51
|
Liu-Shin LPY, Fung A, Malhotra A, Ratnaswamy G. Evidence of disulfide bond scrambling during production of an antibody-drug conjugate. MAbs 2018; 10:1190-1199. [PMID: 30339473 DOI: 10.1080/19420862.2018.1521128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Antibody-drug conjugates (ADCs) that are formed using thiol-maleimide chemistry are commonly produced by reactions that occur at or above neutral pHs. Alkaline environments can promote disulfide bond scrambling, and may result in the reconfiguration of interchain disulfide bonds in IgG antibodies, particularly in the IgG2 and IgG4 subclasses. IgG2-A and IgG2-B antibodies generated under basic conditions yielded ADCs with comparable average drug-to-antibody ratios and conjugate distributions. In contrast, the antibody disulfide configuration affected the distribution of ADCs generated under acidic conditions. The similarities of the ADCs derived from alkaline reactions were attributed to the scrambling of interchain disulfide bonds during the partial reduction step, where conversion of the IgG2-A isoform to the IgG2-B isoform was favored.
Collapse
Affiliation(s)
- Lily Pei-Yao Liu-Shin
- a Analytical and Formulation Development , Agensys, Inc., an affiliate of Astellas, Inc , Santa Monica , CA , USA.,b Department of Biochemistry and Molecular Biology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Adam Fung
- a Analytical and Formulation Development , Agensys, Inc., an affiliate of Astellas, Inc , Santa Monica , CA , USA
| | - Arun Malhotra
- b Department of Biochemistry and Molecular Biology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Gayathri Ratnaswamy
- a Analytical and Formulation Development , Agensys, Inc., an affiliate of Astellas, Inc , Santa Monica , CA , USA
| |
Collapse
|
52
|
Parslow AC, Clayton AHA, Lock P, Scott AM. Confocal Microscopy Reveals Cell Surface Receptor Aggregation Through Image Correlation Spectroscopy. J Vis Exp 2018:57164. [PMID: 30124657 PMCID: PMC6126602 DOI: 10.3791/57164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Confocal microscopy provides an accessible methodology to capture sub-cellular interactions critical for the characterization and further development of pre-clinical agents labeled with fluorescent probes. With recent advancements in antibody based cytotoxic drug delivery systems, understanding the alterations induced by these agents within the realm of receptor aggregation and internalization is of critical importance. This protocol leverages the well-established methodology of fluorescent immunocytochemistry and the open source FIJI distribution of ImageJ, with its inbuilt autocorrelation and image mathematical functions, to perform spatial image correlation spectroscopy (ICS). This protocol quantitates the fluorescent intensity of labeled receptors as a function of the beam area of the confocal microscope. This provides a quantitative measure of the state of target molecule aggregation on the cell surface. This methodology is focused on the characterization of static cells with potential to expand into temporal investigations of receptor aggregation. This protocol presents an accessible methodology to provide quantification of clustering events occurring at the cell surface, utilizing well established techniques and non-specialized imaging apparatus.
Collapse
Affiliation(s)
- Adam C Parslow
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute; School of Cancer Medicine, La Trobe University
| | - Andrew H A Clayton
- Centre for Micro-Photonics, Faculty of Science, Engineering and Technology, Swinburne University of Technology
| | - Peter Lock
- LIMS Bioimaging Facility, La Trobe Institute for Molecular Science, La Trobe University
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute; School of Cancer Medicine, La Trobe University; Department of Medical Oncology, Olivia Newton-John Cancer and Wellnes Centre, Austin Health; Department of Medicine, University of Melbourne; Department of Molecular Imaging and Therapy, Austin Health;
| |
Collapse
|
53
|
Effective cancer therapy based on selective drug delivery into cells across their membrane using receptor-mediated endocytosis. Bioorg Med Chem Lett 2018; 28:3015-3024. [PMID: 30031619 DOI: 10.1016/j.bmcl.2018.07.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/28/2018] [Accepted: 07/04/2018] [Indexed: 01/21/2023]
Abstract
Cancer is one of the major causes of death globally. The current treatment options are insufficient, leading to unmet medical needs in cancer treatment. Off-target side effects, multidrug resistance, selective distribution to cancerous tissues, and cell membrane permeation of anti-cancer agents are critical problems to overcome. There is a method to solve these problems by using receptor-mediated endocytosis (RME). It is well known that proteins such as integrin, HER2, EGFR, or other cancer biomarkers are specifically overexpressed on the surface of target cancer cells. By taking advantage of such specific receptors, payloads can be transported into cells through endocytosis using a conjugate composed of the corresponding ligands connected to the payloads by an appropriate linker. After RME, the payloads released by endosomal escape into the cytoplasm can exhibit the cytotoxic activity against cancer cells. Cell-penetrating peptides (CPPs), tumor-homing peptides (THPs), and monoclonal antibodies (mAbs) are utilized as ligands in this system. Antibody drug conjugates (ADCs) based on RME have already been used to cure cancer. In addition to the canonical conjugate method, nanocarriers for spontaneous accumulation in cancer tissue due to enhanced permeability and retention (EPR) effect are extensively used. In this review, I introduce the possibilities and advantages of drug design and development based on RME for the treatment of cancer.
