151
|
Peters C, Brown S. Antibody-drug conjugates as novel anti-cancer chemotherapeutics. Biosci Rep 2015; 35:e00225. [PMID: 26182432 PMCID: PMC4613712 DOI: 10.1042/bsr20150089] [Citation(s) in RCA: 323] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/18/2015] [Accepted: 05/29/2015] [Indexed: 12/19/2022] Open
Abstract
Over the past couple of decades, antibody-drug conjugates (ADCs) have revolutionized the field of cancer chemotherapy. Unlike conventional treatments that damage healthy tissues upon dose escalation, ADCs utilize monoclonal antibodies (mAbs) to specifically bind tumour-associated target antigens and deliver a highly potent cytotoxic agent. The synergistic combination of mAbs conjugated to small-molecule chemotherapeutics, via a stable linker, has given rise to an extremely efficacious class of anti-cancer drugs with an already large and rapidly growing clinical pipeline. The primary objective of this paper is to review current knowledge and latest developments in the field of ADCs. Upon intravenous administration, ADCs bind to their target antigens and are internalized through receptor-mediated endocytosis. This facilitates the subsequent release of the cytotoxin, which eventually leads to apoptotic cell death of the cancer cell. The three components of ADCs (mAb, linker and cytotoxin) affect the efficacy and toxicity of the conjugate. Optimizing each one, while enhancing the functionality of the ADC as a whole, has been one of the major considerations of ADC design and development. In addition to these, the choice of clinically relevant targets and the position and number of linkages have also been the key determinants of ADC efficacy. The only marketed ADCs, brentuximab vedotin and trastuzumab emtansine (T-DM1), have demonstrated their use against both haematological and solid malignancies respectively. The success of future ADCs relies on improving target selection, increasing cytotoxin potency, developing innovative linkers and overcoming drug resistance. As more research is conducted to tackle these issues, ADCs are likely to become part of the future of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Christina Peters
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K
| | - Stuart Brown
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K.
| |
Collapse
|
152
|
Dua P, Hawkins E, van der Graaf PH. A Tutorial on Target-Mediated Drug Disposition (TMDD) Models. CPT Pharmacometrics Syst Pharmacol 2015; 4:324-37. [PMID: 26225261 PMCID: PMC4505827 DOI: 10.1002/psp4.41] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/07/2015] [Indexed: 12/16/2022] Open
Abstract
Target-mediated drug disposition (TMDD) is the phenomenon in which a drug binds with high affinity to its pharmacological target site (such as a receptor) to such an extent that this affects its pharmacokinetic characteristics.1 The aim of this Tutorial is to provide an introductory guide to the mathematical aspects of TMDD models for pharmaceutical researchers. Examples of Berkeley Madonna2 code for some models discussed in this Tutorial are provided in the Supplementary Materials.
Collapse
Affiliation(s)
- P Dua
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
| | - E Hawkins
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
- Department of Mathematics, University of SurreyGuildford, UK
| | - PH van der Graaf
- Leiden Academic Centre for Drug Research (LACDR), Systems PharmacologyLeiden, The Netherlands
| |
Collapse
|
153
|
Wohlrab J. Pharmakokinetische Besonderheiten von therapeutischen Antikörpern. J Dtsch Dermatol Ges 2015. [DOI: 10.1111/ddg.12648_suppl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Johannes Wohlrab
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie; Universitätsklinikum Halle (Saale) und Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg; Halle (Saale)
| |
Collapse
|
154
|
Zhang Y, Doshi S, Zhu M. Pharmacokinetics and pharmacodynamics of rilotumumab: a decade of experience in preclinical and clinical cancer research. Br J Clin Pharmacol 2015; 80:957-64. [PMID: 25912961 DOI: 10.1111/bcp.12663] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/06/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022] Open
Abstract
Rilotumumab is a fully human monoclonal antibody against hepatocyte growth factor, the only known ligand of the MET receptor. Over the last decade, rilotumumab has been extensively tested in preclinical studies and in clinical studies in a variety of cancer types. In this review, we examine the pharmacokinetic and pharmacodynamic data that have been collected in the rilotumumab programme to date, and discuss retrospectively how the knowledge acquired in this programme can be applied to a number of key issues in oncology drug development, including: (i) using preclinical data to inform first-in-human study design; (ii) the role of biomarkers in the identification of a target patient population; (iii) the potential for drug interactions between therapeutic proteins and other anticancer agents; and (iv) pharmacokinetic and pharmacodynamic considerations in phase 3 study design.
