PMCID: PMC10848714 DOI: 10.1039/d3sc05539f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/31/2023] [Indexed: 02/10/2024] Open
Abstract
Neoplasias pose a significant threat to aging society, underscoring the urgent need to overcome the limitations of traditional chemotherapy through pioneering strategies. Targeted drug delivery is an evolving frontier in cancer therapy, aiming to enhance treatment efficacy while mitigating undesirable side effects. One promising avenue utilizes cell membrane receptors like the folate receptor to guide drug transporters precisely to malignant cells. Based on the cellular folate receptor as a cancer cell hallmark, targeted nanocarriers and small molecule-drug conjugates have been developed that comprise different (bio) chemistries and/or mechanical properties with individual advantages and challenges. Such modern folic acid-conjugated stimuli-responsive drug transporters provide systemic drug delivery and controlled release, enabling reduced dosages, circumvention of drug resistance, and diminished adverse effects. Since the drug transporters' structure-based de novo design is increasingly relevant for precision cancer remediation and diagnosis, this review seeks to collect and debate the recent approaches to deliver therapeutics or diagnostics based on folic acid conjugated Trojan Horses and to facilitate the understanding of the relevant chemistry and biochemical pathways. Focusing exemplarily on brain and breast cancer, recent advances spanning 2017 to 2023 in conjugated nanocarriers and small molecule drug conjugates were considered, evaluating the chemical and biological aspects in order to improve accessibility to the field and to bridge chemical and biomedical points of view ultimately guiding future research in FR-targeted cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| | - Christoph A Ritter
- Institute of Pharmacy, Section Clinical Pharmacy, University of Greifswald Greifswald Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center Ferdinand-Sauerbruch-Straße 17475 Greifswald Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Clinic and Policlinic for Dermatology and Venereology, Rostock University Medical Center Strempelstr. 13 18057 Rostock Germany
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| |
Collapse
|
19
|
Joseph J, Sandel G, Kulkarni R, Alatrash R, Herrera BB, Jain P. Antibody and Cell-Based Therapies against Virus-Induced Cancers in the Context of HIV/AIDS. Pathogens 2023; 13:14. [PMID: 38251321 PMCID: PMC10821063 DOI: 10.3390/pathogens13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Infectious agents, notably viruses, can cause or increase the risk of cancer occurrences. These agents often disrupt normal cellular functions, promote uncontrolled proliferation and growth, and trigger chronic inflammation, leading to cancer. Approximately 20% of all cancer cases in humans are associated with an infectious pathogen. The International Agency for Research on Cancer (IARC) recognizes seven viruses as direct oncogenic agents, including Epstein-Barr Virus (EBV), Kaposi's Sarcoma-associated herpesvirus (KSHV), human T-cell leukemia virus type-1 (HTLV-1), human papilloma virus (HPV), hepatitis C virus (HCV), hepatitis B virus (HBV), and human immunodeficiency virus type 1 (HIV-1). Most viruses linked to increased cancer risk are typically transmitted through contact with contaminated body fluids and high-risk behaviors. The risk of infection can be reduced through vaccinations and routine testing, as well as recognizing and addressing risky behaviors and staying informed about public health concerns. Numerous strategies are currently in pre-clinical phases or undergoing clinical trials for targeting cancers driven by viral infections. Herein, we provide an overview of risk factors associated with increased cancer incidence in people living with HIV (PLWH) as well as other chronic viral infections, and contributing factors such as aging, toxicity from ART, coinfections, and comorbidities. Furthermore, we highlight both antibody- and cell-based strategies directed against virus-induced cancers while also emphasizing approaches aimed at discovering cures or achieving complete remission for affected individuals.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Grace Sandel
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Ratuja Kulkarni
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Reem Alatrash
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Bobby Brooke Herrera
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pooja Jain
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| |
Collapse
|
20
|
Bisbal Lopez L, Ravazza D, Bocci M, Zana A, Principi L, Dakhel Plaza S, Galbiati A, Gilardoni E, Scheuermann J, Neri D, Pignataro L, Gennari C, Cazzamalli S, Dal Corso A. Ex vivo mass spectrometry-based biodistribution analysis of an antibody-Resiquimod conjugate bearing a protease-cleavable and acid-labile linker. Front Pharmacol 2023; 14:1320524. [PMID: 38125888 PMCID: PMC10731371 DOI: 10.3389/fphar.2023.1320524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Immune-stimulating antibody conjugates (ISACs) equipped with imidazoquinoline (IMD) payloads can stimulate endogenous immune cells to kill cancer cells, ultimately inducing long-lasting anticancer effects. A novel ISAC was designed, featuring the IMD Resiquimod (R848), a tumor-targeting antibody specific for Carbonic Anhydrase IX (CAIX) and the protease-cleavable Val-Cit-PABC linker. In vitro stability analysis showed not only R848 release in the presence of the protease Cathepsin B but also under acidic conditions. The ex vivo mass spectrometry-based biodistribution data confirmed the low stability of the linker-drug connection while highlighting the selective accumulation of the IgG in tumors and its long circulatory half-life.
Collapse
Affiliation(s)
| | | | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Switzerland
| | | | | | | | | | | | - Jörg Scheuermann
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Dario Neri
- R&D Department, Philochem AG, Otelfingen, Switzerland
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
- Philogen S.p.A, Siena, Italy
| | - Luca Pignataro
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| | - Cesare Gennari
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| | | | - Alberto Dal Corso
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
21
|
Huet S, Zeisser Labouebe M, Castro R, Jacquot P, Pedrault J, Viollet S, Van Simaeys G, Doumont G, Larbanoix L, Zindy E, Cunha AE, Scapozza L, Cinier M. Targeted Nanofitin-drug Conjugates Achieve Efficient Tumor Delivery and Therapeutic Effect in an EGFRpos Mouse Xenograft Model. Mol Cancer Ther 2023; 22:1343-1351. [PMID: 37578807 PMCID: PMC10618730 DOI: 10.1158/1535-7163.mct-22-0805] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/13/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Adjusting the molecular size, the valency and the pharmacokinetics of drug conjugates are as many leverages to improve their therapeutic window, notably by affecting tumor penetration, renal clearance, and short systemic exposure. In that regard, small tumor-targeting ligands are gaining attention. In this study, we demonstrate the benefits of the small Nanofitin alternative scaffolds (7 kDa) as selective tumor-targeting modules for the generation of drug conjugates, focusing on Nanofitins B10 and D8 directed against the EGFR. Owing to their small size and monovalent format, the two Nanofitins displayed a fast and deep tumor penetration in EGFR-positive A431 xenografts in BALB/c nude mice after intravenous administration, yielding to a targeting of respectively 67.9% ± 14.1 and 98.9% ± 0.7 of the tumor cells as demonstrated by IHC. Conjugation with the monomethyl auristatin E toxin provided homogeneous Nanofitin-drug conjugates, with an overall yield of ≥97%, for in vivo assessment in a curative xenograft model using bioluminescent, EGFR-positive, A431 cells in BALB/c nude mice. Internalization was found critical for efficient release of the toxin. Hence, the intravenous administration of the D8-based construct showed significant antitumor effect in vivo as determined by monitoring tumor volumes and bioluminescence levels over 2 months.
