1
|
Jamadade P, Nupur N, Maharana KC, Singh S. Therapeutic Monoclonal Antibodies for Metabolic Disorders: Major Advancements and Future Perspectives. Curr Atheroscler Rep 2024; 26:549-571. [PMID: 39008202 DOI: 10.1007/s11883-024-01228-0] [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] [Accepted: 06/28/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE OF REVIEW Globally, the prevalence of metabolic disorders is rising. Elevated low-density lipoprotein (LDL) cholesterol is a hallmark of familial hypercholesterolemia, one of the most prevalent hereditary metabolic disorders and another one is Diabetes mellitus (DM) that is more common globally, characterised by hyperglycemia with low insulin-directed glucose by target cells. It is still known that low-density lipoprotein cholesterol (LDL-C) increases the risk of cardiovascular disease (CVD). LDL-C levels are thought to be the main therapeutic objectives. RECENT FINDINGS The primary therapy for individuals with elevated cholesterol levels is the use of statins and other lipid lowering drugs like ezetimibe for hypercholesterolemia. Even after taking statin medication to the maximum extent possible, some individuals still have a sizable residual cardiovascular risk. To overcome this proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors-monoclonal antibodies (mAbs) are a novel class of systemic macromolecules that have enhanced LDL-C-lowering efficacy. Along with this other inhibitor are used like Angiopoeitin like 3 inhibitors. Research on both humans and animals has shown that anti-CD3 antibodies can correct autoimmune disorders like diabetes mellitus. Individuals diagnosed with familial hypercholesterolemia (FH) may need additional treatment options beyond statins, especially when facing challenges such as statin tolerance or the inability of even the highest statin doses to reach the desired target cholesterol level. Here is the summary of PCSK9, ANGPTL-3 and CD3 inhibitors and their detailed information. In this review we discuss the details of PCSK9, ANGPTL-3 and CD3 inhibitors and the current therapeutic interventions of using the monoclonal antibodies in case of the metabolic disorder. We further present the present studies and the future prospective of the same.
Collapse
Affiliation(s)
- Pratiksha Jamadade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Vaishali, Hajipur, 844102, Bihar, India
| | - Neh Nupur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Vaishali, Hajipur, 844102, Bihar, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Vaishali, Hajipur, 844102, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Vaishali, Hajipur, 844102, Bihar, India.
| |
Collapse
|
2
|
Kaplon H, Crescioli S, Chenoweth A, Visweswaraiah J, Reichert JM. Antibodies to watch in 2023. MAbs 2023; 15:2153410. [PMID: 36472472 PMCID: PMC9728470 DOI: 10.1080/19420862.2022.2153410] [Citation(s) in RCA: 187] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022] Open
Abstract
In this 14th installment of the annual Antibodies to Watch article series, we discuss key events in commercial monoclonal antibody therapeutics development that occurred in 2022 and forecast events that might occur in 2023. As of mid-November, 12 antibody therapeutics had been granted first approvals in either the United States or European Union (tebentafusp (Kimmtrak), faricimab (Vabysmo), sutimlimab (Enjaymo), relatlimab (Opdualag), tixagevimab/cilgavimab (Evusheld), mosunetuzumab (Lunsumio), teclistamab (TECVAYLI), spesolimab (SPEVIGO), tremelimumab (Imjudo; combo with durvalumab), nirsevimab (Beyfortus), mirvetuximab soravtansine (ELAHERE™), and teplizumab (TZIELD)), including 4 bispecific antibodies and 1 ADC. Based on FDA action dates, several additional product candidates could be approved by the end of 2022. An additional seven were first approved in China or Japan in 2022, including two bispecific antibodies (cadonilimab and ozoralizumab). Globally, at least 24 investigational antibody therapeutics are undergoing review by regulatory agencies as of mid-November 2022. Our data show that, with antibodies for COVID-19 excluded, the late-stage commercial clinical pipeline grew by ~20% in the past year to include nearly 140 investigational antibody therapeutics that were designed using a wide variety of formats and engineering techniques. Of those in late-stage development, marketing application submissions for at least 23 may occur by the end of 2023, of which 5 are bispecific (odronextamab, erfonrilimab, linvoseltamab, zanidatamab, and talquetamab) and 2 are ADCs (datopotamab deruxtecan, and tusamitamab ravtansine).
Collapse
Affiliation(s)
- Hélène Kaplon
- Translational Medicine Department, Institut de Recherches Internationales ServierSuresnes, France
| | - Silvia Crescioli
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, LondonUK
| | - Alicia Chenoweth
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, LondonUK
| | | | | |
Collapse
|
3
|
Schwarting R, Behling E, Allen A, Arguello-Guerra V, Budak-Alpdogan T. CD30+ Lymphoproliferative Disorders as Potential Candidates for CD30-Targeted Therapies. Arch Pathol Lab Med 2022; 146:415-432. [PMID: 35299246 DOI: 10.5858/arpa.2021-0338-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— In the early 1980s, a monoclonal antibody termed Ki-1 was developed against a cell line derived from a patient with Hodgkin lymphoma. This antibody detected a limited number of benign activated lymphocytes in lymphoid tissue, whereas in Hodgkin lymphoma it appeared to be nearly specific for Reed-Sternberg cells and their mononuclear variants. Subsequent studies showed that Ki-1 expression defined a new type of lymphoma that was later designated anaplastic large cell lymphoma with or without anaplastic large cell kinase expression/translocation. In the past 30 years, numerous new lymphoma entities have been defined, many of which are variably positive for CD30. Many virally transformed lymphoproliferative disorders are also frequently positive for CD30. OBJECTIVE.— To illustrate the broad spectrum of CD30+ hematologic malignancies and to provide an update of CD30-targeted therapies. DATA SOURCES.— Personal experiences and published works in PubMed. CONCLUSIONS.— Because of its low expression in normal tissue, CD30 was studied as a therapeutic target for many years. However, the first functional humanized antibody against CD30 was developed only about 10 years ago. Brentuximab vedotin is a humanized anti-CD30 antibody linked to a cytotoxin, and was approved by the US Food and Drug Administration in 2012 for treating refractory Hodgkin lymphoma and anaplastic large cell lymphoma. Since then, the list of Food and Drug Administration-approved CD30-targeted hematologic malignancies has grown. Recently, the therapies using tumor antigen-specific chimeric antigen receptor T cells targeting CD30 have incited a great deal of enthusiasm and are studied in clinical trials.
Collapse
Affiliation(s)
- Roland Schwarting
- From the Department of Pathology, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey (Schwarting, Behling, Allen, Arguello-Guerra)
| | - Eric Behling
- From the Department of Pathology, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey (Schwarting, Behling, Allen, Arguello-Guerra)
| | - Ashleigh Allen
- From the Department of Pathology, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey (Schwarting, Behling, Allen, Arguello-Guerra)
| | - Vivian Arguello-Guerra
- From the Department of Pathology, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey (Schwarting, Behling, Allen, Arguello-Guerra)
| | - Tulin Budak-Alpdogan
- MD Anderson Cancer Center at Cooper, Department of Medicine, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, New Jersey (Budak-Alpdogan)
| |
Collapse
|
4
|
Ma X, Shi L, Zhang B, Liu L, Fu Y, Zhang X. Recent advances in bioprobes and biolabels based on cyanine dyes. Anal Bioanal Chem 2022; 414:4551-4573. [PMID: 35359180 DOI: 10.1007/s00216-022-03995-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/19/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
As a functional dye, cyanine dye promotes the widespread application of bioprobes in the fields of medicine, genetics and environment, owing to its advantages of good photophysical properties, excellent biocompatibility and low toxicity to biological systems. Nowadays, it is mainly used in the fields of life sciences such as fluorescent labeling of biological macromolecules, disease diagnosis, immunoassay and DNA detection, all of which lie at the core of this review. First, we briefly introduced the characteristics and principles of the cyanine dye bioprobe. Afterward, we paid attention to the recent progress of cyanine dye bioprobes widely used in the 10 years from 2010 to 2020. The application of cyanine dyes as bioprobes with different identification elements, including enzymes, organelles, immunity and DNAs, was mainly summarized. Finally, this review gave an outlook on the future development trend of cyanine dye bioprobes. This facilitates the construction of a new type of multifunctional fluorescent probe and promotes its clinical application.
Collapse
Affiliation(s)
- Xiaoying Ma
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China
| | - Lei Shi
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China.
| | - Buyue Zhang
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China
| | - Lu Liu
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China
| | - Yao Fu
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China
| | - Xiufeng Zhang
- College of Chemical Engineering, Hebei and Tangshan Key Laboratory of Medical-Industrial Integration Precision Medicine, North China University of Science and Technology, 063210, Tangshan, China.
| |
Collapse
|
5
|
Naik J, Kulkarni D, Kadu P, Pandya A, Kale P. Use of In silico tools for screening buffers to overcome physical instability of Abatacept. Transpl Immunol 2022; 71:101551. [PMID: 35122959 DOI: 10.1016/j.trim.2022.101551] [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: 08/31/2021] [Revised: 12/30/2021] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
Abstract
Rheumatoid arthritis is an autoimmune disorder. Abatacept (CTLA4-Ig) is used for the treatment of Rheumatoid arthritis. Abatacept is a monoclonal antibody. Monoclonal antibodies undergo chemical (e.g. oxidation, deamidation, hydrolysis) and physical (e.g. aggregation, unfolding) instabilities while handling and storage. Abatacept is also prone to aggregation. Stabilizing agents such as buffers are used to stabilize monoclonal antibodies. But, the selection of the appropriate buffer is a time-consuming process because after testing many buffers based on the analysis of the results the appropriate buffer is identified. To overcome this issue in the current study computational tools were utilized to virtually screen different buffers to select the appropriate buffer. Ligand binding is the principal mechanism of conformational stability of proteins. For the buffers as well ligand binding is the most common mechanism for enhancing the thermodynamic stability of proteins. Generally it is observed that by enhancing the thermodynamic stability there is reduction in the rate of aggregation of proteins. Buffer (ligand) binds to the native state of the protein preferentially; it results in stabilization of the protein, while in the case of denatured protein it has no impact. There are many studies conducted involving the proteins in buffer solutions but very limited information is available about the mechanism of protein-buffer interactions. In the current study ligand binding mechanism of protein - buffer interaction was studied using molecular docking. After the docking buffers were ranked according to their energy value. The lower energy scores represent better protein-buffer (ligand) binding affinity compared to high energy values. It was observed that Phosphate with a binding affinity of -107.9 kcal/mol was the buffer with the least binding energy followed by Citrate (-70.6 kcal/mol), Melglumine (-66.6 kcal/mol), Arginine (-64.5 kcal/mol), Glucono delta lactone (-62.6 kcal/mol), Sodium citrate (-56.5 kcal/mol), Tromethamine (-52.3 kcal/mol), Glycine HCl (-37.2 kcal/mol), Sulfuric acid (-37.7 kcal/mol), Ammonium acetate (-31.1 kcal/mol), Acetic acid (-30.7 kcal/mol). With lower binding energy higher is the affinity between the ligand and protein. So phosphate was identified as a buffer with the highest affinity with Abatacept.