Collapse
Affiliation(s)
- Y Zhang
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - S Doshi
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - M Zhu
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| |
Collapse
|
155
|
Shen H, Shi S, Zhang Z, Gong T, Sun X. Coating Solid Lipid Nanoparticles with Hyaluronic Acid Enhances Antitumor Activity against Melanoma Stem-like Cells. Theranostics 2015; 5:755-71. [PMID: 25897340 PMCID: PMC4402499 DOI: 10.7150/thno.10804] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/17/2015] [Indexed: 01/05/2023] Open
Abstract
Successful anticancer chemotherapy requires targeting tumors efficiently and further potential to eliminate cancer stem cell (CSC) subpopulations. Since CD44 is present on many types of CSCs, and it binds specially to hyaluronic acid (HA), we tested whether coating solid lipid nanoparticles with hyaluronan (HA-SLNs)would allow targeted delivery of paclitaxel (PTX) to CD44-overexpressing B16F10 melanoma cells. First, we developed a model system based on melanoma stem-like cells for experiments in vitro and in mouse xenografts, and we showed that cells expressing high levels of CD44 (CD44+) displayed a strong CSC phenotype while cells expressing low levels of CD44 (CD44-) did not. This phenotype included sphere and colony formation, higher proportion of side population cells, expression of CSC-related markers (ALDH, CD133, Oct-4) and tumorigenicity in vivo. Next we showed that administering PTX-loaded HA-SLNs led to efficient intracellular delivery of PTX and induced substantial apoptosis in CD44+ cells in vitro. In the B16F10-CD44+ lung metastasis model, PTX-loaded HA-SLNs targeted the tumor-bearing lung tissues well and subsequently exhibited significant antitumor effects with a relative low dose of PTX, which provided significant survival benefit without evidence of adverse events. These findings suggest that the HA-SLNs targeting system shows promise for enhancing cancer therapy.
Collapse
|
156
|
Panoilia E, Schindler E, Samantas E, Aravantinos G, Kalofonos HP, Christodoulou C, Patrinos GP, Friberg LE, Sivolapenko G. A pharmacokinetic binding model for bevacizumab and VEGF165 in colorectal cancer patients. Cancer Chemother Pharmacol 2015; 75:791-803. [PMID: 25687989 PMCID: PMC4365273 DOI: 10.1007/s00280-015-2701-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/10/2015] [Indexed: 12/30/2022]
Abstract
PURPOSE To characterize the population pharmacokinetics of bevacizumab, its binding properties to VEGF165 and the effect of demographic data and VEGF-A polymorphisms on the interplay between bevacizumab serum pharmacokinetics and VEGF165 serum concentrations in patients with colorectal cancer stage IV. METHODS Bevacizumab and VEGF165 data were collected from 19 adult patients with metastatic colorectal cancer enrolled in an observational clinical study. Bevacizumab was administered with one of the following combinations: 5-FU/Leucovorin/Irinotecan, 5-FU/Leucovorin/Oxaliplatin, Capecitabine/Irinotecan at doses ranging from 5 to 10 mg/kg every 2 or 3 weeks. Data analysis was performed using nonlinear mixed-effects modeling implemented in NONMEM 7.3. RESULTS A target-mediated drug disposition model adequately described bevacizumab concentration changes over time and its binding characteristics to VEGF165. The estimated clearance of bevacizumab was 0.18 L/day, the free VEGF165 levels at baseline were 212 ng/L, and the elimination rate constant of free VEGF165 was 0.401 day(-1). Body weight was allometrically included in all PK parameters. CONCLUSION The final model adequately described the pre- and post-dose concentrations of total bevacizumab and free VEGF165 in patients with colorectal cancer. Model parameters were consistent with those previously reported for patients with solid tumors. Correlations between the binding affinity of bevacizumab and the VEGF-2578C/A and VEGF-634G/C polymorphisms were noticed.