Collapse
Affiliation(s)
| | - Magali Zeisser Labouebe
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Rute Castro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | | | | | | | - Gaetan Van Simaeys
- CMMI, Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi (Gosselies), Belgium
| | - Gilles Doumont
- CMMI, Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi (Gosselies), Belgium
| | - Lionel Larbanoix
- CMMI, Center for Microscopy and Molecular Imaging, Université de Mons, Charleroi (Gosselies), Belgium
| | - Egor Zindy
- CMMI, Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi (Gosselies), Belgium
| | - António E. Cunha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
22
|
Lerchen HG, Stelte-Ludwig B, Heroult M, Zubov D, Gericke KM, Wong H, Frigault MM, Johnson AJ, Izumi R, Hamdy A. Discovery of VIP236, an αvβ3-Targeted Small-Molecule-Drug Conjugate with Neutrophil Elastase-Mediated Activation of 7-Ethyl Camptothecin Payload for Treatment of Solid Tumors. Cancers (Basel) 2023; 15:4381. [PMID: 37686656 PMCID: PMC10486604 DOI: 10.3390/cancers15174381] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The emerging field of small-molecule-drug conjugates (SMDCs) using small-molecule biomarker-targeted compounds for tumor homing may provide new perspectives for targeted delivery. Here, for the first time, we disclose the structure and the synthesis of VIP236, an SMDC designed for the treatment of metastatic solid tumors by targeting αvβ3 integrins and extracellular cleavage of the 7-ethyl camptothecin payload by neutrophil elastase in the tumor microenvironment. Imaging studies in the Lewis lung mouse model using an elastase cleavable quenched substrate showed pronounced elastase activity in the tumor. Pharmacokinetics studies of VIP236 in tumor-bearing mice demonstrated high stability of the SMDC in plasma and high tumor accumulation of the cleaved payload. Studies in bile-duct-cannulated rats showed that biliary excretion of the unmodified conjugate is the primary route of elimination. Treatment- and time-dependent phosphorylation of H2AX, a marker of DNA damage downstream of topoisomerase 1 inhibition, verified the on-target activity of the payload cleaved from VIP236 in vivo. Treatment with VIP236 resulted in long-lasting tumor regression in subcutaneous patient-derived xenograft (PDX) models from patients with non-small-cell lung, colon, and renal cancer as well as in two orthotopic metastatic triple-negative breast cancer PDX models. In these models, a significant reduction of brain and lung metastases also was observed.
Collapse
Affiliation(s)
| | | | | | - Dmitry Zubov
- Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (K.M.G.)
| | | | - Harvey Wong
- Vincerx Pharma, Inc., Palo Alto, CA 94306, USA; (H.W.); (M.M.F.); (A.J.J.); (R.I.); (A.H.)
| | - Melanie M. Frigault
- Vincerx Pharma, Inc., Palo Alto, CA 94306, USA; (H.W.); (M.M.F.); (A.J.J.); (R.I.); (A.H.)
| | - Amy J. Johnson
- Vincerx Pharma, Inc., Palo Alto, CA 94306, USA; (H.W.); (M.M.F.); (A.J.J.); (R.I.); (A.H.)
| | - Raquel Izumi
- Vincerx Pharma, Inc., Palo Alto, CA 94306, USA; (H.W.); (M.M.F.); (A.J.J.); (R.I.); (A.H.)
| | - Ahmed Hamdy
- Vincerx Pharma, Inc., Palo Alto, CA 94306, USA; (H.W.); (M.M.F.); (A.J.J.); (R.I.); (A.H.)
| |
Collapse
|
23
|
Mortensen M, Bertolini M, Mock J, Scheuermann J, Oehler S. Site-Specific Chemical Modification of a Cytokine Mimic for Small Molecule-Based Tumor Targeting. Bioconjug Chem 2023; 34:1374-1379. [PMID: 37462264 DOI: 10.1021/acs.bioconjchem.3c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The targeted delivery of bioactive proteins, such as cytokines, for cancer immunotherapy approaches mostly relies on antibodies or antibody fragments. However, fusion proteins may display low tissue penetration due to a large molecular size. Small molecule ligands with high affinity toward tumor-associated antigens provide a promising alternative for the selective delivery of cytokines to tumor lesions. We developed a one-pot procedure for the site-specific thiazolidine formation between an aldehyde bearing small molecule and the in situ generated N-terminal cysteine of a bioactive protein. Thereby, neoleukin-2/15 (Neo-2/15), a computationally engineered interleukin-2 and -15 mimic, was chemically conjugated to acetazolamide plus, a potent carbonic anhydrase IX (CAIX) ligand. The conjugate retained the biological activity of Neo-2/15 and revealed its ability to accumulate in renal cell carcinoma (SK-RC-52) xenografts upon systemic intravenous administration. The results highlight the potential of small molecule targeting moieties to drive the accumulation of a protein cargo to the respective disease site while conserving the small construct size.
Collapse
Affiliation(s)
- Michael Mortensen
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8112 Zürich, Switzerland
| | - Marco Bertolini
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8112 Zürich, Switzerland
| | - Jacqueline Mock
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8112 Zürich, Switzerland
| | - Jörg Scheuermann
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8112 Zürich, Switzerland
| | - Sebastian Oehler
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8112 Zürich, Switzerland
| |
Collapse
|
24
|
Zana A, Puig-Moreno C, Bocci M, Gilardoni E, Di Nitto C, Principi L, Ravazza D, Rotta G, Prodi E, De Luca R, Neri D, Cazzamalli S. A Comparative Analysis of Fibroblast Activation Protein-Targeted Small Molecule-Drug, Antibody-Drug, and Peptide-Drug Conjugates. Bioconjug Chem 2023. [PMID: 37399501 DOI: 10.1021/acs.bioconjchem.3c00244] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
We present the first in vivo comparative evaluation of chemically defined antibody-drug conjugates (ADCs), small molecule-drug conjugates (SMDCs), and peptide-drug conjugates (PDCs) targeting and activated by fibroblast activation protein (FAP) in solid tumors. Both the SMDC (OncoFAP-Gly-Pro-MMAE) and the ADC (7NP2-Gly-Pro-MMAE) candidates delivered high amounts of active payload (i.e., MMAE) selectively at the tumor site, thus producing a potent antitumor activity in a preclinical cancer model.