Collapse
|
54
|
Moore KN, Vergote I, Oaknin A, Colombo N, Banerjee S, Oza A, Pautier P, Malek K, Birrer MJ. FORWARD I: a Phase III study of mirvetuximab soravtansine versus chemotherapy in platinum-resistant ovarian cancer. Future Oncol 2018; 14:1669-1678. [DOI: 10.2217/fon-2017-0646] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Mirvetuximab soravtansine, an antibody–drug conjugate that binds with high affinity to folate receptor-α to provide tumor-directed delivery of the potent microtubule-disrupting agent DM4, has emerged as a promising investigational agent for the treatment of ovarian cancer, particularly in the setting of platinum-resistant disease. Here we describe the rationale and design of FORWARD I (NCT02631876), the first randomized, multicenter Phase III study to compare the safety and efficacy of mirvetuximab soravtansine versus investigator's choice of chemotherapy in women with folate receptor-α-positive, platinum-resistant epithelial ovarian, primary peritoneal or fallopian tube cancer. Patients will be randomized in a 2:1 ratio. The primary end point is progression-free survival, and key secondary objectives include comparison of overall response rates, overall survival and duration of response.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics & Gynecology, Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ignace Vergote
- Gynaecological Oncology, Leuven Cancer Institute, Leuven 3000, Belgium
| | - Ana Oaknin
- Medical Oncology Department, Vall D'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain
| | - Nicoletta Colombo
- Gynecologic Oncology, The European Institute of Oncology, Milan 20141, Italy
| | - Susana Banerjee
- Gynaecology Unit, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Amit Oza
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto M5G 2M9, Canada
| | - Patricia Pautier
- Department of Adult Medicine, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Karim Malek
- Clinical Development, ImmunoGen, Inc., Waltham, MA 02451, USA
| | - Michael J Birrer
- Division of Hematology–Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL 35294, USA
| |
Collapse
|
55
|
Bangalore Kumar A, Maus R, Markovic SN. Pharmacologic Modulation of Human Immunity in the Era of Immuno-oncology: Something Old, Something New. Mayo Clin Proc 2018; 93:917-936. [PMID: 29887221 DOI: 10.1016/j.mayocp.2018.03.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
The concept of exploiting the immune system to treat cancer forms the basis of immuno-oncology. Since its birth in the late 1800s, immuno-oncology, or cancer immunotherapy, has come a long way. With better understanding of the complex relationship between tumor and the immune system, we have been able to explore and develop various modalities of anticancer therapies. In this review, we summarize the main strategies of immunotherapy that are available today: monoclonal antibodies, anticancer vaccines, cytokines, and adoptive T-cell therapy. We also highlight the unique set of adverse effects associated with modern immunotherapy and propose nonsteroidal immunomodulators and anticytokine antibodies as treatment options for toxicities. The future of immuno-oncology is discussed, including combination therapy, drug-antibody conjugates, epigenetic drugs, using nanoparticles for drug delivery, new antigen discovery, and developing biomarkers to assess treatment responses. A data search was conducted using PubMed and included studies published through November 1, 2017. Search terms used include cancer immunotherapy, pembrolizumab, ipilimumab, nivolumab, PD-1 inhibitors, PD-L1 inhibitors, checkpoint inhibitors, anticancer vaccines, TVEC, and adoptive cell therapy.