Collapse
MESH Headings
- Adult
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/blood
- Angiogenesis Inhibitors/pharmacokinetics
- Angiogenesis Inhibitors/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/blood
- Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bevacizumab
- Colorectal Neoplasms/blood
- Colorectal Neoplasms/drug therapy
- Drug Administration Schedule
- Female
- Humans
- Male
- Middle Aged
- Models, Biological
- Polymorphism, Single Nucleotide
- Protein Binding
- Vascular Endothelial Growth Factor A/blood
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Eirini Panoilia
- Department of Pharmacy, University of Patras, Rio-Patras, Greece,
| | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Rodríguez-Vera L, Ramos-Suzarte M, Fernández-Sánchez E, Soriano JL, Guitart CP, Hernández GC, Jacobo-Cabral CO, de Castro Suárez N, Codina HC. Semimechanistic model to characterize nonlinear pharmacokinetics of nimotuzumab in patients with advanced breast cancer. J Clin Pharmacol 2015; 55:888-98. [PMID: 25760761 DOI: 10.1002/jcph.496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study aimed (1) to develop a semimechanistic pharmacokinetic (PK) model for nimotuzumab in patients with advanced breast cancer and (2) to identify demographic, biochemical, and clinical predictive factors of the PK variability. Data from a phase 1 study were analyzed using the nonlinear mixed-effects approach (NONMEM). A target-mediated disposition model that included 2 open PK compartments, the monoclonal antibody (mAb)-target binding, and target and mAb-target complex turnovers best described the linear and nonlinear PK. Covariates had no influence on the PK parameters. The final parameter estimates were 19.93 L (steady-state volume), 0.0045-0.0172 L/h (range of total clearance values), 6.96 μg/mL (steady-state binding constant), 5.50 h(-1) (target degradation rate constant), 1.43 (μg/mL) · h(-1) (complex formation rate), and 0.148 h(-1) (complex internalization rate constant). The model described the effect of the mAb-target binding, and target and mAb-target complex turnovers on nimotuzumab PK. Simulations showed that doses above 200 mg maintained the 50% target occupancy during all of the treatment. This model can be very useful for knowing the dosing schedules required for efficacy and supports further investigation of the pharmacokinetic/pharmacodynamic relationships of nimotuzumab to improve its therapeutic use.
Collapse
Affiliation(s)
- Leyanis Rodríguez-Vera
- Laboratory of Pharmacokinetic, Department of Pharmacology & Clinical Pharmacy, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | | | - Eduardo Fernández-Sánchez
- Center for Biological Evaluation and Research, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | | | - Concepción Peraire Guitart
- Pharmacy and Pharmaceutical Technology Department, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | | | | | - Niurys de Castro Suárez
- Laboratory of Pharmacokinetic, Department of Pharmacology & Clinical Pharmacy, Institute of Pharmacy & Foods, University of Havana, Havana, Cuba
| | - Helena Colom Codina
- Pharmacy and Pharmaceutical Technology Department, School of Pharmacy, University of Barcelona, Barcelona, Spain
| |
Collapse
|
158
|
Glassman PM, Abuqayyas L, Balthasar JP. Assessments of antibody biodistribution. J Clin Pharmacol 2015; 55 Suppl 3:S29-38. [DOI: 10.1002/jcph.365] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/14/2014] [Indexed: 01/24/2023]
Affiliation(s)
- Patrick M. Glassman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences; University at Buffalo, The State University of New York; Buffalo NY 14214 USA
| | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences; University at Buffalo, The State University of New York; Buffalo NY 14214 USA
| |
Collapse
|
159
|
Pharmaceutical aspects of anti-inflammatory TNF-blocking drugs. Inflammopharmacology 2015; 23:71-7. [PMID: 25687751 DOI: 10.1007/s10787-015-0229-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 01/24/2015] [Indexed: 12/13/2022]
Abstract
Tumor necrosis factor (TNF) is a key regulator of inflammatory processes in several immune-mediated inflammatory diseases such as rheumatoid arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, psoriasis and psoriatic arthritis. Inactivating TNF has been found to be a plausible approach in treating these conditions. Two major strategies have been adopted by scientists to inactivate TNF: one is to use monoclonal antibodies (mAbs) that bind to TNF, and the other is to use fusion proteins that bind to TNF, both inactivate TNF and help to prevent TNF-mediated inflammatory processes. Monoclonal antibodies (mAbs) are biological products that selectively bind to specific antigen molecules, and fusion proteins are soluble receptors that bind to TNF. These types of drugs are generally known as biologics and there has been an explosion in the development and testing of biologics since the 1994 US approval and launch of abciximab, a mAb that binds to GPIIb/IIIa on platelets. Anti-TNF drugs that are currently approved by FDA for treating inflammatory conditions include adalimumab, certolizumab pegol, golimumab, infliximab and etanercept. Since these agents are complex protein molecules, the pharmacodynamics and pharmacokinetics of these drugs are different from small-molecule anti-inflammatory agents. This review focuses on the pharmaceutical aspects of these drugs such as mechanism of action, adverse effects, pharmacokinetics and drug interactions. An effort was also taken to compare the pharmacodynamics and pharmacokinetic properties of these drugs, with the available data at this time.