Collapse
Affiliation(s)
- Aureliano Zana
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Claudia Puig-Moreno
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, CH-8093 Zürich, Switzerland
| | - Matilde Bocci
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Ettore Gilardoni
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Cesare Di Nitto
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Lucrezia Principi
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Domenico Ravazza
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Giulia Rotta
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Eleonora Prodi
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Roberto De Luca
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Dario Neri
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, CH-8093 Zürich, Switzerland
- Philogen S.p.A., 53100 Siena, Italy
| | - Samuele Cazzamalli
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| |
Collapse
|
25
|
Kim JH, Park S, Jung E, Shin J, Kim YJ, Kim JY, Sessler JL, Seo JH, Kim JS. A dual-action niclosamide-based prodrug that targets cancer stem cells and inhibits TNBC metastasis. Proc Natl Acad Sci U S A 2023; 120:e2304081120. [PMID: 37186828 PMCID: PMC10214212 DOI: 10.1073/pnas.2304081120] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Chemotherapy typically destroys the tumor mass but rarely eradicates the cancer stem cells (CSCs) that can drive metastatic recurrence. A key current challenge is finding ways to eradicate CSCs and suppress their characteristics. Here, we report a prodrug, Nic-A, created by combining a carbonic anhydrase IX (CAIX) inhibitor, acetazolamide, with a signal transducer and transcriptional activator 3 (STAT3) inhibitor, niclosamide. Nic-A was designed to target triple-negative breast cancer (TNBC) CSCs and was found to inhibit both proliferating TNBC cells and CSCs via STAT3 dysregulation and suppression of CSC-like properties. Its use leads to a decrease in aldehyde dehydrogenase 1 activity, CD44high/CD24low stem-like subpopulations, and tumor spheroid-forming ability. TNBC xenograft tumors treated with Nic-A exhibited decreased angiogenesis and tumor growth, as well as decreased Ki-67 expression and increased apoptosis. In addition, distant metastases were suppressed in TNBC allografts derived from a CSC-enriched population. This study thus highlights a potential strategy for addressing CSC-based cancer recurrence.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department of Chemistry, Korea University, Seoul02841, Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jinwoo Shin
- Department of Chemistry, Korea University, Seoul02841, Korea
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, TX78712-1224
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul02841, Korea
| |
Collapse
|
26
|
Aigbogun OP, Phenix CP, Krol ES, Price EW. The Chemistry of Creating Chemically Programmed Antibodies (cPAbs): Site-Specific Bioconjugation of Small Molecules. Mol Pharm 2023; 20:853-874. [PMID: 36696533 DOI: 10.1021/acs.molpharmaceut.2c00821] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Small-molecule drugs have been employed for years as therapeutics in the pharmaceutical industry. However, small-molecule drugs typically have short in vivo half-lives which is one of the largest impediments to the success of many potentially valuable pharmacologically active small molecules. The undesirable pharmacokinetics and pharmacology associated with some small molecules have led to the development of a new class of bioconjugates known as chemically programmed antibodies (cPAbs). cPAbs are bioconjugates in which antibodies are used to augment small molecules with effector functions and prolonged pharmacokinetic profiles, where the pharmacophore of the small molecule is harnessed for target binding and therefore biological targeting. Many different small molecules can be conjugated to large proteins such as full monoclonal antibodies (IgG), fragment crystallizable regions (Fc), or fragment antigen binding regions (Fab). In order to successfully and site-specifically conjugate small molecules to any class of antibodies (IgG, Fc, or Fab), the molecules must be derivatized with a functional group for ease of conjugation without altering the pharmacology of the small molecules. In this Review, we summarize the different synthetic or biological methods that have been employed to produce cPAbs. These unique chemistries have potential to be applied to other fields of antibody modification such as antibody drug conjugates, radioimmunoconjugates, and fluorophore-tagged antibodies.
Collapse
Affiliation(s)
- Omozojie P Aigbogun
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| | - Christopher P Phenix
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| | - Ed S Krol
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, S7N-5E5 Saskatchewan, Canada
| | - Eric W Price
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| |
Collapse
|
27
|
Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. "Smart" drug delivery: A window to future of translational medicine. Front Chem 2023; 10:1095598. [PMID: 36688039 PMCID: PMC9846181 DOI: 10.3389/fchem.2022.1095598] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy is the mainstay of cancer treatment today. Chemotherapeutic drugs are non-selective and can harm both cancer and healthy cells, causing a variety of adverse effects such as lack of specificity, cytotoxicity, short half-life, poor solubility, multidrug resistance, and acquiring cancer stem-like characteristics. There is a paradigm shift in drug delivery systems (DDS) with the advent of smarter ways of targeted cancer treatment. Smart Drug Delivery Systems (SDDSs) are stimuli responsive and can be modified in chemical structure in response to light, pH, redox, magnetic fields, and enzyme degradation can be future of translational medicine. Therefore, SDDSs have the potential to be used as a viable cancer treatment alternative to traditional chemotherapy. This review focuses mostly on stimuli responsive drug delivery, inorganic nanocarriers (Carbon nanotubes, gold nanoparticles, Meso-porous silica nanoparticles, quantum dots etc.), organic nanocarriers (Dendrimers, liposomes, micelles), antibody-drug conjugates (ADC) and small molecule drug conjugates (SMDC) based SDDSs for targeted cancer therapy and strategies of targeted drug delivery systems in cancer cells.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meheli Adhikary
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Praveen Kumar Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Bhudev C. Das
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,*Correspondence: Seema Bhatnagar,
| |
Collapse
|
28
|
Zana A, Galbiati A, Gilardoni E, Bocci M, Millul J, Sturm T, Stucchi R, Elsayed A, Nadal L, Cirillo M, Roll W, Stegger L, Asmus I, Backhaus P, Schäfers M, Neri D, Cazzamalli S. Fibroblast Activation Protein Triggers Release of Drug Payload from Non-internalizing Small Molecule Drug Conjugates in Solid Tumors. Clin Cancer Res 2022; 28:5440-5454. [PMID: 36215129 DOI: 10.1158/1078-0432.ccr-22-1788] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Small molecule drug conjugates (SMDC) are modular anticancer prodrugs that include a tumor-targeting small organic ligand, a cleavable linker, and a potent cytotoxic agent. Most of the SMDC products that have been developed for clinical applications target internalizing tumor-associated antigens on the surface of tumor cells. We have recently described a novel non-internalizing small organic ligand (named OncoFAP) of fibroblast activation protein (FAP), a tumor-associated antigen highly expressed in the stroma of most solid human malignancies. EXPERIMENTAL DESIGN In this article, we describe a new series of OncoFAP-Drug derivatives based on monomethyl auristatin E (MMAE; a potent cytotoxic tubulin poison) and dipeptide linkers that are selectively cleaved by FAP in the tumor microenvironment. RESULTS The tumor-targeting potential of OncoFAP was confirmed in patients with cancer using nuclear medicine procedures. We used mass spectrometry methodologies to quantify the amount of prodrug delivered to tumors and normal organs, as well as the efficiency of the drug release process. Linkers previously exploited for anticancer drug conjugates were used as benchmark. We identified OncoFAP-Gly-Pro-MMAE as the best performing SMDC, which has now been prioritized for further clinical development. OncoFAP-Gly-Pro-MMAE selectively delivered more than 10% injected dose per gram of MMAE to FAP-positive tumors, with a tumor-to-kidney ratio of 16:1 at 24 hours post-injection. CONCLUSIONS The FAP-specific drug conjugates described in this article promise to be efficacious for the targeting of human malignancies. The extracellular release of potent anticancer payloads mediates durable complete remission in difficult-to-treat animal models of cancer.