Collapse
Affiliation(s)
| | - Rachel Maus
- Department of Medical Oncology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
56
|
Umlauf BJ, Mix KA, Grosskopf VA, Raines RT, Shusta EV. Site-Specific Antibody Functionalization Using Tetrazine-Styrene Cycloaddition. Bioconjug Chem 2018; 29:1605-1613. [PMID: 29694034 DOI: 10.1021/acs.bioconjchem.8b00114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biologics, such as antibody-drug conjugates, are becoming mainstream therapeutics. Consequently, methods to functionalize biologics without disrupting their native properties are essential for identifying, characterizing, and translating candidate biologics from the bench to clinical practice. Here, we present a method for site-specific, carboxy-terminal modification of single-chain antibody fragments (scFvs). ScFvs displayed on the surface of yeast were isolated and functionalized by combining intein-mediated expressed protein ligation (EPL) with inverse electron-demand Diels-Alder (IEDDA) cycloaddition using a styrene-tetrazine pair. The high thiol concentration required to trigger EPL can hinder the subsequent chemoselective ligation reactions; therefore, the EPL reaction was used to append styrene to the scFv, limiting tetrazine exposure to damaging thiols. Subsequently, the styrene-functionalized scFv was reacted with tetrazine-conjugated compounds in an IEDDA cycloaddition to generate functionalized scFvs that retain their native binding activity. Rapid functionalization of yeast surface-derived scFv in a site-directed manner could find utility in many downstream laboratory and preclinical applications.
Collapse
|
57
|
Rautio J, Meanwell NA, Di L, Hageman MJ. The expanding role of prodrugs in contemporary drug design and development. Nat Rev Drug Discov 2018; 17:559-587. [DOI: 10.1038/nrd.2018.46] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
58
|
Sioud M, Westby P, Vasovic V, Fløisand Y, Peng Q. Development of a new high-affinity human antibody with antitumor activity against solid and blood malignancies. FASEB J 2018; 32:5063-5077. [PMID: 29913558 DOI: 10.1096/fj.201701544r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
mAbs have emerged as a promising strategy for the treatment of cancer. However, in several malignancies, no effective antitumor mAbs are yet available. Identifying therapeutic mAbs that recognize common tumor antigens could render the treatment widely applicable. Here, a human single-chain variable fragment (scFv) antibody library was sequentially affinity selected against a panel of human cancer cell lines and an antibody fragment (named MS5) that bound to solid and blood cancer cells was identified. The MS5 scFv was fused to the human IgG1 Fc domain to generate an antibody (MS5-Fc fusion) that induced antibody-dependent cellular cytotoxicity and phagocytosis of cancer cells by macrophages. In addition, the MS5-Fc antibody bound to primary leukemia cells and induced antibody-dependent cellular cytotoxicity. In the majority of analyzed cancer cells, the MS5-Fc antibody induced cell surface redistribution of the receptor complexes, but not internalization, thus maximizing the accessibility of the IgG1 Fc domain to immune effector cells. In vitro stability studies showed that the MS5-Fc antibody was stable after 6 d of incubation in human serum, retaining ∼60% of its initial intact form. After intravenous injections, the antibody localized into tumor tissues and inhibited the growth of 3 different human tumor xenografts (breast, lymphoma, and leukemia). These antitumor effects were associated with tumor infiltration by macrophages and NK cells. In the Ramos B-cell lymphoma xenograft model, the MS5-Fc antibody exhibited a comparable antitumor effect as rituximab, a chimeric anti-CD20 IgG1 mAb. These results indicate that human antibodies with pan-cancer abilities can be generated from phage display libraries, and that the engineered MS5-Fc antibody could be an attractive agent for further clinical investigation.-Sioud, M., Westby, P., Vasovic, V., Fløisand, Y., Peng, Q. Development of a new high-affinity human antibody with antitumor activity against solid and blood malignancies.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Rikshospitalet-Radiumhospitalet, University Hospital, Oslo, Norway