Collapse
|
160
|
Simulations of site-specific target-mediated pharmacokinetic models for guiding the development of bispecific antibodies. J Pharmacokinet Pharmacodyn 2015; 42:1-18. [PMID: 25559227 DOI: 10.1007/s10928-014-9401-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/11/2014] [Indexed: 12/30/2022]
Abstract
Bispecific antibodies (BAbs) are novel constructs that are under development and show promise as new therapeutic modalities for cancer and autoimmune disorders. The aim of this study is to develop a semi-mechanistic modeling approach to elucidate the disposition of BAbs in plasma and possible sites of action in humans. Here we present two case studies that showcase the use of modeling to guide BAb development. In case one, a BAb is directed against a soluble and a membrane-bound ligand for treating systemic lupus erythematosus, and in case two, a BAb targets two soluble ligands as a potential treatment for ulcerative colitis and asthma. Model simulations revealed important differences between plasma and tissues, when evaluated for drug disposition and target suppression. Target concentrations at tissue sites and type (soluble vs membrane-bound), tissue-site binding, and binding affinity are all major determinants of BAb disposition and subsequently target suppression. For the presented case studies, higher doses and/or frequent dosing regimens are required to achieve 80 % target suppression in site specific tissue (the more relevant matrix) as compared to plasma. Site-specific target-mediated models may serve to guide the selection of first-in-human doses for new BAbs.
Collapse
|
161
|
Chetty M, Li L, Rose R, Machavaram K, Jamei M, Rostami-Hodjegan A, Gardner I. Prediction of the Pharmacokinetics, Pharmacodynamics, and Efficacy of a Monoclonal Antibody, Using a Physiologically Based Pharmacokinetic FcRn Model. Front Immunol 2015; 5:670. [PMID: 25601866 PMCID: PMC4283607 DOI: 10.3389/fimmu.2014.00670] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/14/2014] [Indexed: 12/27/2022] Open
Abstract
Although advantages of physiologically based pharmacokinetic models (PBPK) are now well established, PBPK models that are linked to pharmacodynamic (PD) models to predict pharmacokinetics (PK), PD, and efficacy of monoclonal antibodies (mAbs) in humans are uncommon. The aim of this study was to develop a PD model that could be linked to a physiologically based mechanistic FcRn model to predict PK, PD, and efficacy of efalizumab. The mechanistic FcRn model for mAbs with target-mediated drug disposition within the Simcyp population-based simulator was used to simulate the pharmacokinetic profiles for three different single doses and two multiple doses of efalizumab administered to virtual Caucasian healthy volunteers. The elimination of efalizumab was modeled with both a target-mediated component (specific) and catabolism in the endosome (non-specific). This model accounted for the binding between neonatal Fc receptor (FcRn) and efalizumab (protective against elimination) and for changes in CD11a target concentration. An integrated response model was then developed to predict the changes in mean Psoriasis Area and Severity Index (PASI) scores that were measured in a clinical study as an efficacy marker for efalizumab treatment. PASI scores were approximated as continuous and following a first-order asymptotic progression model. The reported steady state asymptote (Y ss) and baseline score [Y (0)] was applied and parameter estimation was used to determine the half-life of progression (T p) of psoriasis. Results suggested that simulations using this model were able to recover the changes in PASI scores (indicating efficacy) observed during clinical studies. Simulations of both single dose and multiple doses of efalizumab concentration-time profiles as well as suppression of CD11a concentrations recovered clinical data reasonably well. It can be concluded that the developed PBPK FcRn model linked to a PD model adequately predicted PK, PD, and efficacy of efalizumab.