Collapse
Affiliation(s)
- Janhavi Naik
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Duttraj Kulkarni
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Pramod Kadu
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India.
| | - Aditya Pandya
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Pravin Kale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| |
Collapse
|
6
|
Abstract
This 2020 installment of the annual 'Antibodies to Watch' series documents the antibody therapeutics approved in 2019 and in regulatory review in the United States or European Union, as well as those in late-stage clinical studies, as of November 2019*. At this time, a total of 5 novel antibody therapeutics (romosozumab, risankizumab, polatuzumab vedotin, brolucizumab, and crizanlizumab) had been granted a first approval in either the US or EU, and marketing applications for 13 novel antibody therapeutics (eptinezumab, teprotumumab, enfortumab vedotin, isatuximab, [fam-]trastuzumab deruxtecan, inebilizumab, leronlimab, sacituzumab govitecan, satralizumab, narsoplimab, tafasitamab, REGNEB3 and naxituximab) were undergoing review in these regions, which represent the major markets for antibody therapeutics. Also as of November 2019, 79 novel antibodies were undergoing evaluation in late-stage clinical studies. Of the 79 antibodies, 39 were undergoing evaluation in late-stage studies for non-cancer indications, with 2 of these (ublituximab, pamrevlumab) also in late-stage studies for cancer indications. Companies developing 7 (tanezumab, aducanumab, evinacumab, etrolizumab, sutimlimab, anifrolumab, and teplizumab) of the 39 drugs have indicated that they may submit a marketing application in either the US or EU in 2020. Of the 79 antibodies in late-stage studies, 40 were undergoing evaluation as treatments for cancer, and potentially 9 of these (belantamab mafodotin, oportuzumab monatox, margetuximab, dostarlimab, spartalizumab, 131I-omburtamab, loncastuximab tesirine, balstilimab, and zalifrelimab) may enter regulatory review in late 2019 or in 2020. Overall, the biopharmaceutical industry's clinical pipeline of antibody therapeutics is robust, and should provide a continuous supply of innovative products for patients in the future. *Note on key updates through December 18, 2019: 1) the US Food and Drug Administration granted accelerated approval to enfortumab vedotin-ejfv (Padcev) on December 18, 2019, bringing the total number of novel antibody therapeutics granted a first approval in either the US or EU during 2019 to 6; 2) the European Commission approved romosozumab on December 9, 2019; 3) the European Medicines Agency issued a positive opinion for brolucizumab; 4) Sesen Bio initiated a rolling biologics license application (BLA) on December 6, 2019; 5) GlaxoSmithKline submitted a BLA for belantamab mafodotin; and 6) the status of the Phase 3 study (NCT04128696) of GSK3359609, a humanized IgG4 anti-ICOS antibody, in patients with head and neck squamous cell carcinoma was updated to recruiting from not yet recruiting.
Collapse
Affiliation(s)
- Hélène Kaplon
- Division of Biotechnology & Biomarker Research, Institut de Recherches Servier, Croissy-sur-Seine, France
| | | | | | | |
Collapse
|
7
|
Abstract
In this 12th annual installment of the Antibodies to Watch article series, we discuss key events in antibody therapeutics development that occurred in 2020 and forecast events that might occur in 2021. The coronavirus disease 2019 (COVID-19) pandemic posed an array of challenges and opportunities to the healthcare system in 2020, and it will continue to do so in 2021. Remarkably, by late November 2020, two anti-SARS-CoV antibody products, bamlanivimab and the casirivimab and imdevimab cocktail, were authorized for emergency use by the US Food and Drug Administration (FDA) and the repurposed antibodies levilimab and itolizumab had been registered for emergency use as treatments for COVID-19 in Russia and India, respectively. Despite the pandemic, 10 antibody therapeutics had been granted the first approval in the US or EU in 2020, as of November, and 2 more (tanezumab and margetuximab) may be granted approvals in December 2020.* In addition, prolgolimab and olokizumab had been granted first approvals in Russia and cetuximab saratolacan sodium was first approved in Japan. The number of approvals in 2021 may set a record, as marketing applications for 16 investigational antibody therapeutics are already undergoing regulatory review by either the FDA or the European Medicines Agency. Of these 16 mAbs, 11 are possible treatments for non-cancer indications and 5 are potential treatments for cancer. Based on the information publicly available as of November 2020, 44 antibody therapeutics are in late-stage clinical studies for non-cancer indications, including 6 for COVID-19, and marketing applications for at least 6 (leronlimab, tezepelumab, faricimab, ligelizumab, garetosmab, and fasinumab) are planned in 2021. In addition, 44 antibody therapeutics are in late-stage clinical studies for cancer indications. Of these 44, marketing application submissions for 13 may be submitted by the end of 2021. *Note added in proof on key events announced during December 1-21, 2020: margetuximab-cmkb and ansuvimab-zykl were approved by FDA on December 16 and 21, 2020, respectively; biologics license applications were submitted for ublituximab and amivantamab.
Collapse
Affiliation(s)
- Hélène Kaplon
- Institut De Recherches Internationales Servier, Translational Medicine Department, Suresnes, France
| | | |
Collapse
|
8
|
Tandale JB, Badgujar SB, Tandale BU, Angre U, Daftary SB, Lala S, Gaur VP. An improved protocol for large scale production of high purity 'Fc' fragment of human immunoglobulin G (IgG-Fc). J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1159:122400. [PMID: 33126073 DOI: 10.1016/j.jchromb.2020.122400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/30/2020] [Accepted: 09/29/2020] [Indexed: 01/09/2023]
Abstract
We describe a simplified approach for purification and characterization of human 'IgG-Fc' fragment used widely as immunochemical tool and for therapeutic purposes. The 'Fc' fragment was purified from human IgG in a 3-stage column chromatography. The purified 'Fc' fragment appeared as a dimer glycoprotein with an apparent molecular mass of 52,981 Dalton (Ultraflex MALDI TOF/TOF). The Size-exclusion HPLC profile of the purified 'Fc' fragment of human IgG matched that of a commercially procured reference 'Fc' fragment material. The purity of the 'Fc' fragments was >99% by SDS-PAGE and size-exclusion HPLC. The results of Western blotting, immunoelectrophoresis, and mass spectrometry analysis indicate a high purity of the 'Fc' fragment. Peptide mass fingerprint analysis of the purified 'Fc' protein yielded peptides that partially match the known database sequences of FCG3B_HUMAN (Uniprot ID: O75015). This method of purification of the 'Fc' fragment is suitable for achieving high purity level of 'Fc' fragment protein. With this purification approach, the cost of the purified 'Fc' fragment of human IgG is significantly reduced as compared with the current market price of IgG-Fc fragment protein in international market. The purified 'IgG-Fc' fragment protein was found to be negative for major viral markers.
Collapse
Affiliation(s)
- Jatin B Tandale
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India
| | - Shamkant B Badgujar
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India.
| | - Babasaheb U Tandale
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India
| | - Unmesh Angre
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India
| | - Siddharth B Daftary
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India
| | - Sanjeev Lala
- Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane 400604, Maharashtra, India
| | - Vinod P Gaur
- Northwest Lipid Metabolism and Diabetes Research Laboratories (NWRL), Department of Medicine, University of Washington, 401 Queen Anne Ave North, Seattle, WA 98109, USA
| |
Collapse
|
9
|
McDaid WJ, Greene MK, Johnston MC, Pollheimer E, Smyth P, McLaughlin K, Van Schaeybroeck S, Straubinger RM, Longley DB, Scott CJ. Repurposing of Cetuximab in antibody-directed chemotherapy-loaded nanoparticles in EGFR therapy-resistant pancreatic tumours. NANOSCALE 2019; 11:20261-20273. [PMID: 31626255 PMCID: PMC6861736 DOI: 10.1039/c9nr07257h] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The anti-Epidermal Growth Factor Receptor (EGFR) antibody Cetuximab (CTX) has demonstrated limited anti-cancer efficacy in cells overexpressing EGFR due to activating mutations in RAS in solid tumours, such as pancreatic cancer. The utilisation of antibodies as targeting components of antibody-drug conjugates, such as trastuzumab emtansine (Kadcyla), demonstrates that antibodies may be repurposed to direct therapeutic agents to antibody-resistant cancers. Here we investigated the use of CTX as a targeting agent for camptothecin (CPT)-loaded polymeric nanoparticles (NPs) directed against KRAS mutant CTX-resistant cancer cells. CPT was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs using the solvent evaporation method. CTX conjugation improved NP binding and delivery of CPT to CTX-resistant cancer cell lines. CTX successfully targeted CPT-loaded NPs to mutant KRAS PANC-1 tumours in vivo and reduced tumour growth. This study highlights that CTX can be repurposed as a targeting agent against CTX-resistant cancers and that antibody repositioning may be applicable to other antibodies restricted by resistance.
Collapse
Affiliation(s)
- William J McDaid
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michelle K Greene
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michael C Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Ellen Pollheimer
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Peter Smyth
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Kirsty McLaughlin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | | | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, NY 14260-1200, USA
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
10
|
Toll-Like Receptors and Relevant Emerging Therapeutics with Reference to Delivery Methods. Pharmaceutics 2019; 11:pharmaceutics11090441. [PMID: 31480568 PMCID: PMC6781272 DOI: 10.3390/pharmaceutics11090441] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/24/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023] Open
Abstract
The built-in innate immunity in the human body combats various diseases and their causative agents. One of the components of this system is Toll-like receptors (TLRs), which recognize structurally conserved molecules derived from microbes and/or endogenous molecules. Nonetheless, under certain conditions, these TLRs become hypofunctional or hyperfunctional, thus leading to a disease-like condition because their normal activity is compromised. In this regard, various small-molecule drugs and recombinant therapeutic proteins have been developed to treat the relevant diseases, such as rheumatoid arthritis, psoriatic arthritis, Crohn’s disease, systemic lupus erythematosus, and allergy. Some drugs for these diseases have been clinically approved; however, their efficacy can be enhanced by conventional or targeted drug delivery systems. Certain delivery vehicles such as liposomes, hydrogels, nanoparticles, dendrimers, or cyclodextrins can be employed to enhance the targeted drug delivery. This review summarizes the TLR signaling pathway, associated diseases and their treatments, and the ways to efficiently deliver the drugs to a target site.