| | - Phuong Westby
- Department of Cancer Immunology, Rikshospitalet-Radiumhospitalet, University Hospital, Oslo, Norway
| | - Vlada Vasovic
- Department of Pathology, Rikshospitalet-Radiumhospitalet, University Hospital, Oslo, Norway; and
| | - Yngvar Fløisand
- Department of Hematology, Rikshospitalet-Radiumhospitalet, University Hospital, Oslo, Norway
| | - Qian Peng
- Department of Pathology, Rikshospitalet-Radiumhospitalet, University Hospital, Oslo, Norway; and
| |
Collapse
|
59
|
Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, Chauhan SC. Antibody-Drug Conjugates for Cancer Therapy: Chemistry to Clinical Implications. Pharmaceuticals (Basel) 2018; 11:ph11020032. [PMID: 29642542 PMCID: PMC6027311 DOI: 10.3390/ph11020032] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023] Open
Abstract
Chemotherapy is one of the major therapeutic options for cancer treatment. Chemotherapy is often associated with a low therapeutic window due to its poor specificity towards tumor cells/tissues. Antibody-drug conjugate (ADC) technology may provide a potentially new therapeutic solution for cancer treatment. ADC technology uses an antibody-mediated delivery of cytotoxic drugs to the tumors in a targeted manner, while sparing normal cells. Such a targeted approach can improve the tumor-to-normal tissue selectivity and specificity in chemotherapy. Considering its importance in cancer treatment, we aim to review recent efforts for the design and development of ADCs. ADCs are mainly composed of an antibody, a cytotoxic payload, and a linker, which can offer selectivity against tumors, anti-cancer activity, and stability in systemic circulation. Therefore, we have reviewed recent updates and principal considerations behind ADC designs, which are not only based on the identification of target antigen, cytotoxic drug, and linker, but also on the drug-linker chemistry and conjugation site at the antibody. Our review focuses on site-specific conjugation methods for producing homogenous ADCs with constant drug-antibody ratio (DAR) in order to tackle several drawbacks that exists in conventional conjugation methods.
Collapse
Affiliation(s)
- Nirnoy Dan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Vivek K Kashyap
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sheema Khan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
60
|
Nasiri H, Valedkarimi Z, Aghebati‐Maleki L, Majidi J. Antibody‐drug conjugates: Promising and efficient tools for targeted cancer therapy. J Cell Physiol 2018; 233:6441-6457. [DOI: 10.1002/jcp.26435] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Hadi Nasiri
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Zahra Valedkarimi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Leili Aghebati‐Maleki
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Jafar Majidi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
61
|
Liu-Shin L, Fung A, Malhotra A, Ratnaswamy G. Influence of disulfide bond isoforms on drug conjugation sites in cysteine-linked IgG2 antibody-drug conjugates. MAbs 2018; 10:583-595. [PMID: 29436897 PMCID: PMC5973704 DOI: 10.1080/19420862.2018.1440165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cysteine-linked antibody-drug conjugates (ADCs) produced from IgG2 monoclonal antibodies (mAbs) are more heterogeneous than ADCs generated from IgG1 mAbs, as IgG2 ADCs are composed of a wider distribution of molecules, typically containing 0 – 12 drug-linkers per antibody. The three disulfide isoforms (A, A/B, and B) of IgG2 antibodies confer differences in solvent accessibilities of the interchain disulfides and contribute to the structural heterogeneity of cysteine-linked ADCs. ADCs derived from either IgG2-A or IgG2-B mAbs were compared to better understand the role of disulfide isoforms on attachment sites and distribution of conjugated species. Our characterization of these ADCs demonstrated that the disulfide configuration affects the kinetics of disulfide bond reduction, but has minimal effect on the primary sites of reduction. The IgG2-A mAbs yielded ADCs with higher drug-to-antibody ratios (DARs) due to the easier reduction of its interchain disulfides. However, hinge-region cysteines were the primary conjugation sites for both IgG2-A and IgG2-B mAbs.