Collapse
Affiliation(s)
| | - Linzhong Li
- Simcyp Limited (a Certara Company) , Sheffield , UK
| | - Rachel Rose
- Simcyp Limited (a Certara Company) , Sheffield , UK
| | | | - Masoud Jamei
- Simcyp Limited (a Certara Company) , Sheffield , UK
| | - Amin Rostami-Hodjegan
- Simcyp Limited (a Certara Company) , Sheffield , UK ; Manchester Pharmacy School, Manchester University , Manchester , UK
| | - Iain Gardner
- Simcyp Limited (a Certara Company) , Sheffield , UK
| |
Collapse
|
162
|
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]
|
163
|
Kuo F, Histed S, Xu B, Bhadrasetty V, Szajek LP, Williams MR, Wong K, Wu H, Lane K, Coble V, Vasalatiy O, Griffiths G, Paik CH, Elbuluk O, Szot C, Chaudhary A, St. Croix B, Choyke P, Jagoda EM. Immuno-PET imaging of tumor endothelial marker 8 (TEM8). Mol Pharm 2014; 11:3996-4006. [PMID: 24984190 PMCID: PMC4224515 DOI: 10.1021/mp500056d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 05/30/2014] [Accepted: 07/01/2014] [Indexed: 01/26/2023]
Abstract
Tumor endothelial marker 8 (TEM8) is a cell surface receptor that is highly expressed in a variety of human tumors and promotes tumor angiogenesis and cell growth. Antibodies targeting TEM8 block tumor angiogenesis in a manner distinct from the VEGF receptor pathway. Development of a TEM8 imaging agent could aid in patient selection for specific antiangiogenic therapies and for response monitoring. In these studies, L2, a therapeutic anti-TEM8 monoclonal IgG antibody (L2mAb), was labeled with (89)Zr and evaluated in vitro and in vivo in TEM8 expressing cells and mouse xenografts (NCI-H460, DLD-1) as a potential TEM8 immuno-PET imaging agent. (89)Zr-df-L2mAb was synthesized using a desferioxamine-L2mAb conjugate (df-L2mAb); (125)I-L2mAb was labeled directly. In vitro binding studies were performed using human derived cell lines with high, moderate, and low/undetectable TEM8 expression. (89)Zr-df-L2mAb in vitro autoradiography studies and CD31 IHC staining were performed with cryosections from human tumor xenografts (NCI-H460, DLD-1, MKN-45, U87-MG, T-47D, and A-431). Confirmatory TEM8 Western blots were performed with the same tumor types and cells. (89)Zr-df-L2mAb biodistribution and PET imaging studies were performed in NCI-H460 and DLD-1 xenografts in nude mice. (125)I-L2mAb and (89)Zr-df-L2mAb exhibited specific and high affinity binding to TEM8 that was consistent with TEM8 expression levels. In NCI-H460 and DLD-1 mouse xenografts nontarget tissue uptake of (89)Zr-df-L2mAb was similar; the liver and spleen exhibited the highest uptake at all time points. (89)Zr-L2mAb was highly retained in NCI-H460 tumors with <10% losses from day 1 to day 3 with the highest tumor to muscle ratios (T:M) occurring at day 3. DLD-1 tumors exhibited similar pharmacokinetics, but tumor uptake and T:M ratios were reduced ∼2-fold in comparison to NCI-H460 at all time points. NCI-H460 and DLD-1 tumors were easily visualized in PET imaging studies despite low in vitro TEM8 expression in DLD-1 cells indicating that in vivo expression might be higher in DLD-1 tumors. From in vitro autoradiography studies (89)Zr-df-L2mAb specific binding was found in 6 tumor types (U87-MG, NCI-H460, T-47D MKN-45, A-431, and DLD-1) which highly correlated to vessel density (CD31 IHC). Westerns blots confirmed the presence of TEM8 in the 6 tumor types but found undetectable TEM8 levels in DLD-1 and MKN-45 cells. This data would indicate that TEM8 is associated with the tumor vasculature rather than the tumor tissue, thus explaining the increased TEM8 expression in DLD-1 tumors compared to DLD-1 cell cultures. (89)Zr-df-L2mAb specifically targeted TEM8 in vitro and in vivo although the in vitro expression was not necessarily predictive of in vivo expression which seemed to be associated with the tumor vasculature. In mouse models, (89)Zr-df-L2mAb tumor uptakes and T:M ratios were sufficient for visualization during PET imaging. These results would suggest that a TEM8 targeted PET imaging agent, such as (89)Zr-df-L2mAb, may have potential clinical, diagnostic, and prognostic applications by providing a quantitative measure of tumor angiogenesis and patient selection for future TEM8 directed therapies.