Collapse
Affiliation(s)
- Aureliano Zana
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Andrea Galbiati
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | | | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Jacopo Millul
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Theo Sturm
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | | | - Abdullah Elsayed
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland
| | - Lisa Nadal
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Martina Cirillo
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna, Italy
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Lars Stegger
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Inga Asmus
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Philipp Backhaus
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Dario Neri
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland.,Philogen S.p.A., Siena, Italy
| | | |
Collapse
|
29
|
Testa C, Papini AM, Zeidler R, Vullo D, Carta F, Supuran CT, Rovero P. First studies on tumor associated carbonic anhydrases IX and XII monoclonal antibodies conjugated to small molecule inhibitors. J Enzyme Inhib Med Chem 2022; 37:592-596. [PMID: 35057692 PMCID: PMC8786240 DOI: 10.1080/14756366.2021.2004593] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022] Open
Abstract
We report for the first time Antibody-Drug-Conjugates (ADCs) containing human (h) Carbonic Anhydrase (CA; EC 4.2.1.1) directed Monoclonal Antibodies (MAbs) linked to low molecular weight inhibitors of the same enzymes by means of hydrophilic peptide spacers. In agreement with the incorporated CA directed MAb fragments, in vitro inhibition data of the obtained ADCs showed sub-nanomolar KI values for the tumour associated CAs IX and XII which were up to 10-fold more potent when compared to the corresponding unconjugated MAbs. In addition, the introduction of the CA inhibitor (CAI) benzenesulfonamide allowed the ADCs to potently inhibit the housekeeping tumoral off-target human CA II isoform. Such results are supporting the definition of an unprecedented reported class of ADCs able to hit simultaneously multiple hCAs physiologically cooperative in maintaining altered cellular metabolic pathways, and therefore ideal for the treatment of chronic diseases such as cancers and inflammation diseases.
Collapse
Affiliation(s)
- Chiara Testa
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology”, Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology”, Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino, Italy
| | - Reinhard Zeidler
- Research Group Therapeutic Antibodies, Helmholtz Centre Munich German Research Centre for Environmental Health, Munich, Germany
- Department of Otorhinolaryngology, Klinikum der Universitaet, Munich, Germany
| | - Daniela Vullo
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, via Ugo Schff 6, Sesto Fiorentino (Florence), 50019Italy
| | - Fabrizio Carta
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, via Ugo Schff 6, Sesto Fiorentino (Florence), 50019Italy
| | - Claudiu T. Supuran
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, via Ugo Schff 6, Sesto Fiorentino (Florence), 50019Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology”, Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
30
|
Amin TU, Emara R, Pal A, Aldawod H, Jiang G, Liang D, Haque Tuhin MT, Balgoname A, Patel AD, Alhamadsheh MM. Enhancing the Safety and Efficacy of PSMA-Based Small-Molecule Drug Conjugates by Linker Stabilization and Conjugation to Transthyretin Binding Ligand. J Med Chem 2022; 65:15473-15486. [PMID: 36327103 DOI: 10.1021/acs.jmedchem.2c01423] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This work describes the enhancement of a novel antitumor therapeutic platform that combines advantages from small-molecule drug conjugates (SMDCs) and antibody drug conjugates (ADCs). Valine-citrulline (VCit) dipeptide linkers are commonly used cathepsin B cleavable linkers for ADCs. However, the instability of these linkers in mouse serum makes translating efficacy data from mouse to human more challenging. Replacing the VCit linker with glutamic acid-valine-citrulline (EVCit) has been reported to enhance the stability of ADCs in mouse serum. However, the effect of EVCit linker on the stability of SMDCs has not been reported. Here, we report that incorporating the EVCit linker in prostate-specific membrane antigen-targeting SMDCs, equipped with the transthyretin ligand AG10, resulted in conjugates with lower toxicity, an extended half-life, and superior therapeutic efficacy to docetaxel in a xenograft mouse model of prostate cancer. This should make SMDCs' preclinical toxicity and efficacy data from mice more reliable for predicting human results.
Collapse
Affiliation(s)
- Toufiq Ul Amin
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Rasha Emara
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Arindom Pal
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Hala Aldawod
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Guanming Jiang
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Dengpan Liang
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Md Tariqul Haque Tuhin
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Abdulmalek Balgoname
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Arjun D Patel
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| | - Mamoun M Alhamadsheh
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, California 95211, United States
| |
Collapse
|
31
|
Lo CF, Chiu TY, Liu YT, Pan PY, Liu KL, Hsu CY, Fang MY, Huang YC, Yeh TK, Hsu TA, Chen CT, Huang LR, Tsou LK. Targeting the Phosphatidylserine-Immune Checkpoint with a Small-Molecule Maytansinoid Conjugate. J Med Chem 2022; 65:12802-12824. [PMID: 36153998 PMCID: PMC9574934 DOI: 10.1021/acs.jmedchem.2c00631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Ligand-targeting drug delivery systems have made significant
strides
for disease treatments with numerous clinical approvals in this era
of precision medicine. Herein, we report a class of small molecule-based
immune checkpoint-targeting maytansinoid conjugates. From the ligand
targeting ability, pharmacokinetics profiling, in vivo anti-pancreatic cancer, triple-negative breast cancer, and sorafenib-resistant
liver cancer efficacies with quantitative mRNA analysis of treated-tumor
tissues, we demonstrated that conjugate 40a not only
induced lasting regression of tumor growth, but it also rejuvenated
the once immunosuppressive tumor microenvironment to an “inflamed
hot tumor” with significant elevation of gene expressions that
were not accessible in the vehicle-treated tumor. In turn, the immune
checkpoint-targeting small molecule drug conjugate from this work
represents a new pharmacodelivery strategy that can be expanded with
combination therapy with existing immune-oncology treatment options.
Collapse
Affiliation(s)
- Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Yu-Tzu Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Pei-Yun Pan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Chia-Yu Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| |
Collapse
|
32
|
In vivo therapeutic effects of small molecule-drug conjugates enhanced by Fc grafting. Biomaterials 2022; 290:121820. [DOI: 10.1016/j.biomaterials.2022.121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
|
33
|
Migliorini F, Cini E, Dreassi E, Finetti F, Ievoli G, Macrì G, Petricci E, Rango E, Trabalzini L, Taddei M. A pH-responsive crosslinker platform for antibody-drug conjugate (ADC) targeting delivery. Chem Commun (Camb) 2022; 58:10532-10535. [PMID: 36043993 DOI: 10.1039/d2cc03052g] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a new 1-6 self-immolative, traceless crosslinker derived from the natural product gallic acid. The linker acts through a pH-dependent mechanism for drug release. This 5-(hydroxymethyl)pyrogallol orthoester derivative (HMPO) was stable for 24 hours at pH values of 7.4 and 6.6 and in plasma, releasing molecules bound to the hydroxymethyl moiety under acid-dependent stimuli at pH 5.5. The linker was non-toxic and was used for the conjugation of Doxorubicin (Doxo) or Combretastatin A4 with Cetuximab. The ADCs formed showed their pH responsivity reducing cell viability of A431 and A549 cancer cells better than Cetuximab alone.