Collapse
Affiliation(s)
- Lily Liu-Shin
- a Analytical and Formulation Development, Agensys, Inc., an affiliate of Astellas, Inc. , Santa Monica , CA.,b Department of Biochemistry and Molecular Biology , University of Miami Miller School of Medicine , Miami , FL
| | - Adam Fung
- a Analytical and Formulation Development, Agensys, Inc., an affiliate of Astellas, Inc. , Santa Monica , CA
| | - Arun Malhotra
- b Department of Biochemistry and Molecular Biology , University of Miami Miller School of Medicine , Miami , FL
| | - Gayathri Ratnaswamy
- a Analytical and Formulation Development, Agensys, Inc., an affiliate of Astellas, Inc. , Santa Monica , CA
| |
Collapse
|
62
|
Taplin S, Vashisht K, Walles M, Calise D, Kluwe W, Bouchard P, Johnson R. Hepatotoxicity with antibody maytansinoid conjugates: A review of preclinical and clinical findings. J Appl Toxicol 2018; 38:600-615. [DOI: 10.1002/jat.3582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Sarah Taplin
- Novartis Pharmaceuticals Inc.; East Hanover NJ USA
| | | | | | | | | | | | | |
Collapse
|
63
|
Wagh A, Song H, Zeng M, Tao L, Das TK. Challenges and new frontiers in analytical characterization of antibody-drug conjugates. MAbs 2018; 10:222-243. [PMID: 29293399 DOI: 10.1080/19420862.2017.1412025] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a growing class of biotherapeutics in which a potent small molecule is linked to an antibody. ADCs are highly complex and structurally heterogeneous, typically containing numerous product-related species. One of the most impactful steps in ADC development is the identification of critical quality attributes to determine product characteristics that may affect safety and efficacy. However, due to the additional complexity of ADCs relative to the parent antibodies, establishing a solid understanding of the major quality attributes and determining their criticality are a major undertaking in ADC development. Here, we review the development challenges, especially for reliable detection of quality attributes, citing literature and new data from our laboratories, highlight recent improvements in major analytical techniques for ADC characterization and control, and discuss newer techniques, such as two-dimensional liquid chromatography, that have potential to be included in analytical control strategies.
Collapse
Affiliation(s)
- Anil Wagh
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Hangtian Song
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Ming Zeng
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Li Tao
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Tapan K Das
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| |
Collapse
|
64
|
Moore KN, Martin LP, O'Malley DM, Matulonis UA, Konner JA, Vergote I, Ponte JF, Birrer MJ. A review of mirvetuximab soravtansine in the treatment of platinum-resistant ovarian cancer. Future Oncol 2018; 14:123-136. [DOI: 10.2217/fon-2017-0379] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Resistance to platinum-based therapy poses a significant clinical challenge for the management of advanced ovarian cancer, a leading cause of cancer mortality among women. Mirvetuximab soravtansine is a novel antibody–drug conjugate that targets folate receptor-α, a validated molecular target for therapeutic intervention in this disease. Here, we examine mirvetuximab soravtansine's mechanism of action and pharmacology, and review its clinical evaluation in ovarian cancer to date. We focus on the favorable tolerability and encouraging signals of efficacy that have emerged, most notably in patients with platinum-resistant disease. Ongoing Phase III monotherapy and Phase Ib/II combination trials evaluating its activity in the setting of platinum resistance are emphasized, which will help define its role in the evolving landscape of ovarian cancer therapy.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics & Gynecology, Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lainie P Martin
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - David M O'Malley
- Department of Obstetrics & Gynecology, The Ohio State University, Columbus, OH 43210, USA
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Jason A Konner
- Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ignace Vergote
- Gynaecological Oncology, Leuven Cancer Institute, Leuven 3000, Belgium
| | - Jose F Ponte
- Pharmacology, ImmunoGen, Inc, Waltham, MA 02451, USA
| | - Michael J Birrer