Collapse
|
11
|
Enhancement of Binding Affinity of Folate to Its Receptor by Peptide Conjugation. Int J Mol Sci 2019; 20:ijms20092152. [PMID: 31052315 PMCID: PMC6539678 DOI: 10.3390/ijms20092152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 12/12/2022] Open
Abstract
(1) Background: The folate receptor (FR) is a target for cancer treatment and detection. Expression of the FR is restricted in normal cells but overexpressed in many types of tumors. Folate was conjugated with peptides for enhancing binding affinity to the FR. (2) Materials and Methods: For conjugation, folate was coupled with propargyl or dibenzocyclooctyne, and 4-azidophenylalanine was introduced in peptides for “click” reactions. We measured binding kinetics including the rate constants of association (ka) and dissociation (kd) of folate-peptide conjugates with purified FR by biolayer interferometry. After optimization of the conditions for the click reaction, we successfully conjugated folate with designed peptides. (3) Results: The binding affinity, indicated by the equilibrium dissociation constant (KD), of folate toward the FR was enhanced by peptide conjugation. The enhanced FR binding affinity by peptide conjugation is a result of an increase in the number of interaction sites. (4) Conclusion: Such peptide-ligand conjugates will be important in the design of ligands with higher affinity. These high affinity ligands can be useful for targeted drug delivery system.
Collapse
|
12
|
In Silico Evaluation of Two Targeted Chimeric Proteins Based on Bacterial Toxins for Breast Cancer Therapy. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2019. [DOI: 10.5812/ijcm.83315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Abstract
For the past 10 years, the annual 'Antibodies to watch' articles have provided updates on key events in the late-stage development of antibody therapeutics, such as first regulatory review or approval, that occurred in the year before publication or were anticipated to occur during the year of publication. To commemorate the 10th anniversary of the article series and to celebrate the 2018 Nobel Prizes in Chemistry and in Physiology or Medicine, which were given for work that is highly relevant to antibody therapeutics research and development, we expanded the scope of the data presented to include an overview of all commercial clinical development of antibody therapeutics and approval success rates for this class of molecules. Our data indicate that: 1) antibody therapeutics are entering clinical study, and being approved, in record numbers; 2) the commercial pipeline is robust, with over 570 antibody therapeutics at various clinical phases, including 62 in late-stage clinical studies; and 3) Phase 1 to approval success rates are favorable, ranging from 17-25%, depending on the therapeutic area (cancer vs. non-cancer). In 2018, a record number (12) of antibodies (erenumab (Aimovig), fremanezumab (Ajovy), galcanezumab (Emgality), burosumab (Crysvita), lanadelumab (Takhzyro), caplacizumab (Cablivi), mogamulizumab (Poteligeo), moxetumomab pasudodox (Lumoxiti), cemiplimab (Libtayo), ibalizumab (Trogarzo), tildrakizumab (Ilumetri, Ilumya), emapalumab (Gamifant)) that treat a wide variety of diseases were granted a first approval in either the European Union (EU) or United States (US). As of November 2018, 4 antibody therapeutics (sacituzumab govitecan, ravulizumab, risankizumab, romosozumab) were being considered for their first marketing approval in the EU or US, and an additional 3 antibody therapeutics developed by Chinese companies (tislelizumab, sintilimab, camrelizumab) were in regulatory review in China. In addition, our data show that 3 product candidates (leronlimab, brolucizumab, polatuzumab vedotin) may enter regulatory review by the end of 2018, and at least 12 (eptinezumab, teprotumumab, crizanlizumab, satralizumab, tanezumab, isatuximab, spartalizumab, MOR208, oportuzumab monatox, TSR-042, enfortumab vedotin, ublituximab) may enter regulatory review in 2019. Finally, we found that approximately half (18 of 33) of the late-stage pipeline of antibody therapeutics for cancer are immune checkpoint modulators or antibody-drug conjugates. Of these, 7 (tremelimumab, spartalizumab, BCD-100, omburtamab, mirvetuximab soravtansine, trastuzumab duocarmazine, and depatuxizumab mafodotin) are being evaluated in clinical studies with primary completion dates in late 2018 and in 2019, and are thus 'antibodies to watch'. We look forward to documenting progress made with these and other 'antibodies to watch' in the next installment of this article series.
Collapse
Affiliation(s)
- Hélène Kaplon
- a Institut de Recherches Servier , Croissy-sur-Seine, the Division of Biotechnology & Biomarker Research , France
| | | |
Collapse
|
14
|
Toth RT, Pace SE, Mills BJ, Joshi SB, Esfandiary R, Middaugh CR, Weis DD, Volkin DB. Evaluation of Hydrogen Exchange Mass Spectrometry as a Stability-Indicating Method for Formulation Excipient Screening for an IgG4 Monoclonal Antibody. J Pharm Sci 2018; 107:1009-1019. [DOI: 10.1016/j.xphs.2017.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/25/2017] [Accepted: 12/06/2017] [Indexed: 12/23/2022]
|
15
|
Ramos-Tomillero I, Perez-Chacon G, Somovilla-Crespo B, Sanchez-Madrid F, Domínguez JM, Cuevas C, Zapata JM, Rodríguez H, Albericio F. Bioconjugation through Mesitylene Thiol Alkylation. Bioconjug Chem 2018; 29:1199-1208. [DOI: 10.1021/acs.bioconjchem.7b00828] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Iván Ramos-Tomillero
- Institute for Research in Biomedicine, 08028-Barcelona, Spain
- Department of Organic Chemistry, University of Barcelona, 08028-Barcelona, Spain
| | - Gema Perez-Chacon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, 28029-Madrid, Spain
| | - Beatriz Somovilla-Crespo
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital de la Princesa, 28006-Madrid, Spain
| | - Francisco Sanchez-Madrid
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital de la Princesa, 28006-Madrid, Spain
| | | | - Carmen Cuevas
- Research Department, PharmaMar S.A., Colmenar Viejo, 28770-Madrid, Spain
| | - Juan Manuel Zapata
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, 28029-Madrid, Spain
| | | | - Fernando Albericio
- Institute for Research in Biomedicine, 08028-Barcelona, Spain
- Department of Organic Chemistry, University of Barcelona, 08028-Barcelona, Spain
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, 08028-Barcelona, Spain
- School of Chemistry, University of KwaZulu-Natal, 4001-Durban, South Africa
| |
Collapse
|
16
|
Abstract
The pace of antibody therapeutics development accelerated in 2017, and this faster pace is projected to continue through 2018. Notably, the annual number of antibody therapeutics granted a first approval in either the European Union (EU) or United States (US) reached double-digits (total of 10) for the first time in 2017. The 10 antibodies granted approvals are: brodalumab, dupilumab, sarilumab, guselkumab, benralizumab, ocrelizumab, inotuzumab ozogamicin, avelumab, duvalumab, and emicizumab. Brodalumab, however, had already been approved in Japan in 2016. As of December 1, 2017, nine antibody therapeutics (ibalizumab, burosumab, tildrakizumab, caplacizumab, erenumab, fremanezumab, galcanezumab, romosozumab, mogamulizumab) were in regulatory review in the EU or US, and regulatory actions on their marketing applications are expected by the end of 2018. Based on company announcements and estimated clinical study primary completion dates, and assuming the study results are positive, marketing applications for at least 12 antibody therapeutics that are now being evaluated in late-stage clinical studies may be submitted by the end of 2018. Of the 12 candidates, 8 are for non-cancer indications (lanadelumab, crizanlizumab, ravulizumab, eptinezumab, risankizumab, satralizumab, brolucizumab, PRO140) and 4 are for cancer (sacituzumab govitecan, moxetumomab pasudotox, cemiplimab, ublituximab). Additional antibody therapeutics to watch in 2018 include 19 mAbs undergoing evaluation in late-stage studies with primary completion dates in late 2017 or during 2018. Of these mAbs, 9 are for non-cancer indications (lampalizumab, roledumab, emapalumab, fasinumab, tanezumab, etrolizumab, NEOD001, gantenerumab, anifrolumab) and 10 are for cancer indications (tremelimumab, isatuximab, BCD-100, carotuximab, camrelizumab, IBI308, glembatumumab vedotin, mirvetuximab soravtansine, oportuzumab monatox, L19IL2/L19TNF). Positive clinical study results may enable marketing application submissions in 2018. Brief summaries of these antibody therapeutics are provided in this installment of the 'Antibodies to watch' article series.
Collapse
Affiliation(s)
- Hélène Kaplon
- Laboratory UMRS 1138 “Cancer, Immune Control and Escape”, Cordeliers Research Centre, Paris, France
| | | |
Collapse
|
17
|
Qin H, Wei G, Sakamaki I, Dong Z, Cheng WA, Smith DL, Wen F, Sun H, Kim K, Cha S, Bover L, Neelapu SS, Kwak LW. Novel BAFF-Receptor Antibody to Natively Folded Recombinant Protein Eliminates Drug-Resistant Human B-cell Malignancies In Vivo. Clin Cancer Res 2017; 24:1114-1123. [PMID: 29180606 DOI: 10.1158/1078-0432.ccr-17-1193] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/20/2017] [Accepted: 11/21/2017] [Indexed: 02/05/2023]
Abstract
Purpose: mAbs such as anti-CD20 rituximab are proven therapies in B-cell malignancies, yet many patients develop resistance. Novel therapies against alternative targets are needed to circumvent resistance mechanisms. We sought to generate mAbs against human B-cell-activating factor receptor (BAFF-R/TNFRSF13C), which has not yet been targeted successfully for cancer therapy.Experimental Design: Novel mAbs were generated against BAFF-R, expressed as a natively folded cell surface immunogen on mouse fibroblast cells. Chimeric BAFF-R mAbs were developed and assessed for in vitro and in vivo monotherapy cytotoxicity. The chimeric mAbs were tested against human B-cell tumor lines, primary patient samples, and drug-resistant tumors.Results: Chimeric antibodies bound with high affinity to multiple human malignant B-cell lines and induced potent antibody-dependent cellular cytotoxicity (ADCC) against multiple subtypes of human lymphoma and leukemia, including primary tumors from patients who had relapsed after anti-CD20 therapy. Chimeric antibodies also induced ADCC against ibrutinib-resistant and rituximab-insensitive CD20-deficient variant lymphomas, respectively. Importantly, they demonstrated remarkable in vivo growth inhibition of drug-resistant tumor models in immunodeficient mice.Conclusions: Our method generated novel anti-BAFF-R antibody therapeutics with remarkable single-agent antitumor effects. We propose that these antibodies represent an effective new strategy for targeting and treating drug-resistant B-cell malignancies and warrant further development. Clin Cancer Res; 24(5); 1114-23. ©2017 AACR.