Collapse
Affiliation(s)
- Francesca Migliorini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Cini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Dreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Federica Finetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Giovanni Ievoli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Giulia Macrì
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Petricci
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Enrico Rango
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Lorenza Trabalzini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Maurizio Taddei
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| |
Collapse
|
34
|
Dakhel S, Galbiati A, Migliorini F, Comacchio C, Oehler S, Prati L, Scheuermann J, Cazzamalli S, Neri D, Bassi G, Favalli N. Isolation of a Natural Killer Group 2D Small-Molecule Ligand from DNA-Encoded Chemical Libraries. ChemMedChem 2022; 17:e202200350. [PMID: 35929380 DOI: 10.1002/cmdc.202200350] [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: 06/30/2022] [Revised: 08/04/2022] [Indexed: 11/11/2022]
Abstract
Natural Killer Group 2D (NKG2D) is a homo-dimeric transmembrane protein which is typically expressed on the surface of natural killer (NK) cells, natural killer T (NKT) cells, gamma delta T (γδT) cells, activated CD8 positive T-cells and activated macrophages. Bispecific molecules, capable of bridging NKG2D with a target protein expressed on the surface of tumor cells, may be used to redirect the cytotoxic activity of NK-cells towards antigen-positive malignanT-cells. In this work, we report the discovery of a novel NKG2D small molecule binder [K D = (410±60) nM], isolated from a DNA-Encoded Chemical Library (DEL). The discovery of small organic NKG2D ligands may facilitate the generation of fully synthetic bispecific adaptors, which may serve as an alternative to bispecific antibody products and which may benefit from better tumor targeting properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Luca Prati
- Philogen SpA, R&D (Philochem), SWITZERLAND
| | - Jörg Scheuermann
- ETH Zürich: Eidgenossische Technische Hochschule Zurich, chemistry and applied biosciences, SWITZERLAND
| | | | | | | | - Nicholas Favalli
- Philogen SpA, R&D (Philochem), Libernstrasse 3, 8112, Otelfingen, SWITZERLAND
| |
Collapse
|
35
|
Dannheim FM, Walsh SJ, Orozco CT, Hansen AH, Bargh JD, Jackson SE, Bond NJ, Parker JS, Carroll JS, Spring DR. All-in-one disulfide bridging enables the generation of antibody conjugates with modular cargo loading. Chem Sci 2022; 13:8781-8790. [PMID: 35975158 PMCID: PMC9350601 DOI: 10.1039/d2sc02198f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/10/2022] [Indexed: 12/30/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are valuable therapeutic entities which leverage the specificity of antibodies to selectively deliver cytotoxins to antigen-expressing targets such as cancer cells. However, current methods for their construction still suffer from a number of shortcomings. For instance, using a single modification technology to modulate the drug-to-antibody ratio (DAR) in integer increments while maintaining homogeneity and stability remains exceptionally challenging. Herein, we report a novel method for the generation of antibody conjugates with modular cargo loading from native antibodies. Our approach relies on a new class of disulfide rebridging linkers, which can react with eight cysteine residues, thereby effecting all-in-one bridging of all four interchain disulfides in an IgG1 antibody with a single linker molecule. Modification of the antibody with the linker in a 1 : 1 ratio enabled the modulation of cargo loading in a quick and selective manner through derivatization of the linker with varying numbers of payload attachment handles to allow for attachment of either 1, 2, 3 or 4 payloads (fluorescent dyes or cytotoxins). Assessment of the biological activity of these conjugates demonstrated their exceptional stability in human plasma and utility for cell-selective cytotoxin delivery or imaging/diagnostic applications.
Collapse
Affiliation(s)
| | - Stephen J Walsh
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Cancer Research UK Cambridge Institute, University of Cambridge Cambridge CB2 0RE UK
| | - Carolina T Orozco
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Anders Højgaard Hansen
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Department of Chemistry, Technical University of Denmark (DTU) 2800 Kgs. Lyngby Denmark
| | - Jonathan D Bargh
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Sophie E Jackson
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Nicholas J Bond
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca Granta Park Cambridge CB21 6GH UK
| | - Jeremy S Parker
- Early Chemical Development, Pharmaceutical Development, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge Cambridge CB2 0RE UK
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| |
Collapse
|
36
|
Jin Y, Edalatian Zakeri S, Bahal R, Wiemer AJ. New Technologies Bloom Together for Bettering Cancer Drug Conjugates. Pharmacol Rev 2022; 74:680-711. [PMID: 35710136 PMCID: PMC9553120 DOI: 10.1124/pharmrev.121.000499] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug conjugates, including antibody-drug conjugates, are a step toward realizing Paul Ehrlich's idea from over 100 years ago of a "magic bullet" for cancer treatment. Through balancing selective targeting molecules with highly potent payloads, drug conjugates can target specific tumor microenvironments and kill tumor cells. A drug conjugate consists of three parts: a targeting agent, a linker, and a payload. In some conjugates, monoclonal antibodies act as the targeting agent, but new strategies for targeting include antibody derivatives, peptides, and even small molecules. Linkers are responsible for connecting the payload to the targeting agent. Payloads impact vital cellular processes to kill tumor cells. At present, there are 12 antibody-drug conjugates on the market for different types of cancers. Research on drug conjugates is increasing year by year to solve problems encountered in conjugate design, such as tumor heterogeneity, poor circulation, low drug loading, low tumor uptake, and heterogenous expression of target antigens. This review highlights some important preclinical research on drug conjugates in recent years. We focus on three significant areas: improvement of antibody-drug conjugates, identification of new conjugate targets, and development of new types of drug conjugates, including nanotechnology. We close by highlighting the critical barriers to clinical translation and the open questions going forward. SIGNIFICANCE STATEMENT: The development of anticancer drug conjugates is now focused in three broad areas: improvements to existing antibody drug conjugates, identification of new targets, and development of new conjugate forms. This article focuses on the exciting preclinical studies in these three areas and advances in the technology that improves preclinical development.