- Gillette Center for Gynecologic Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
65
|
Martin LP, Konner JA, Moore KN, Seward SM, Matulonis UA, Perez RP, Su Y, Berkenblit A, Ruiz-Soto R, Birrer MJ. Characterization of folate receptor alpha (FRα) expression in archival tumor and biopsy samples from relapsed epithelial ovarian cancer patients: A phase I expansion study of the FRα-targeting antibody-drug conjugate mirvetuximab soravtansine. Gynecol Oncol 2017; 147:402-407. [PMID: 28843653 PMCID: PMC6893864 DOI: 10.1016/j.ygyno.2017.08.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/08/2017] [Accepted: 08/12/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE To characterize folate receptor alpha (FRα) expression in archival and fresh biopsy tumor samples from relapsed ovarian cancer patients. METHODS Patients with ovarian tumors amenable to biopsy were eligible to enroll. Eligibility included a minimum requirement of FRα positivity in archival tumor samples (≥25% of cells with ≥2+ staining intensity). Patients received mirvetuximab soravtansine at 6mg/kg once every 3weeks. Core needle biopsies were collected before and after treatment and FRα levels assessed by immunohistochemistry. Descriptive statistics were used to summarize the association between receptor expression and response. RESULTS Twenty-seven heavily pre-treated patients were enrolled. Six individuals (22%) did not have evaluable pre-treatment biopsies due to insufficient tumor cells. The concordance of FRα expression in archival and biopsy tissues was 71%, and no major shifts in receptor expression were seen in matched pre- and post-treatment biopsy samples. Adverse events were generally mild (≤grade 2) with keratopathy (48%), fatigue (44%), diarrhea, and blurred vision (each 37%) being the most common treatment-related toxicities. The confirmed objective response rate (ORR) was 22%, including two complete responses and four partial responses. Superior efficacy measures were observed in the subset of patients with the highest FRα levels (ORR, 31%; progression-free survival, 5.4months). CONCLUSION Concordance of FRα expression in biopsy versus archival tumor samples suggests that archival tissue can reliably identify patients with receptor-positive tumors and is appropriate for patient selection in mirvetuximab soravtansine clinical trials. Regardless of the tissue source analyzed, higher FRα expression was associated with greater antitumor activity.
Collapse
Affiliation(s)
| | - Jason A Konner
- Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| | - Kathleen N Moore
- Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| | - Shelly M Seward
- Wayne State University - Karmanos Cancer Institute, Huntington Woods, MI, United States.
| | | | | | - Ying Su
- ImmunoGen, Inc., Waltham, MA, United States.
| | | | | | | |
Collapse
|
66
|
Bashari O, Redko B, Cohen A, Luboshits G, Gellerman G, Firer MA. Discovery of peptide drug carrier candidates for targeted multi-drug delivery into prostate cancer cells. Cancer Lett 2017; 408:164-173. [PMID: 28888997 DOI: 10.1016/j.canlet.2017.08.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 11/24/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains essentially incurable. Targeted Drug Delivery (TDD) systems may overcome the limitations of current mCRPC therapies. We describe the use of strict criteria to isolate novel prostate cancer cell targeting peptides that specifically deliver drugs into target cells. Phage from a libraries displaying 7mer peptides were exposed to PC-3 cells and only internalized phage were recovered. The ability of these phage to internalize into other prostate cancer cells (LNCaP, DU-145) was validated. The displayed peptides of selected phage clones were synthesized and their specificity for target cells was validated in vitro and in vivo. One peptide (P12) which specifically targeted PC-3 tumors in vivo was incorporated into mono-drug (Chlorambucil, Combretastatin or Camptothecin) and dual-drug (Chlorambucil/Combretastatin or Chlorambucil/Camptothecin) PDCs and the cytotoxic efficacy of these conjugates for target cells was tested. Conjugation of P12 into dual-drug PDCs allowed discovery of new drug combinations with synergistic effects. The use of strict selection criteria can lead to discovery of novel peptides for use as drug carriers for TDD. PDCs represent an effective alternative to current modes of free drug chemotherapy for prostate cancer.