Collapse
Affiliation(s)
- Hong Qin
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.
| | - Guowei Wei
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Ippei Sakamaki
- Department of Clinical Infectious diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Zhenyuan Dong
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Wesley A Cheng
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - D Lynne Smith
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Feng Wen
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.,Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Han Sun
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Kunhwa Kim
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soungchul Cha
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Bover
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Larry W Kwak
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.
| |
Collapse
|
18
|
Recent advances in the development of novel protein scaffolds based therapeutics. Int J Biol Macromol 2017; 102:630-641. [DOI: 10.1016/j.ijbiomac.2017.04.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
|
19
|
Wang Y, Yang YJ, Wang Z, Liao J, Liu M, Zhong XR, Zheng H, Wang YP. CD55 and CD59 expression protects HER2-overexpressing breast cancer cells from trastuzumab-induced complement-dependent cytotoxicity. Oncol Lett 2017; 14:2961-2969. [PMID: 28928834 PMCID: PMC5588148 DOI: 10.3892/ol.2017.6555] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/28/2017] [Indexed: 02/05/2023] Open
Abstract
A large proportion (40-60%) of patients with human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer do not benefit from trastuzumab treatment, potentially due to the lack of complement-dependent cytotoxicity (CDC) activation. In the present study, the effect of complement decay-accelerating factor (CD55) and CD59 glycoprotein precursor (CD59) expression on trastuzumab-induced CDC in HER2-positive breast cancer cell lines was investigated. The CD55 and CD59-overexpressing and HER2-positive cell lines SK-BR-3 and BT474 were selected for subsequent experiments. Blocking CD55 and CD59 function using targeting monoclonal antibodies significantly enhanced the cell lysis of SK-BR-3 and BT474 cells following treatment with trastuzumab. In addition, following treatment with 0.1 U/ml phosphatidylinositol-specific phospholipase C (PI-PLC) for 1 h, CD55 and CD59 surface expression was significantly decreased, and the cell lysis rate was further enhanced. Treatment of SK-BR-3 cells with short hairpin RNA (shRNA) targeting CD55 and CD59 downregulated CD55 and CD59 expression at the mRNA and protein levels, and resulted in significantly enhanced trastuzumab-induced CDC-dependent lysis. The data from the present study suggested that CD55 and CD59 serve roles in blocking trastuzumab-induced CDC, therefore strategies targeting CD55 and CD59 may overcome breast cancer cell resistance to trastuzumab. The results from the present study may provide a basis for developing suitable, personalized treatment strategies to improve the clinical efficacy of trastuzumab for patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jun Yang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Liao
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mei Liu
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Breast Cancer Research Center, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Xiao-Rong Zhong
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Zheng
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Ping Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
20
|
Golay J. Direct targeting of cancer cells with antibodies: What can we learn from the successes and failure of unconjugated antibodies for lymphoid neoplasias? J Autoimmun 2017; 85:6-19. [PMID: 28666691 DOI: 10.1016/j.jaut.2017.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022]
Abstract
Following approval in 1997 of the anti-CD20 antibody rituximab for the treatment of B-NHL and CLL, many other unconjugated IgG1 MAbs have been tested in pre-clinical and clinical trials for the treatment of lymphoid neoplasms. Relatively few have been approved however and these are directed against a limited number of target antigens (CD20, CD52, CCR4, CD38, CD319). We review here the known biological properties of these antibodies and discuss which factors may have led to their success or may, on the contrary, limit their clinical application. Common factors of the approved MAbs are that the target antigen is expressed at relatively high levels on the neoplastic targets and their mechanism of action is mostly immune-mediated. Indeed most of these MAbs induce ADCC and phagocytosis by macrophages, and many also activate complement, leading to target cell lysis. In contrast direct cell death induction is not a common feature but may enhance efficacy in some cases. Interestingly, a key factor for the success of several MAbs appears to be their capacity to skew immunity towards an anti-tumour mode, by inhibiting/depleting suppressor cells and/or activating immune cells within the microenvironment, independently of FcγRs. We also expose here some of the strategies employed by industry to expand the clinical use of these molecules beyond their original indication. Interestingly, due to the central role of lymphocytes in the control of the immune response, several of the antibodies are now successfully used to treat many different autoimmune diseases and have also been formally approved for some of these new indications. There is little doubt that this trend will continue and that the precise mechanisms of therapeutic MAbs will be further dissected and better understood in the context of both tumour immunology and autoimmunity.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy "G. Lanzani", USC Haematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Via Garibaldi 11-13, 24128, Bergamo, Italy.
| |
Collapse
|
21
|
Abstract
Over 50 investigational monoclonal antibody (mAb) therapeutics are currently undergoing evaluation in late-stage clinical studies, which is expected to drive a trend toward first marketing approvals of at least 6–9 mAbs per year in the near-term. In the United States (US), a total of 6 and 9 mAbs were granted first approvals during 2014 and 2015, respectively; all these products are also approved in the European Union (EU). As of December 1, 2016, 6 mAbs (atezolizumab, olaratumab, reslizumab, ixekizumab, bezlotoxumab, oblitoxaximab) had been granted first approvals during 2016 in either the EU or US. Brodalumab, was granted a first approval in Japan in July 2016. Regulatory actions on marketing applications for brodalumab in the EU and US are not expected until 2017. In 2017, first EU or US approvals may also be granted for at least nine mAbs (ocrelizumab, avelumab, Xilonix, inotuzumab ozogamicin, dupilumab, sirukumab, sarilumab, guselkumab, romosozumab) that are not yet approved in any country. Based on announcements of company plans for regulatory submissions and the estimated completion dates for late-stage clinical studies, and assuming the study results are positive, marketing applications for at least 6 antibody therapeutics (benralizumab, tildrakizumab, emicizumab, galcanezumab, ibalizumab, PRO-140) that are now being evaluated in late-stage clinical studies may be submitted during December 2016* or 2017. Other ‘antibodies to watch' in 2017 include 20 mAbs are undergoing evaluation in pivotal studies that have estimated primary completion dates in late 2016 or during 2017. Of these, 5 mAbs are for cancer (durvalumab, JNJ-56022473, ublituximab, anetumab ravtansine, glembatumumab vedotin) and 15 mAbs are for non-cancer indications (caplacizumab, lanadelumab, roledumab, tralokinumab, risankizumab, SA237, emapalumab, suptavumab, erenumab, eptinezumab, fremanezumab, fasinumab, tanezumab, lampalizumab, brolucizumab). Positive results from these studies may enable submission of marketing applications in 2017 or 2018, or provide justification for additional studies. *See note added in proof for update through December 31, 2016.
Collapse
|
22
|
Li W, Yang B, Zhou D, Xu J, Ke Z, Suen WC. Discovery and characterization of antibody variants using mass spectrometry-based comparative analysis for biosimilar candidates of monoclonal antibody drugs. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1025:57-67. [DOI: 10.1016/j.jchromb.2016.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
|
23
|
FAT1: a potential target for monoclonal antibody therapy in colon cancer. Br J Cancer 2016; 115:40-51. [PMID: 27328312 PMCID: PMC4931367 DOI: 10.1038/bjc.2016.145] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the major causes of cancer-associated mortality worldwide. The currently approved therapeutic agents have limited efficacy. Methods: The atypical cadherin FAT1 was discovered as a novel CRC-associated protein by using a monoclonal antibody (mAb198.3). FAT1 expression was assessed in CRC cells by immunohistochemistry (IHC), immunoblots, flow cytometry and confocal microscopy. In addition, in vitro and in vivo tumour models were done to assess FAT1 potential value for therapeutic applications. Results: The study shows that FAT1 is broadly expressed in primary and metastatic CRC stages and detected by mAb198.3, regardless of KRAS and BRAF mutations. FAT1 mainly accumulates at the plasma membrane of cancer cells, whereas it is only marginally detected in normal human samples. Moreover, the study shows that FAT1 has an important role in cell invasiveness while it does not significantly influence apoptosis. mAb198.3 specifically recognises FAT1 on the surface of colon cancer cells and is efficiently internalised. Furthermore, it reduces cancer growth in a colon cancer xenograft model. Conclusions: This study provides evidence that FAT1 and mAb198.3 may offer new therapeutic opportunities for CRC including the tumours resistant to current EGFR-targeted therapies.
Collapse
|
24
|
McLean LP, Cross RK. Pharmacodynamic assessment of vedolizumab for the treatment of ulcerative colitis. Expert Opin Drug Metab Toxicol 2016; 12:833-42. [PMID: 27096357 DOI: 10.1080/17425255.2016.1181171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Vedolizumab is an anti-integrin approved for the treatment of Crohn's disease and ulcerative colitis. By binding the α4β7-integrin heterodimer, vedolizumab blocks leukocyte translocation into gastrointestinal tissue. AREAS COVERED This review discusses the chemistry, pharmacologic properties, clinical efficacy, and safety of vedolizumab in ulcerative colitis. Other medications available for the treatment of ulcerative colitis are also discussed. EXPERT OPINION Vedolizumab is a promising new agent for the treatment of ulcerative colitis. Its mechanism of action differs from TNF-α inhibitors and immune suppressants, allowing it to be used in cases of TNF-α inhibitor failure or non-response, or as a first-line biologic drug. Available safety data suggests that vedolizumab is not associated with an increased risk of infection or malignancy; however, additional post-marketing data are required to confirm these initial reports. Vedolizumab is likely to be used in growing numbers of patients over the coming years.