Collapse
Affiliation(s)
- Yiming Jin
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | | | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
37
|
Huang KH, Liu YT, Pan PY, Lo CF, Liu KL, Yeh TK, Huang LR, Tsou LK. Rejuvenating hepatic tumor microenvironment immunity with a phosphatidylserine-targeting small molecule drug conjugate. Biomed Pharmacother 2022; 151:113084. [PMID: 35567985 DOI: 10.1016/j.biopha.2022.113084] [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: 03/20/2022] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022] Open
Abstract
We report the design, synthesis and evaluation of a class of phosphatidylserine-targeting zinc (II) dipicolylamine drug conjugates and show that conjugate 16b elicits immune cell infiltration and remodels the "cold" hepatic tumor microenvironment to the inflamed "hot" tumor. Structure-property relationship study via linker modifications and subsequent pharmacokinetics profiling were carried out to improve the solubility and stability of the conjugates in vivo. In a spontaneous hepatocellular carcinoma mouse model, we showed that conjugate 16b exhibited better antitumor efficacy than sorafenib. In particular, significant increase of CD8+ T cell infiltration and granzyme B level was observed, providing insights in sensitizing tumors from intrinsic immune suppressive microenvironment. Evaluation of tumor inflammation-related mRNA expression profile revealed that conjugate 16b, through inductions of key gene expressions including STAT1, CXCL9, CCL5, and PD-L1, rejuvenated tumor microenvironment with enhancement in T cell-, macrophage-, NK cell-, chemokines and cytokines'- functions. Our study establishes that an apoptosis-targeting theranostic enables enrichment of multifaceted immune cells into the tumor mass, which provides potential therapeutic strategies in the combination with immune checkpoint blockade treatment.
Collapse
Affiliation(s)
- Kuan-Hsun Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Yu-Tzu Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Pei-Yun Pan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC.
| | - Lun K Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC.
| |
Collapse
|
38
|
Synthesis and Evaluation of Small Molecule Drug Conjugates Harnessing Thioester-Linked Maytansinoids. Pharmaceutics 2022; 14:pharmaceutics14071316. [PMID: 35890212 PMCID: PMC9323955 DOI: 10.3390/pharmaceutics14071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
Ligand-targeting drug conjugates are a class of clinically validated biopharmaceutical drugs constructed by conjugating cytotoxic drugs with specific disease antigen targeting ligands through appropriate linkers. The integrated linker-drug motif embedded within such a system can prevent the premature release during systemic circulation, thereby allowing the targeting ligand to engage with the disease antigen and selective accumulation. We have designed and synthesized new thioester-linked maytansinoid conjugates. By performing in vitro cytotoxicity, targeting ligand binding assay, and in vivo pharmacokinetic studies, we investigated the utility of this new linker-drug moiety in the small molecule drug conjugate (SMDC) system. In particular, we conjugated the thioester-linked maytansinoids to the phosphatidylserine-targeting small molecule zinc dipicolylamine and showed that Zn8_DM1 induced tumor regression in the HCC1806 triple-negative breast cancer xenograft model. Moreover, in a spontaneous sorafenib-resistant liver cancer model, Zn8_DM1 exhibited potent antitumor growth efficacy. From quantitative mRNA analysis of Zn8_DM1 treated-tumor tissues, we observed the elevation of gene expressions associated with a “hot inflamed tumor” state. With the identification and validation of a plethora of cancer-associated antigens in the “omics” era, this work provided the insight that antibody- or small molecule-based targeting ligands can be conjugated similarly to generate new ligand-targeting drug conjugates.
Collapse
|
39
|
Petrini I, Sollini M, Bartoli F, Barachini S, Montali M, Pardini E, Burzi IS, Erba PA. ED-B-Containing Isoform of Fibronectin in Tumor Microenvironment of Thymomas: A Target for a Theragnostic Approach. Cancers (Basel) 2022; 14:cancers14112592. [PMID: 35681572 PMCID: PMC9179240 DOI: 10.3390/cancers14112592] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The extra-domain B fibronectin (ED-B FN) is highly expressed in thymic epithelial tumors (TETs), as demonstrated by in vivo targeting using 131I-labeled L19 small immunoprotein (131I-L19-SIP) and immunohistochemistry with a predominant expression by stromal cells of a thymoma microenvironment rather than epithelial cells. Such high expression derived from the induction of stromal cells shifts FN production to the ED-B subtype. Our results suggest that Radretumab radioimmunotherapy (R-RIT) inefficacy is not related to low TET ED-B expression but to multifactorial aspects including patients’ inherent characteristics, the pattern expression of the target, the biological characteristics of the tumor, and the format of the target agent, which contribute to the resistance of tumor cells to treatment. Abstract Aim: to exploit tissue-specific interactions among thymic epithelial tumor (TETs) cells and extra-domain B fibronectin (ED-B FN). Material and methods: The stromal pattern of ED-B FN expression was investigated through tumor specimen collection and molecular profiling in 11 patients with recurrent TETs enrolled in prospective theragnostic phase I/II trials with Radretumab, an ED-B FN specific recombinant human antibody. Radretumab radioimmunotherapy (R-RIT) was offered to patients who exhibited the target expression. Experiments included immunochemical analysis (ICH), cell cultures, immunophenotypic analysis, Western blot, slot-blot assay, and quantitative RT-PCR of two primary thymoma cultures we obtained from patients’ samples and in the Ty82 cell line. Results: The in vivo scintigraphic demonstration of ED-B FN expression resulted in R-RIT eligibility in 8/11 patients, of which seven were treated. The best observed response was disease stabilization (n = 5/7) with a duration of 4.3 months (range 3–5 months). IHC data confirmed high ED-B FN expression in the peripherical microenvironment rather than in the center of the tumor, which was more abundant in B3 thymomas. Further, there was a predominant expression of ED-B FN by the stromal cells of the thymoma microenvironment rather than the epithelial cells. Conclusions: Our data support the hypothesis that thymomas induce stromal cells to shift FN production to the ED-B subtype, likely representing a favorable hallmark for tumor progression and metastasis. Collectively, results derived from clinical experience and molecular insights of the in vitro experiments suggested that R-RIT inefficacy is unlikely related to low target expression in TET, being the mechanism of R-RIT resistance eventually related to patients’ susceptibility (i.e., inherent characteristics), the pattern expression of the target (i.e., at periphery), the biological characteristics of the tumor (i.e., aggressive and resistant phenotypes), and/or to format of the target agent (i.e., 131I-L19-SIP).
Collapse
Affiliation(s)
- Iacopo Petrini
- General Pathology, Department of Translational Research & New Technologies in Surgery and Medicine, University of Pisa and Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy;
| | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy;
- Diagnostic Imaging Department, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Francesco Bartoli
- Regional Center of Nuclear Medicine, Department of Translational Research and New Technology in Medicine, University of Pisa and Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
| | - Serena Barachini
- Laboratory of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.B.); (M.M.); (E.P.); (I.S.B.)
| | - Marina Montali
- Laboratory of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.B.); (M.M.); (E.P.); (I.S.B.)
| | - Eleonora Pardini
- Laboratory of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.B.); (M.M.); (E.P.); (I.S.B.)
| | - Irene Sofia Burzi
- Laboratory of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.B.); (M.M.); (E.P.); (I.S.B.)