Collapse
Affiliation(s)
- O Bashari
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - B Redko
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - A Cohen
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Luboshits
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Gellerman
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - M A Firer
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| |
Collapse
|
67
|
Woitok M, Klose D, Di Fiore S, Richter W, Stein C, Gresch G, Grieger E, Barth S, Fischer R, Kolberg K, Niesen J. Comparison of a mouse and a novel human scFv-SNAP-auristatin F drug conjugate with potent activity against EGFR-overexpressing human solid tumor cells. Onco Targets Ther 2017; 10:3313-3327. [PMID: 28740407 PMCID: PMC5505605 DOI: 10.2147/ott.s140492] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Antibody–drug conjugates (ADCs) can deliver toxins to specific targets such as tumor cells. They have shown promise in preclinical/clinical development but feature stoichiometrically undefined chemical linkages, and those based on full-size antibodies achieve only limited tumor penetration. SNAP-tag technology can overcome these challenges by conjugating benzylguanine-modified toxins to single-chain fragment variables (scFvs) with 1:1 stoichiometry while preserving antigen binding. Two (human and mouse) scFv-SNAP fusion proteins recognizing the epidermal growth factor receptor (EGFR) were expressed in HEK 293T cells. The purified fusion proteins were conjugated to auristatin F (AURIF). Binding activity was confirmed by flow cytometry/immunohistochemistry, and cytotoxic activity was confirmed by cell viability/apoptosis and cell cycle arrest assays, and a novel microtubule dynamics disassembly assay was performed. Both ADCs bound specifically to their target cells in vitro and ex vivo, indicating that the binding activity of the scFv-SNAP fusions was unaffected by conjugation to AURIF. Cytotoxic assays confirmed that the ADCs induced apoptosis and cell cycle arrest at nanomolar concentrations and microtubule disassembly. The SNAP-tag technology provides a platform for the development of novel ADCs with defined conjugation sites and stoichiometry. We achieved the stable and efficient linkage of AURIF to human or murine scFvs using the SNAP-tag technology, offering a strategy to improve the development of personalized medicines.
Collapse
Affiliation(s)
- Mira Woitok
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany.,Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Diana Klose
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Stefano Di Fiore
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | | | - Christoph Stein
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Gerrit Gresch
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Elena Grieger
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Stefan Barth
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany.,Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Katharina Kolberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| | - Judith Niesen
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany
| |
Collapse
|
68
|
Moore KN, Borghaei H, O'Malley DM, Jeong W, Seward SM, Bauer TM, Perez RP, Matulonis UA, Running KL, Zhang X, Ponte JF, Ruiz-Soto R, Birrer MJ. Phase 1 dose-escalation study of mirvetuximab soravtansine (IMGN853), a folate receptor α-targeting antibody-drug conjugate, in patients with solid tumors. Cancer 2017; 123:3080-3087. [PMID: 28440955 DOI: 10.1002/cncr.30736] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/02/2017] [Accepted: 03/20/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mirvetuximab soravtansine (IMGN853) is an antibody-drug conjugate that selectively targets folate receptor α (FRα). In this phase 1 dose-escalation study, the authors investigated IMGN853 in patients with FRα-positive solid tumors. METHODS Patients received IMGN853 on day 1 of a 21-day cycle (once every 3 weeks dosing), with cycles repeated until patients experienced dose-limiting toxicity or progression. Dose escalation commenced in single-patient cohorts for the first 4 planned dose levels and then followed a standard 3 + 3 scheme. The primary objectives were to determine the maximum tolerated dose and the recommended phase 2 dose. Secondary objectives were to determine safety and tolerability, to characterize the pharmacokinetic profile, and to describe preliminary clinical activity. RESULTS In total, 44 patients received treatment at doses escalating from 0.15 to 7.0 mg/kg. No meaningful drug accumulation was observed with the dosing regimen of once every 3 weeks. The most common treatment-related adverse events were fatigue, blurred vision, and diarrhea, the majority of which were grade 1 or 2. The dose-limiting toxicities observed were grade 3 hypophosphatemia (5.0 mg/kg) and grade 3 punctate keratitis (7.0 mg/kg). Two patients, both of whom were individuals with epithelial ovarian cancer, achieved confirmed tumor responses according to Response Evaluation Criteria in Solid Tumors 1.1, and each was a partial response. CONCLUSIONS IMGN853 demonstrated a manageable safety profile and encouraging preliminary clinical activity, particularly in patients with ovarian cancer. The results establish a recommended phase 2 dosing of 6.0 mg/kg (based on adjusted ideal body weight) once every 3 weeks. Cancer 2017. © 2017 American Cancer Society. Cancer 2017;123:3080-7. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - David M O'Malley
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Woondong Jeong
- Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas
| | - Shelly M Seward
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Raymond P Perez
- Division of Hematology/Oncology, University of Kansas, Fairway, Kansas
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | - Michael J Birrer
- Gillette Center for Gynecologic Oncology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|