Collapse
Affiliation(s)
- Leon P McLean
- a Department of Medicine, Division of Gastroenterology and Hepatology , University of Maryland , Baltimore , MD , USA
| | - Raymond K Cross
- a Department of Medicine, Division of Gastroenterology and Hepatology , University of Maryland , Baltimore , MD , USA
| |
Collapse
|
25
|
Singh H, Grewal N, Arora E, Kumar H, Kakkar AK. Vedolizumab: A novel anti-integrin drug for treatment of inflammatory bowel disease. J Nat Sci Biol Med 2016; 7:4-9. [PMID: 27003961 PMCID: PMC4780165 DOI: 10.4103/0976-9668.175016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is the chronic inflammatory disorder of gastrointestinal tract consisting of two subtypes: Ulcerative colitis and Crohn's disease. IBD occurs due to infiltration of leukocytes in intestinal mucosa and derangements in intestinal barrier function. One of the most important steps in pathogenesis of IBD is the interactions between integrins on the surface of leukocyte. The α4β7 integrin expressing T-cell is an important leukocyte involved in pathogenesis and represents a new drug target for the treatment of IBD. Vedolizumab is a humanized monoclonal antibody, which acts against α4β7 integrin heterodimer and blocks the interaction of α4β7 integrin with MAdCAM-1. It prevents leukocyte binding to endothelial surface and its extravasation into affected tissue. The efficacy and safety of the vedolizumab have been established in many clinical studies. It has shown promising results in various clinical studies where a greater percentage of patients as compared to a placebo achieved and maintained clinical response, clinical remission, and corticosteroid-free clinical remission. Vedolizumab has been shown to be well tolerated with slightly higher risk of infections, headache, naspharyngitis as compared to placebo. This review focuses on the potential role of vedolizumab for the treatment of IBD.
Collapse
Affiliation(s)
- Harmanjit Singh
- Department of Pharmacology, All India Institutes of Medical Sciences (AIIMS), New Delhi, India
| | - Nipunjot Grewal
- Department of Pharmacology, Punjab Institute of Medical Sciences, Jalandhar, Punjab, India
| | - Ekta Arora
- Department of Pharmacology, All India Institutes of Medical Sciences (AIIMS), New Delhi, India
| | - Harish Kumar
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | |
Collapse
|
26
|
Brunn ND, Mauze S, Gu D, Wiswell D, Ueda R, Hodges D, Beebe AM, Zhang S, Escandón E. The Role of Anti-Drug Antibodies in the Pharmacokinetics, Disposition, Target Engagement, and Efficacy of a GITR Agonist Monoclonal Antibody in Mice. J Pharmacol Exp Ther 2016; 356:574-86. [PMID: 26669426 DOI: 10.1124/jpet.115.229864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/14/2015] [Indexed: 03/08/2025] Open
Abstract
Administration of biologics to enhance T-cell function is part of a rapidly growing field of cancer immunotherapy demonstrated by the unprecedented clinical success of several immunoregulatory receptor targeting antibodies. While these biologic agents confer significant anti-tumor activity through targeted immune response modulation, they can also elicit broad immune responses potentially including the production of anti-drug antibodies (ADAs). DTA-1, an agonist monoclonal antibody against GITR, is a highly effective anti-tumor treatment in preclinical models. We demonstrate that repeated dosing with murinized DTA-1 (mDTA-1) generates ADAs with corresponding reductions in drug exposure and engagement of GITR on circulating CD3(+) CD4(+) T cells, due to rapid hepatic drug uptake and catabolism. Mice implanted with tumors after induction of preexisting mDTA-1 ADA show no anti-tumor efficacy when given 3 mg/kg mDTA-1, an efficacious dose in naive mice. Nonetheless, increasing mDTA-1 treatment to 30 mg/kg in ADA-positive mice restores mDTA-1 exposure and GITR engagement on circulating CD3(+) CD4(+) T cells, thereby partially restoring anti-tumor efficacy. Formation of anti-mDTA-1 antibodies and changes in drug exposure and disposition does not occur in GITR(-/-) mice, consistent with a role for GITR agonism in humoral immunity. Finally, the administration of muDX400, a murinized monoclonal antibody against the checkpoint inhibitor PD-1, dosed alone or combined with mDTA-1 did not result in reduced muDX400 exposure, nor did it change the nature of the anti-mDTA-1 response. This indicates that anti-GITR immunogenicity may not necessarily impact the pharmacology of coadministered monoclonal antibodies, supporting combination immunomodulatory strategies.
Collapse
Affiliation(s)
- Nicholas D Brunn
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Smita Mauze
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Danling Gu
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Derek Wiswell
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Roanna Ueda
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Douglas Hodges
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Amy M Beebe
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Shuli Zhang
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| | - Enrique Escandón
- Biologics DMPK and Disposition (N.D.B., D.W., D.H., E.E.), Immuno-oncology (S.M., D.G., R.U., A.M.B.), and Bioanalytics Department (S.Z.), Merck Research Laboratories, Merck & Co. Inc., Palo Alto, California
| |
Collapse
|
27
|
König M, Rharbaoui F, Aigner S, Dälken B, Schüttrumpf J. Tregalizumab - A Monoclonal Antibody to Target Regulatory T Cells. Front Immunol 2016; 7:11. [PMID: 26834751 PMCID: PMC4724712 DOI: 10.3389/fimmu.2016.00011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/11/2016] [Indexed: 12/18/2022] Open
Abstract
Regulatory T cells (Tregs) represent a subpopulation of CD4+ T cells, which are essential for the maintenance of immunological tolerance. The absence or dysfunction of Tregs can lead to autoimmunity and allergies. The restoration of functional Tregs and/or Treg cell numbers represents a novel and attractive approach for the treatment of autoimmune diseases, e.g., rheumatoid arthritis (RA). The CD4 cell surface receptor is a target for modulation of T cell function. Monoclonal antibodies (mAbs) against CD4 have previously been tested for the treatment of autoimmune diseases, including RA. Furthermore, in model systems, anti-CD4 antibodies are able to induce tolerance and mediate immunomodulatory effects through a variety of mechanisms. Despite the availability of innovative and effective therapies for RA, many patients still have persistently active disease or experience adverse events that can limit use. A growing body of evidence suggests that Treg modulation could offer a new therapeutic strategy in RA and other autoimmune disorders. Here, we describe tregalizumab (BT-061), which is a novel, non-depleting IgG1 mAb that binds to a unique epitope of CD4. Tregalizumab represents the first humanized anti-CD4 mAb that selectively induces Treg activation.
Collapse
|
28
|
Jaccoulet E, Smadja C, Taverna M. Quality Control of Therapeutic Monoclonal Antibodies at the Hospital After Their Compounding and Before Their Administration to Patients. Methods Mol Biol 2016; 1466:179-184. [PMID: 27473490 DOI: 10.1007/978-1-4939-4014-1_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Monoclonal antibodies (mAbs) are widely used in cancer therapy and recently many new mAbs have gained EMA and FDA approvals for oncology indications. Here we describe a highly reproducible CZE method, relying on a cationic coating allowing separation and identification of a complex mixture of four compounded mAbs widely used in cancer therapy (cetuximab, rituximab, bevacizumab, and trastuzumab).
Collapse
Affiliation(s)
- Emmanuel Jaccoulet
- Institut Galien Paris-Sud, UMR 8612, Proteins and Nanotechnology in Analytical Science, CNRS, Univ Paris-Sud, Univ Paris-Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France
- Hôpital Européen Georges Pompidou, Paris, France
| | - Claire Smadja
- Institut Galien Paris-Sud, UMR 8612, Proteins and Nanotechnology in Analytical Science, CNRS, Univ Paris-Sud, Univ Paris-Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France.
| | - Myriam Taverna
- Institut Galien Paris-Sud, UMR 8612, Proteins and Nanotechnology in Analytical Science (PNAS), CNRS, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
29
|
Wu SC, Chen YJ, Wang HC, Chou MY, Chang TY, Yuan SS, Chen CY, Hou MF, Hsu JTA, Wang YM. Bispecific Antibody Conjugated Manganese-Based Magnetic Engineered Iron Oxide for Imaging of HER2/neu- and EGFR-Expressing Tumors. Am J Cancer Res 2016; 6:118-30. [PMID: 26722378 PMCID: PMC4679359 DOI: 10.7150/thno.13069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/21/2015] [Indexed: 11/05/2022] Open
Abstract
The overexpression of HER2/neu and EGFR receptors plays important roles in tumorigenesis and tumor progression. Targeting these two receptors simultaneously can have a more widespread application in early diagnosis of cancers. In this study, a new multifunctional nanoparticles (MnMEIO-CyTE777-(Bis)-mPEG NPs) comprising a manganese-doped iron oxide nanoparticle core (MnMEIO), a silane-amino functionalized poly(ethylene glycol) copolymer shell, a near infrared fluorescence dye (CyTE777), and a covalently conjugated anti-HER2/neu and anti-EGFR receptors bispecific antibody (Bis) were successfully developed. In vitro T2-weighted MR imaging studies in SKBR-3 and A431 tumor cells incubated with MnMEIO-CyTE777-(Bis)-mPEG NPs showed - 94.8 ± 3.8 and - 84.1 ± 2.8% negative contrast enhancement, respectively. Pharmacokinetics study showed that MnMEIO-CyTE777-(Bis)-mPEG NPs were eliminated from serum with the half-life of 21.3 mins. In vivo MR imaging showed that MnMEIO-CyTE777-(Bis)-mPEG NPs could specifically and effectively target to HER2/neu- and EGFR-expressing tumors in mice; the relative contrast enhancements were 11.8 (at 2 hrs post-injection) and 61.5 (at 24 hrs post-injection) fold higher in SKBR-3 tumors as compared to Colo-205 tumors. T2-weighted MR and optical imaging studies revealed that the new contrast agent (MnMEIO-CyTE777-(Bis)-mPEG NPs) could specifically and effectively target to HER2/neu- and/or EGFR-expressing tumors. Our results demonstrate that MnMEIO-CyTE777-(Bis)-mPEG NPs are able to recognize the tumors expressing both HER2/neu and/or EGFR, and may provide a novel molecular imaging tool for early diagnosis of cancers expressing HER2/neu and/or EGFR.