| | - Paola Anna Erba
- Regional Center of Nuclear Medicine, Department of Translational Research and New Technology in Medicine, University of Pisa and Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Centre, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: ; Tel.: +39-050-992115
| |
Collapse
|
40
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
41
|
Jin C, EI‐Sagheer AH, Li S, Vallis KA, Tan W, Brown T. Engineering Enzyme-Cleavable Oligonucleotides by Automated Solid-Phase Incorporation of Cathepsin B Sensitive Dipeptide Linkers. Angew Chem Int Ed Engl 2022; 61:e202114016. [PMID: 34953094 PMCID: PMC9306542 DOI: 10.1002/anie.202114016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Indexed: 12/04/2022]
Abstract
Oligonucleotides containing cleavable linkers have emerged as versatile tools to achieve stimulus-responsive and site-specific cleavage of DNA. However, the limitations of previously reported cleavable linkers including photolabile and disulfide linkers have restricted their applications in vivo. Inspired by the cathepsin B-sensitive dipeptide linkers in antibody-drug conjugates (ADCs) such as Adcetris, we have developed Val-Ala-02 and Val-Ala-Chalcone phosphoramidites for the automated synthesis of enzyme-cleavable oligonucleotides. Cathepsin B digests Val-Ala-02 and Val-Ala-Chalcone linkers efficiently, enabling cleavage of oligonucleotides into two components or release of small-molecule payloads. Based on the prior success of dipeptide linkers in ADCs, we believe that these dipeptide linker phosphoramidites will promote new clinical applications of therapeutic oligonucleotides.
Collapse
Affiliation(s)
- Cheng Jin
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| | - Afaf H. EI‐Sagheer
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
- Department of Science and MathematicsSuez University, Faculty of Petroleum and Mining EngineeringSuez43721Egypt
| | - Siqi Li
- Medical Research CouncilOxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Katherine A. Vallis
- Medical Research CouncilOxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Weihong Tan
- The Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer Hospital)Institute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouZhejiang310022China
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Tom Brown
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| |
Collapse
|
42
|
Jin C, EI‐Sagheer AH, Li S, Vallis KA, Tan W, Brown T. Engineering Enzyme-Cleavable Oligonucleotides by Automated Solid-Phase Incorporation of Cathepsin B Sensitive Dipeptide Linkers. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202114016. [PMID: 38505643 PMCID: PMC10946720 DOI: 10.1002/ange.202114016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Indexed: 11/09/2022]
Abstract
Oligonucleotides containing cleavable linkers have emerged as versatile tools to achieve stimulus-responsive and site-specific cleavage of DNA. However, the limitations of previously reported cleavable linkers including photolabile and disulfide linkers have restricted their applications in vivo. Inspired by the cathepsin B-sensitive dipeptide linkers in antibody-drug conjugates (ADCs) such as Adcetris, we have developed Val-Ala-02 and Val-Ala-Chalcone phosphoramidites for the automated synthesis of enzyme-cleavable oligonucleotides. Cathepsin B digests Val-Ala-02 and Val-Ala-Chalcone linkers efficiently, enabling cleavage of oligonucleotides into two components or release of small-molecule payloads. Based on the prior success of dipeptide linkers in ADCs, we believe that these dipeptide linker phosphoramidites will promote new clinical applications of therapeutic oligonucleotides.
Collapse
Affiliation(s)
- Cheng Jin
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| | - Afaf H. EI‐Sagheer
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
- Department of Science and MathematicsSuez University, Faculty of Petroleum and Mining EngineeringSuez43721Egypt
| | - Siqi Li
- Medical Research CouncilOxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Katherine A. Vallis
- Medical Research CouncilOxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Weihong Tan
- The Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer Hospital)Institute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouZhejiang310022China
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Tom Brown
- Department of Chemistry, Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| |
Collapse
|
43
|
Harms M, Hansson RF, Carmali S, Almeida-Hernández Y, Sanchez-Garcia E, Münch J, Zelikin AN. Dimerization of the Peptide CXCR4-Antagonist on Macromolecular and Supramolecular Protraction Arms Affords Increased Potency and Enhanced Plasma Stability. Bioconjug Chem 2022; 33:594-607. [PMID: 35293739 DOI: 10.1021/acs.bioconjchem.2c00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peptides are prime drug candidates due to their high specificity of action but are disadvantaged by low proteolytic stability. Here, we focus on the development of stabilized analogues of EPI-X4, an endogenous peptide antagonist of CXCR4. We synthesized macromolecular peptide conjugates and performed side-by-side comparison with their albumin-binding counterparts and considered monovalent conjugates, divalent telechelic conjugates, and Y-shaped peptide dimers. All constructs were tested for competition with the CXCR4 antibody-receptor engagement, inhibition of receptor activation, and inhibition of the CXCR4-tropic human immunodeficiency virus infection. We found that the Y-shaped conjugates were more potent than the parent peptide and at the same time more stable in human plasma, with a favorable outlook for translational studies.
Collapse
Affiliation(s)
- Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rikke Fabech Hansson
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Sheiliza Carmali
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Yasser Almeida-Hernández
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Alexander N Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
44
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
45
|
Tsai CH, Chiu TY, Chen CT, Hsu CY, Tsai YR, Yeh TK, Huang KH, Tsou LK. Click Chemistry and Multicomponent Reaction for Linker Diversification of Zinc Dipicolylamine-Based Drug Conjugates. Front Chem 2022; 9:822587. [PMID: 35242746 PMCID: PMC8886374 DOI: 10.3389/fchem.2021.822587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
An efficient Ugi multicomponent reaction with strain promoted azide-alkyne cycloaddition protocol has been utilized in concert or independently to prepare a small family of bioactive zinc(II) dipicolylamine (ZnDPA)-based SN-38 conjugates. With sequential click chemistry coupling between the cytotoxic payload and phosphatidylserine-targeting ZnDPA ligand derived from structurally diverse carboxylic acids, aldehyde or ketones, and isocyanides, we demonstrated that this convergent synthetic strategy could furnish conjugates harnessing diversified linkers that exhibited different pharmacokinetic profiles in systemic circulation in vivo. Among the eight new conjugates, comparative studies on in vitro cytotoxicities, plasma stabilities, in vivo pharmacokinetic properties, and maximum tolerated doses were then carried out to identify a potent ZnDPA-based SN-38 conjugate that resulted in pancreatic cancer growth regression with an 80% reduction of cytotoxic payload used when compared to that of the marketed irinotecan. Our work provided the roadmap to construct a variety of theranostic agents in a similar manner for cancer treatment.
Collapse
|
46
|
Younis MA, Tawfeek HM, Abdellatif AAH, Abdel-Aleem JA, Harashima H. Clinical translation of nanomedicines: Challenges, opportunities, and keys. Adv Drug Deliv Rev 2022; 181:114083. [PMID: 34929251 DOI: 10.1016/j.addr.2021.114083] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023]
Abstract
Despite the massive interest and recent developments in the field of nanomedicine, only a limited number of formulations have found their way to the clinics. This shortcoming reveals the challenges facing the clinical translation of this technology. In the current article, we summarize and evaluate the status, market situation, and clinical profiles of the reported nanomedicines, the shortcomings limiting their clinical translation, as well as some approaches designed to break through this barrier. Moreover, some emerging technologies that have the potential to compete with nanomedicines are highlighted. Lastly, we identify the key factors that should be considered in nanomedicine-related research to be clinically-translatable. These can be classified into five areas: rational design during the research and development stage, the recruitment of representative preclinical models, careful design of clinical trials, development of specific and uniform regulatory protocols, and calls for non-classic sponsorship. This new field of endeavor was firmly established during the last two decades and more in-depth progress is expected in the coming years.