Collapse
|
30
|
Faghihi H, Khalili F, Amini M, Vatanara A. The effect of freeze-dried antibody concentrations on its stability in the presence of trehalose and hydroxypropyl-β-cyclodextrin: a Box-Behnken statistical design. Pharm Dev Technol 2015; 22:724-732. [PMID: 26654052 DOI: 10.3109/10837450.2015.1116563] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The present study aimed at preparation and optimization of stable freeze-dried immunoglobulin G (IgG) applying proper amount of antibody with efficient combination of trehalose and hydroxypropyl-β-cyclodextrin (HPβCD). Response surface methodology was employed through a three-factor, three-level Box-Behnken design. Amounts of IgG (X1), trehalose (X2) and HPβCD (X3) were independent variables. Aggregation following process (Y1), after one month at 45 °C (Y2), upon two month at 45 °C (Y3) and beta-sheet content of IgG (Y4) were determined as dependent variables. Results were fitted to quadratic models (except for beta-sheet content), describing the inherent relationship between main factors. Optimized formulation composed of 55.85 mg IgG, 52.51 mg trehalose and 16.01 mg HPβCD was prepared. The calculated responses of the optimized formulation were as follows: Y1 = 0.19%, Y2 = 0.78%, Y3 = 1.88% and Y4 = 68.60%, respectively. The thermal analysis confirmed the amorphous nature of optimum formulation and the integrity of IgG was shown to be favorably preserved. Validation of the optimization study demonstrated high degree of prognostic ability. The DOE study successfully predicted the optimum values of antibody as well as stabilizers for desirable process and storage stabilization of freeze-dried IgG.
Collapse
Affiliation(s)
| | | | - Mohsen Amini
- b Department of Medicinal Chemistry , Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | | |
Collapse
|
31
|
Abstract
INTRODUCTION Dermatology is a relatively small field concerned with conditions of the hair, skin, nails and their related diseases; yet there is considerable active research and development within the field. Pharmaceutical companies seek more effective treatments through various therapeutic classes and delivery routes. However, 28 drugs have been discontinued for the treatment of dermatologic diseases in 2014. AREAS COVERED Herein, the authors summarize the details about each discontinued drug in 2014. The dermatological conditions covered are: psoriasis, eczema, leg ulcers, wounds, allergies, acne, scleroderma, lupus erythematosus, urticaria, mastocytosis, epidermolysis bullosa, onychomycosis and one other unspecified disease. The authors also provide suggestions for improving and accelerating the future of dermatological drug development. EXPERT OPINION It is clear that improved metrics, especially for early assessment, emphasizing clinical relevance, are necessary to increase success rate. Transparency and clear communication within the field is necessary to reduce and salvage the waste that accumulates from these costly studies. Focused attention on how preclinical and early clinical studies failed to indicate subsequent toxicity profiles in patients would accelerate drug development. Distinguishing between disappointing study results and business/financial factors is important when analyzing discontinuations. A reformed approach toward study design would aid both. Ultimately, relevance and practicality for the patient must be kept in mind at all times.
Collapse
Affiliation(s)
- Christina Phuong
- a University of California, Department of Dermatology , Surge Building Room 110, 90 Medical Center Way, San Francisco, CA 94143-0989, USA +1 415 673 9690 ; +1 415 673 3533 ;
| | - Howard I Maibach
- a University of California, Department of Dermatology , Surge Building Room 110, 90 Medical Center Way, San Francisco, CA 94143-0989, USA +1 415 673 9690 ; +1 415 673 3533 ;
| |
Collapse
|
32
|
Abstract
The commercial pipeline of recombinant antibody therapeutics is robust and dynamic. As of early December 2014, a total of 6 such products (vedolizumab, siltuximab, ramucirumab, pembrolizumab, nivolumab, blinatumomab) were granted first marketing approvals in 2014. As discussed in this perspective on antibodies in late-stage development, the outlook for additional approvals, potentially still in 2014 and certainly in 2015, is excellent as marketing applications for 7 antibody therapeutics (secukinumab, evolocumab, mepolizumab, dinutuximab, nivolumab, blinatumomab, necitumumab) are undergoing a first regulatory review in the EU or US. Of the 39 novel mAbs currently in Phase 3 studies, a marketing application for one (alirocumab) may be submitted in late 2014, and marketing application submissions for at least 4 (reslizumab, ixekizumab, ocrelizumab, obiltoxaximab) are expected in 2015. Other 'antibodies to watch' are those in Phase 3 studies with estimated primary completion dates in late 2014 or 2015, which includes 13 for non-cancer indications (brodalumab, bimagrumab, bococizumab, MABp1, gevokizumab, dupilumab, sirukumab, sarilumab, tildrakizumab, guselkumab, epratuzumab, combination of actoxumab + bezlotoxumab, romosozumab) and 2 (racotumomab and clivatuzumab tetraxetan) undergoing evaluation as treatments for cancer. In addition to the novel antibody therapeutics mentioned, biosimilar infliximab and biosimilar trastuzumab are 'antibodies to watch' in 2015 because of their potential for entry into the US market and regulatory review, respectively.
Collapse
Key Words
- BLA, biologics license application
- EMA, European Medicines Agency
- European Medicines Agency
- FDA, Food and Drug Administration
- Food and Drug Administration
- IV, intravenous
- MTX, methotrexate
- PA, protective antigen
- PCSK9, proprotein convertase subtilisin/kexin type 9
- RA, rheumatoid arthritis
- SLE, systemic lupus erythematosus
- biosimilar antibodies
- cancer
- clinical studies
- immune-mediated disorders
- interleukin, IL
- mAb, monoclonal antibody
- monoclonal antibodies
- pharmacokinetic, PK
Collapse
|
33
|
Lin X, Zhu H, Luo Z, Hong Y, Zhang H, Liu X, Ding H, Tian H, Yang Z. Near-infrared fluorescence imaging of non-Hodgkin's lymphoma CD20 expression using Cy7-conjugated obinutuzumab. Mol Imaging Biol 2015; 16:877-87. [PMID: 24833041 DOI: 10.1007/s11307-014-0742-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Obinutuzumab is the first fully humanized and glycoengineered monoclonal antibody (mAb) directly targeting CD20 antigen, which is expressed on B cell lymphocytes and the majority of non-Hodgkin's lymphoma (NHL). This study aims to design a diagnostic molecular probe, Cy7-Obinutuzumab (Cy7-Obi), in which Cy7 is a near-infrared fluorescent dye. This probe is used to noninvasively image CD20 antigen expressed in NHL cells. PROCEDURES Cy7-Obi probe was synthesized through nucleophilic substitution reaction between NHS-Cy7 and obinutuzumab. After purification, the conjugate was fully characterized by a series of methods. The immunoreactivity and molecular specificity of the probe were confirmed using flow cytometry and in vitro microscopy on Raji (CD20-positive) cells. For in vivo imaging, Cy7-Obi probe (1 nmol) was injected intravenously in severe combined immunodeficiency (SCID) mice bearing Raji tumors which overexpress CD20 (n = 3) and was imaged with near-infrared fluorescence (NIRF) at 6, 9, 12, 24, 60, and 96 h post-probe injection. For pre-block, obinutuzumab (3.25 mg) was injected intravenously in tumor-bearing mice 6 h before the administration of Cy7-Obi probe. RESULTS The synthesized Cy7-Obi probe in this paper mimics obinutuzumab in both structure and function. Flow cytometry analysis of the probe and obinutuzumab on Raji cells showed minor difference in binding affinity/specificity with CD20. The probe showed significant fluorescence signal when it was examined on Raji cells using in vitro microscopy. The fluorescence signal can be blocked by pretreatment with obinutuzumab. The probe Cy7-Obi also showed high tumor uptake when it was examined by in vivo optical imaging on Raji tumor-bearing mice. The tumor uptake can be blocked by pretreatment with obinutuzumab (n = 3, p < 0.05). The in vivo imaging results were also confirmed by ex vivo imaging of dissected organs. Finally, the probe Cy7-Obi has shown excellent tumor targeting and specificity through immunofluorescence analysis. CONCLUSIONS We have shown that humanized Cy7-Obi probe can be used for NIRF imaging successfully. The probe may be an effective and noninvasive diagnostic molecular probe capable of tracking CD20 overexpression in NHL.
Collapse
Affiliation(s)
- Xinfeng Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, 100142, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Huang J, Tang Q, Wang C, Yu H, Feng Z, Zhu J. Molecularly Targeted Therapy of Human Hepatocellular Carcinoma Xenografts with Radio-iodinated Anti-VEGFR2 Murine-Human Chimeric Fab. Sci Rep 2015; 5:10660. [PMID: 26021484 PMCID: PMC4448128 DOI: 10.1038/srep10660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/21/2015] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) is traditionally regarded as an important therapeutic target in a wide variety of malignancies, such as hepatocellular carcinoma (HCC). We previously generated a murine-human anti-VEGFR2 chimeric Fab (cFab), named FA8H1, which has the potential to treat VEGFR2-overexpressing solid tumors. Here, we investigated whether FA8H1 can be used as a carrier in molecularly targeted therapy in HCC xenograft models. FA8H1 was labeled with 131I, and two HCC xenograft models were generated using BEL-7402 (high VEGFR2-expressing) and SMMC-7721 (low VEGFR2-expressing) cells, which were selected from five HCC cell lines. The biodistribution of 131I-FA8H1 was determined in both models by Single-Photon Emission Computed Tomography and therapeutic effects were monitored in nude mice bearing BEL-7402 xenografts. Finally, we determined the involvement of necrosis and apoptotic pathways in treated mice using immunohistochemistry. 131I-FA8H1 levels were dramatically reduced in blood and other viscera. The therapeutic effect of 131I-labeled FA8H1 in the BEL-7402 model was significantly better than that by 131I and FA8H1 alone. We observed extensive necrosis in the treated tumors, and both FasL and caspase 3 were up-regulated. Thus, 131I-anti-VEGFR2 cFab has the potential to be used for molecularly targeted treatment of HCC overexpressing VEGFR2.
Collapse
Affiliation(s)
- Jianfei Huang
- 1] Key Laboratory of Antibody Technique, Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China [2] Department of Pathology, Affiliated Hospital of Nantong University. Nantong, Jiangsu 226001, China
| | - Qi Tang
- Key Laboratory of Antibody Technique, Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing, Jiangsu 210002, China
| | - Huixin Yu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Zhenqing Feng
- 1] Key Laboratory of Antibody Technique, Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China [2] Jiangsu Key Lab of Cancer Biomarkers, Prevention &Treatment, Cancer Center, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jin Zhu
- 1] Key Laboratory of Antibody Technique, Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China [2] Huadong Medical Institute of Biotechniques, Nanjing, Jiangsu 210002, China
| |
Collapse
|
35
|
Diamant E, Torgeman A, Ozeri E, Zichel R. Monoclonal Antibody Combinations that Present Synergistic Neutralizing Activity: A Platform for Next-Generation Anti-Toxin Drugs. Toxins (Basel) 2015; 7:1854-81. [PMID: 26035486 PMCID: PMC4488679 DOI: 10.3390/toxins7061854] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/07/2015] [Accepted: 05/19/2015] [Indexed: 12/25/2022] Open
Abstract
Monoclonal antibodies (MAbs) are among the fastest-growing therapeutics and are being developed for a broad range of indications, including the neutralization of toxins, bacteria and viruses. Nevertheless, MAbs potency is still relatively low when compared to conventional polyclonal Ab preparations. Moreover, the efficacy of an individual neutralizing MAb may significantly be hampered by the potential absence or modification of its target epitope in a mutant or subtype of the infectious agent. These limitations of individual neutralizing MAbs can be overcome by using oligoclonal combinations of several MAbs with different specificities to the target antigen. Studies conducted in our lab and by others show that such combined MAb preparation may present substantial synergy in its potency over the calculated additive potency of its individual MAb components. Moreover, oligoclonal preparation is expected to be better suited to compensating for reduced efficacy due to epitope variation. In this review, the synergistic neutralization properties of combined oligoclonal Ab preparations are described. The effect of Ab affinity, autologous Fc fraction, and targeting a critical number of epitopes, as well as the unexpected contribution of non-neutralizing clones to the synergistic neutralizing effect are presented and discussed.
Collapse
Affiliation(s)
- Eran Diamant
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Amram Torgeman
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Eyal Ozeri
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Ran Zichel
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| |
Collapse
|
36
|
Alzuguren P, Hervas-Stubbs S, Gonzalez-Aseguinolaza G, Poutou J, Fortes P, Mancheno U, Bunuales M, Olagüe C, Razquin N, Van Rooijen N, Enguita M, Hernandez-Alcoceba R. Transient depletion of specific immune cell populations to improve adenovirus-mediated transgene expression in the liver. Liver Int 2015; 35:1274-89. [PMID: 24754307 DOI: 10.1111/liv.12571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 04/17/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Adenoviral (Ad) vectors are currently one of the most efficient tools for in vivo gene transfer to the liver. However, anti-Ad immune responses limit the safety and efficacy of these vectors. The initial inflammatory reaction is a concern in terms of toxicity, and it favours the development of cellular and humoral responses leading to short transgene persistence and inefficient vector re-administrations. Therefore, safe and simple ways to interfere with these processes are needed. Study ways to deplete specific immune cell populations and their impact on liver-directed gene transfer. METHODS First-generation Ad vectors encoding reporter genes (luciferase or β-galactosidase) were injected intravenously into Balb/c mice. Kupffer cells and splenic macrophages were depleted by intravenous administration of clodronate liposomes. B lymphocytes, CD4(+) , CD8(+) T lymphocytes or NK cells were depleted by intraperitoneal injection of anti-M plus anti-D, anti-CD4, anti-CD8 or anti-asialo-GM1 antibodies respectively. Long-term evolution of luciferase expression in the liver was monitored by bioluminescence imaging. RESULTS The anti-CD4 monoclonal antibody impaired cellular and humoral immune responses, leading to efficient vector re-administration. Clodronate liposomes had no impact on humoral responses but caused a 100-1000 fold increase in liver transduction, stabilized transgene expression, reduced the concentration of inflammatory cytokines, and inhibited lymphocyte activation. CONCLUSIONS Transient CD4(+) T-cell depletion using antibodies is a clinically feasible procedure that allows efficient Ad redosing. Systemic administration of clodronate liposomes may further increase the safety and efficacy of vectors.
Collapse
Affiliation(s)
- Pilar Alzuguren
- Division of Hepatology and Gene Therapy, CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kügler J, Wilke S, Meier D, Tomszak F, Frenzel A, Schirrmann T, Dübel S, Garritsen H, Hock B, Toleikis L, Schütte M, Hust M. Generation and analysis of the improved human HAL9/10 antibody phage display libraries. BMC Biotechnol 2015; 15:10. [PMID: 25888378 PMCID: PMC4352240 DOI: 10.1186/s12896-015-0125-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/09/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Antibody phage display is a proven key technology that allows the generation of human antibodies for diagnostics and therapy. From naive antibody gene libraries - in theory - antibodies against any target can be selected. Here we describe the design, construction and characterization of an optimized antibody phage display library. RESULTS The naive antibody gene libraries HAL9 and HAL10, with a combined theoretical diversity of 1.5×10(10) independent clones, were constructed from 98 healthy donors using improved phage display vectors. In detail, most common phagemids employed for antibody phage display are using a combined His/Myc tag for detection and purification. We show that changing the tag order to Myc/His improved the production of soluble antibodies, but did not affect antibody phage display. For several published antibody libraries, the selected number of kappa scFvs were lower compared to lambda scFvs, probably due to a lower kappa scFv or Fab expression rate. Deletion of a phenylalanine at the end of the CL linker sequence in our new phagemid design increased scFv production rate and frequency of selected kappa antibodies significantly. The HAL libraries and 834 antibodies selected against 121 targets were analyzed regarding the used germline V-genes, used V-gene combinations and CDR-H3/-L3 length and composition. The amino acid diversity and distribution in the CDR-H3 of the initial library was retrieved in the CDR-H3 of selected antibodies showing that all CDR-H3 amino acids occurring in the human antibody repertoire can be functionally used and is not biased by E. coli expression or phage selection. Further, the data underline the importance of CDR length variations. CONCLUSION The highly diverse universal antibody gene libraries HAL9/10 were constructed using an optimized scFv phagemid vector design. Analysis of selected antibodies revealed that the complete amino acid diversity in the CDR-H3 was also found in selected scFvs showing the functionality of the naive CDR-H3 diversity.
Collapse
Affiliation(s)
- Jonas Kügler
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,mAb-factory GmbH, Gelsenkirchenstr. 5, 38108, Braunschweig, Germany.
| | - Sonja Wilke
- mAb-factory GmbH, Gelsenkirchenstr. 5, 38108, Braunschweig, Germany.
| | - Doris Meier
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - Florian Tomszak
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - André Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany.
| | - Thomas Schirrmann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany.
| | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - Henk Garritsen
- Klinikum Braunschweig g GmbH, Institut für Klinische Transfusionsmedizin, Celler Str. 38, 38114, Braunschweig, Germany. .,Department Vaccinology, Helmholtz-Zentrum für Infektionsforschung, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| | | | | | | | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| |
Collapse
|
38
|
Majumdar R, Middaugh C, Weis DD, Volkin DB. Hydrogen-Deuterium Exchange Mass Spectrometry as an Emerging Analytical Tool for Stabilization and Formulation Development of Therapeutic Monoclonal Antibodies. J Pharm Sci 2015; 104:327-45. [DOI: 10.1002/jps.24224] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 12/11/2022]
|
39
|
Jones RGA, Martino A. Targeted localized use of therapeutic antibodies: a review of non-systemic, topical and oral applications. Crit Rev Biotechnol 2015; 36:506-20. [PMID: 25600465 DOI: 10.3109/07388551.2014.992388] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic antibodies provide important tools in the "medicine chest" of today's clinician for the treatment of a range of disorders. Typically monoclonal or polyclonal antibodies are administered in large doses, either directly or indirectly into the circulation, via a systemic route which is well suited for disseminated ailments. Diseases confined within a specific localized tissue, however, may be treated more effectively and at reduced cost by a delivery system which targets directly the affected area. To explore the advantages of the local administration of antibodies, we reviewed current alternative, non-systemic delivery approaches which are in clinical use, being trialed or developed. These less conventional approaches comprise: (a) local injections, (b) topical and (c) peroral administration routes. Local delivery includes intra-ocular injections into the vitreal humor (i.e. Ranibizumab for age-related macular degeneration), subconjunctival injections (e.g. Bevacizumab for corneal neovascularization), intra-articular joint injections (i.e. anti-TNF alpha antibody for persistent inflammatory monoarthritis) and intratumoral or peritumoral injections (e.g. Ipilimumab for cancer). A range of other strategies, such as the local use of antibacterial antibodies, are also presented. Local injections of antibodies utilize doses which range from 1/10th to 1/100th of the required systemic dose therefore reducing both side-effects and treatment costs. In addition, any therapeutic antibody escaping from the local site of disease into the systemic circulation is immediately diluted within the large blood volume, further lowering the potential for unwanted effects. Needle-free topical application routes become an option when the condition is restricted locally to an external surface. The topical route may potentially be utilized in the form of eye drops for infections or corneal neovascularization or be applied to diseased skin for psoriasis, dermatitis, pyoderma gangrenosum, antibiotic resistant bacterial infections or ulcerated wounds. Diseases confined to the gastrointestinal tract can be targeted directly by applying antibody via the injection-free peroral route. The gastrointestinal tract is unusual in that its natural immuno-tolerant nature ensures the long-term safety of repeatedly ingesting heterologous antiserum or antibody materials. Without the stringent regulatory, purity and clean room requirements of manufacturing parenteral (injectable) antibodies, production costs are minimal, with the potential for more direct low-cost targeting of gastrointestinal diseases, especially with those caused by problematic antibiotic resistant or toxigenic bacteria (e.g. Clostridium difficile, Helicobacter pylori), viruses (e.g. rotavirus, norovirus) or inflammatory bowel disease (e.g. ulcerative colitis, Crohn's disease). Use of the oral route has previously been hindered by excessive antibody digestion within the gastrointestinal tract; however, this limitation may be overcome by intelligently applying one or more strategies (i.e. decoy proteins, masking therapeutic antibody cleavage sites, pH modulation, enzyme inhibition or encapsulation). These aspects are additionally discussed in this review and novel insights also provided. With the development of new applications via local injections, topical and peroral routes, it is envisaged that an extended range of ailments will increasingly fall within the clinical scope of therapeutic antibodies further expanding this market.