Collapse
Affiliation(s)
- Mahmoud A Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Hesham M Tawfeek
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Jelan A Abdel-Aleem
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
47
|
Veselov VV, Nosyrev AE, Jicsinszky L, Alyautdin RN, Cravotto G. Targeted Delivery Methods for Anticancer Drugs. Cancers (Basel) 2022; 14:622. [PMID: 35158888 PMCID: PMC8833699 DOI: 10.3390/cancers14030622] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Several drug-delivery systems have been reported on and often successfully applied in cancer therapy. Cell-targeted delivery can reduce the overall toxicity of cytotoxic drugs and increase their effectiveness and selectivity. Besides traditional liposomal and micellar formulations, various nanocarrier systems have recently become the focus of developmental interest. This review discusses the preparation and targeting techniques as well as the properties of several liposome-, micelle-, solid-lipid nanoparticle-, dendrimer-, gold-, and magnetic-nanoparticle-based delivery systems. Approaches for targeted drug delivery and systems for drug release under a range of stimuli are also discussed.
Collapse
Affiliation(s)
- Valery V. Veselov
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - Alexander E. Nosyrev
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - László Jicsinszky
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
| | - Renad N. Alyautdin
- Department of Pharmacology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia
| |
Collapse
|
48
|
Li SS, Zhang CM, Wu JD, Liu C, Liu ZP. A branched small molecule-drug conjugate nanomedicine strategy for the targeted HCC chemotherapy. Eur J Med Chem 2022; 228:114037. [PMID: 34883290 DOI: 10.1016/j.ejmech.2021.114037] [Citation(s) in RCA: 1] [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: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Off-target toxicity is one of the main challenges faced by anticancer chemotherapeutics. For tumor targeted and precision chemotherapy, we take the advantages of the ligand directed tumor active targeting of small molecule drug conjugates (SMDCs) and the passive tumor targeting of nanoparticles via the enhanced penetration and retention (EPR) effects, put forward a branched small molecule drug conjugate (BSMDC) nanomedicine design concept. In a proof of concept, we used pentaerythritol as the branched moiety, galactosamine (GalN) as the hepatocellular carcinoma (HCC) directing ligands, PTX as a payload, and a stearoyl moiety as the amphiphilic property adjusting group, designed and synthesized BSMDC 1 and prepared its NPs. In cellular level, the BSMDC 1 NPs targeted asialoglycoprotein receptor (ASGPR)-overexpressing HepG2 cells, were effectively taken up in the cells and released in tumor microenvironments, inhibited the HepG2 cell proliferation, arrested HepG2 cell in G2/M phase and induced tumor cell apoptosis. In HepG2 xenograft nude mice, the BSMDC 1 NPs were high specific to target the tumor and demonstrated a higher antitumor efficiency than BSMDC 1, having no apparent influences on mice body weights and major organs, supporting our BSMDC nanomedicine design concept. Therefore, this new strategy may find applications for cancer targeted and precision chemotherapy.
Collapse
Affiliation(s)
- Sha-Sha Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Cheng-Mei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
49
|
Lerchen HG, Stelte-Ludwig B, Kopitz C, Heroult M, Zubov D, Willuda J, Schlange T, Kahnert A, Wong H, Izumi R, Hamdy A. A Small Molecule–Drug Conjugate (SMDC) Consisting of a Modified Camptothecin Payload Linked to an αVß3 Binder for the Treatment of Multiple Cancer Types. Cancers (Basel) 2022; 14:cancers14020391. [PMID: 35053556 PMCID: PMC8773721 DOI: 10.3390/cancers14020391] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/07/2021] [Accepted: 01/04/2022] [Indexed: 12/27/2022] Open
Abstract
To improve tumor selectivity of cytotoxic agents, we designed VIP236, a small molecule–drug conjugate consisting of an αVβ3 integrin binder linked to a modified camptothecin payload (VIP126), which is released by the enzyme neutrophil elastase (NE) in the tumor microenvironment (TME). The tumor targeting and pharmacokinetics of VIP236 were studied in tumor-bearing mice by in vivo near-infrared imaging and by analyzing tumor and plasma samples. The efficacy of VIP236 was investigated in a panel of cancer cell lines in vitro, and in MX-1, NCI-H69, and SW480 murine xenograft models. Imaging studies with the αVβ3 binder demonstrated efficient tumor targeting. Administration of VIP126 via VIP236 resulted in a 10-fold improvement in the tumor/plasma ratio of VIP126 compared with VIP126 administered alone. Unlike SN38, VIP126 is not a substrate of P-gp and BCRP drug transporters. VIP236 presented strong cytotoxic activity in the presence of NE. VIP236 treatment resulted in tumor regressions and very good tolerability in all in vivo models tested. VIP236 represents a novel approach for delivering a potent cytotoxic agent by utilizing αVβ3 as a targeting moiety and NE in the TME to release the VIP126 payload—designed for high permeability and low efflux—directly into the tumor stroma.
Collapse
Affiliation(s)
- Hans-Georg Lerchen
- Vincerx Pharma GmbH, 40789 Monheim am Rhein, Germany;
- Correspondence: ; Tel.: +49-157-31993091
| | | | | | - Melanie Heroult
- Crop Science Division, Bayer AG, 65926 Frankfurt am Main, Germany;
| | - Dmitry Zubov
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Joerg Willuda
- Pharmaceuticals R&D, Bayer AG, 13353 Berlin, Germany;
| | - Thomas Schlange
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Antje Kahnert
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Harvey Wong
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| | - Raquel Izumi
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| | - Ahmed Hamdy
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| |
Collapse
|
50
|
Plais L, Scheuermann J. Macrocyclic DNA-encoded chemical libraries: a historical perspective. RSC Chem Biol 2022; 3:7-17. [PMID: 35128404 PMCID: PMC8729180 DOI: 10.1039/d1cb00161b] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/19/2021] [Indexed: 12/25/2022] Open
Abstract
While macrocyclic peptides are extensively researched for therapeutically relevant protein targets, DNA-encoded chemical libraries (DELs) are developed at a quick pace to discover novel small molecule binders. The combination of both fields has been explored since 2004 and the number of macrocyclic peptide DELs is steadily increasing. Macrocycles with high affinity and potency were identified for diverse classes of proteins, revealing DEL's huge potential. By giving a historical perspective, we would like to review the methods which permitted the rise of macrocyclic peptide DELs, describe the different DELs which were created and discuss the achievements and challenges of this emerging field.
Collapse
Affiliation(s)
- Louise Plais
- Department of Chemistry and Applied Biosciences, ETH Zürich (Swiss Federal Institute of Technology) Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| | - Jörg Scheuermann
- Department of Chemistry and Applied Biosciences, ETH Zürich (Swiss Federal Institute of Technology) Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| |
Collapse
|