Collapse
Affiliation(s)
| | - Angela Martino
- a Department of Chemistry , University of Warwick , Coventry , UK
| |
Collapse
|
40
|
Quantification of Patient-Specific Assay Interference in Different Formats of Enzyme-Linked Immunoassays for Therapeutic Monoclonal Antibodies. Ther Drug Monit 2014; 36:765-70. [DOI: 10.1097/ftd.0000000000000090] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Figueira TN, Veiga AS, Castanho MA. The interaction of antibodies with lipid membranes unraveled by fluorescence methodologies. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2014.02.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Joshi V, Shivach T, Yadav N, Rathore AS. Circular Dichroism Spectroscopy as a Tool for Monitoring Aggregation in Monoclonal Antibody Therapeutics. Anal Chem 2014; 86:11606-13. [DOI: 10.1021/ac503140j] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Varsha Joshi
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi 110016, India
| | - Tarun Shivach
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi 110016, India
| | - Nitin Yadav
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi 110016, India
| | - Anurag S. Rathore
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
43
|
|
44
|
Podhorecka M, Markowicz J, Szymczyk A, Pawlowski J. Target Therapy in Hematological Malignances: New Monoclonal Antibodies. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:701493. [PMID: 27433507 PMCID: PMC4897146 DOI: 10.1155/2014/701493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 11/17/2022]
Abstract
Apart from radio- and chemotherapy, monoclonal antibodies (MoAbs) represent a new, more selective tool in the treatment of hematological malignancies. MoAbs bind with the specific antigens of the tumors. This interaction is a basis for targeted therapies which exhibit few side effects and significant antitumor activity. This review provides an overview of the functional characteristics of MoAbs, with some examples of their clinical application. The promising results in the treatment of hematological malignancies have led to the more frequent usage of MoAbs in the therapy. Development of MoAbs is a subject of extensive research. They are a promising method of cancer treatment in the future.
Collapse
Affiliation(s)
- Monika Podhorecka
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Justyna Markowicz
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Szymczyk
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Johannes Pawlowski
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
45
|
Josephs DH, Spicer JF, Karagiannis P, Gould HJ, Karagiannis SN. IgE immunotherapy: a novel concept with promise for the treatment of cancer. MAbs 2014; 6:54-72. [PMID: 24423620 DOI: 10.4161/mabs.27029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The importance of antibodies in activating immune responses against tumors is now better appreciated with the emergence of checkpoint blockade antibodies and with engineered antibody Fc domains featuring enhanced capacity to focus potent effector cells against cancer cells. Antibodies designed with Fc regions of the IgE class can confer natural, potent, long-lived immune surveillance in tissues through tenacious engagement of high-affinity cognate Fc receptors on distinct, often tumor-resident immune effector cells, and through ability to activate these cells under tumor-induced Th2-biased conditions. Here, we review the properties that make IgE a contributor to the allergic response and a critical player in the protection against parasites, which also support IgE as a novel anti-cancer modality. We discuss IgE-based active and passive immunotherapeutic approaches in disparate in vitro and in vivo model systems, collectively suggesting the potential of IgE immunotherapies in oncology. Translation toward clinical application is now in progress.
Collapse
Affiliation(s)
- Debra H Josephs
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK; Division of Cancer Studies; King's College London; Guy's Hospital; London, UK
| | - James F Spicer
- Division of Cancer Studies; King's College London; Guy's Hospital; London, UK
| | - Panagiotis Karagiannis
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK
| | - Hannah J Gould
- Randall Division of Cell and Molecular Biophysics & Division of Asthma; Allergy and Lung Biology; MRC and Asthma UK Centre for Allergic Mechanisms of Asthma; King's College London; Guy's Campus; London, UK
| | - Sophia N Karagiannis
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK
| |
Collapse
|
46
|
Schubert I, Saul D, Nowecki S, Mackensen A, Fey GH, Oduncu FS. A dual-targeting triplebody mediates preferential redirected lysis of antigen double-positive over single-positive leukemic cells. MAbs 2014; 6:286-96. [PMID: 24135631 DOI: 10.4161/mabs.26768] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The single-chain triplebody HLA-ds16-hu19 consists of three single-chain Fv (scFv) antibody fragments connected in a single polypeptide chain. This protein with dual-targeting capacity mediated preferential lysis of antigen double positive(dp) over single-positive (sp) leukemic cells by recruitment of natural killer (NK) cells as effectors. The two distal scFv modules were specific for the histocompatibility protein HLA-DR and the lymphoid antigen CD19, the central one for the Fc gamma receptor CD16. In antibody-dependent cellular cytotoxicity (ADCC) experiments with a mixture of leukemic target cells comprising both HLA-DR sp HuT-78 or Kasumi-1 cells and (HLA-DR plus CD19) dp SEM cells, the triplebody mediated preferential lysis of the dp cells even when the sp cells were present in ≤ 20-fold numerical excess.The triplebody promoted equal lysis of SEM cells at 2.5-fold and 19.5-fold lower concentrations than the parental antibodies specific for HLA-DR and CD19, respectively. Finally, the triplebody also eliminated primary leukemic cells at lower concentrations than an equimolar mixture of bispecific single-chain Fv fragments (bsscFvs) separately addressing each target antigen (hu19-ds16 and HLA-ds16). The increased selectivity of targeting and the preferential lysis of dp over sp cells achieved by dual-targeting open attractive new perspectives for the use of dual-targeting agents in cancer therapy.
Collapse
|
47
|
Chan CEZ, Lim APC, MacAry PA, Hanson BJ. The role of phage display in therapeutic antibody discovery. Int Immunol 2014; 26:649-57. [PMID: 25135889 PMCID: PMC7185696 DOI: 10.1093/intimm/dxu082] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Phage display involves the expression of selected proteins on the surface of filamentous phage through fusion with phage coat protein, with the genetic sequence packaged within, linking phenotype to genotype selection. When combined with antibody libraries, phage display allows for rapid in vitro selection of antigen-specific antibodies and recovery of their corresponding coding sequence. Large non-immune and synthetic human libraries have been constructed as well as smaller immune libraries based on capturing a single individual’s immune repertoire. This completely in vitro process allows for isolation of antibodies against poorly immunogenic targets as well as those that cannot be obtained by animal immunization, thus further expanding the utility of the approach. Phage antibody display represents the first developed methodology for high throughput screening for human therapeutic antibody candidates. Recently, other methods have been developed for generation of fully human therapeutic antibodies, such as single B-cell screening, next-generation genome sequencing and transgenic mice with human germline B-cell genes. While each of these have their particular advantages, phage display has remained a key methodology for human antibody discovery due its in vitro process. Here, we review the continuing role of this technique alongside other developing technologies for therapeutic antibody discovery.
Collapse
Affiliation(s)
- Conrad E Z Chan
- Biological Defence Program, Defense Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Angeline P C Lim
- Biological Defence Program, Defense Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Paul A MacAry
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore Immunology Program, Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Brendon J Hanson
- Biological Defence Program, Defense Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
| |
Collapse
|
48
|
Hamakubo T, Kusano-Arai O, Iwanari H. Generation of antibodies against membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1920-1924. [PMID: 25135856 DOI: 10.1016/j.bbapap.2014.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 07/30/2014] [Accepted: 08/12/2014] [Indexed: 12/12/2022]
Abstract
The monoclonal antibody has become an important therapeutic in the treatment of both hematological malignancies and solid tumors. The recent success of antibody-drug conjugates (ADCs) has broadened the extent of the potential target molecules in cancer immunotherapy. As a result, even molecules of low abundance have become targets for cytotoxic reagents. The multi-pass membrane proteins are an emerging target for the next generation antibody therapeutics. One outstanding challenge is the difficulty in preparing a sufficient amount of these membrane proteins so as to be able to generate the functional antibody. We have pursued the expression of various membrane proteins on the baculovirus particle and the utilization of displayed protein for immunization. The strong antigenicity of the virus acts either as a friend or foe in the making of an efficient antibody against an immunologically tolerant antigen. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.
Collapse
Affiliation(s)
- Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro, Tokyo 153-8904, Japan.
| | - Osamu Kusano-Arai
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro, Tokyo 153-8904, Japan; Institute of Immunology Co. Ltd, .1-1-10 Koraku, Bunkyo, Tokyo 112-0004, Japan
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro, Tokyo 153-8904, Japan
| |
Collapse
|
49
|
Ju MS, Jung ST. Aglycosylated full-length IgG antibodies: steps toward next-generation immunotherapeutics. Curr Opin Biotechnol 2014; 30:128-39. [PMID: 25035939 DOI: 10.1016/j.copbio.2014.06.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/17/2014] [Accepted: 06/15/2014] [Indexed: 12/17/2022]
Abstract
Albeit the removal of Asn297 glycans of IgG perturbs the overall conformation and flexibility of the IgG CH2 domain, resulting in the loss of Fc-ligand interactions and therapeutically critical immune effector functions, aglycosylated full-length IgG antibodies are nearly identical to the glycosylated counterparts in terms of antigen binding, stability at physiological or low temperature conditions, pharmacokinetics, and biodistribution. To bypass the drawbacks of glycosylated antibodies that include glycan heterogeneity and requirement of high capital investment for biomanufacturing, aglycosylated antibodies have been developed and several are under clinical trials. Comprehensive cellular and bioprocess engineering has enabled to produce highly complex aglycosylated IgGs in a simple bacterial cultivation with comparable production level as that of mammalian cells. Moreover, extensive engineering of aglycosylated Fc has converted the aglycosylated IgG antibodies into a new class of effector functional human immunotherapeutics.
Collapse
Affiliation(s)
- Man-Seok Ju
- Department of Bio and Nano Chemistry, Kookmin University, Seoul 136-702, Republic of Korea
| | - Sang Taek Jung
- Department of Bio and Nano Chemistry, Kookmin University, Seoul 136-702, Republic of Korea.
| |
Collapse
|
50
|
Wållberg H, Ståhl S. Design and evaluation of radiolabeled tracers for tumor imaging. Biotechnol Appl Biochem 2014; 60:365-83. [PMID: 24033592 DOI: 10.1002/bab.1111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/20/2013] [Indexed: 12/22/2022]
Abstract
The growing understanding of tumor biology and the identification of tumor-specific genetic and molecular alterations, such as the overexpression of membrane receptors and other proteins, allows for personalization of patient management using targeted therapies. However, this puts stringent demands on the diagnostic tools used to identify patients who are likely to respond to a particular treatment. Radionuclide molecular imaging is a promising noninvasive method to visualize and characterize the expression of such targets. A number of different proteins, from full-length antibodies and their derivatives to small scaffold proteins and peptide receptor-ligands, have been applied to molecular imaging, each demonstrating strengths and weaknesses. Here, we discuss the concept of molecular targeting and, in particular, molecular imaging of cancer-associated targets. Additionally, we describe important biotechnological considerations and desired features when designing and developing tracers for radionuclide molecular imaging.
Collapse
Affiliation(s)
- Helena Wållberg
- Division of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|