Collapse
Affiliation(s)
- Frank Kuo
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Stephanie Histed
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Biying Xu
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Veerendra Bhadrasetty
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Lawrence P. Szajek
- PET Department and Nuclear Medicine Division,
Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mark R. Williams
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Karen Wong
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Haitao Wu
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Kelly Lane
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Vincent Coble
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Olga Vasalatiy
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Gary
L. Griffiths
- Clinical Research Directorate/CMRP, Leidos
Biomedical Research Inc. (formerly SAIC-Frederick, Inc.), Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Chang H. Paik
- PET Department and Nuclear Medicine Division,
Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Osama Elbuluk
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Christopher Szot
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Amit Chaudhary
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Brad St. Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Elaine M. Jagoda
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
164
|
Kagan L. Pharmacokinetic modeling of the subcutaneous absorption of therapeutic proteins. Drug Metab Dispos 2014; 42:1890-905. [PMID: 25122564 DOI: 10.1124/dmd.114.059121] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Subcutaneous injection is an important route of administration for therapeutic proteins that provides several advantages over other modes of parenteral delivery. Despite extensive clinical use, the exact mechanism underlying subcutaneous absorption of proteins is not completely understood, and the accuracy of prediction of absorption of biotherapeutics in humans remains unsatisfactory. This review summarizes a variety of models that have been developed for describing the pharmacokinetics of therapeutic proteins administered by subcutaneous injection, including single- and dual-pathway absorption models. Modeling of the lymphatic uptake and redistribution, absorption of monoclonal antibodies and insulin, and population analysis of protein absorption are discussed. The review also addresses interspecies modeling and prediction of absorption in humans, highlights important factors affecting the absorption processes, and suggests approaches for future development of mechanism-based absorption models.
Collapse
Affiliation(s)
- Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
165
|
Salimi-Moosavi H, Winters A, Abbott C, Patel J, Hager T, Patel V, Shih J, Zhuang Y, Ma M. A multifactorial screening strategy to identify anti-idiotypic reagents for bioanalytical support of antibody therapeutics. Anal Biochem 2014; 470:52-60. [PMID: 25447458 DOI: 10.1016/j.ab.2014.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/24/2014] [Accepted: 10/13/2014] [Indexed: 01/28/2023]
Abstract
Antibodies are critical tools for protein bioanalysis; their quality and performance dictate the caliber and robustness of ligand binding assays. After immunization, polyclonal B cells generate a diverse antibody repertoire against constant and variable regions of the therapeutic antibody immunogen. Herein we describe a comprehensive and multifactorial screening strategy to eliminate undesirable constant region-specific antibodies and select for anti-idiotypic antibodies with specificity for the unique variable region. Application of this strategy is described for the therapeutic antibody Mab-A case study. Five different factors were evaluated to select a final antibody pair for the quantification of therapeutics in biological matrices: (i) matrix effect in preclinical and clinical matrices, (ii) assay sensitivity with lower limit of quantification goal of single-digit ng/ml (low pM) at a signal-to-background ratio greater than 5, (iii) epitope distinction or nonbridging antibody pair, (iv) competition with target and inhibitory capacity enabling measurement of free drug, and (v) neutralizing bioactivity using bioassay. The selected antibody pair demonstrated superior assay sensitivity with no or minimal matrix effect in common biological samples, recognized two distinct binding epitopes on the therapeutic antibody variable region, and featured inhibitory and neutralizing effects with respect to quantification of free drug levels.
Collapse
Affiliation(s)
- Hossein Salimi-Moosavi
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA.
| | - Aaron Winters
- Department of Discovery Science, Amgen, Thousand Oaks, CA 91320, USA
| | - Christina Abbott
- Department of Protein Technologies, Amgen, Thousand Oaks, CA 91320, USA
| | - Jennifer Patel
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA 91320, USA
| | - Todd Hager
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Vimal Patel
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Judy Shih
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| | - Yao Zhuang
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA 91320, USA
| | - Mark Ma
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA 91320, USA
| |
Collapse
|
166
|
Dunbar J, Knapp B, Fuchs A, Shi J, Deane CM. Examining variable domain orientations in antigen receptors gives insight into TCR-like antibody design. PLoS Comput Biol 2014; 10:e1003852. [PMID: 25233457 PMCID: PMC4168974 DOI: 10.1371/journal.pcbi.1003852] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
The variable domains of antibodies and T-Cell receptors (TCRs) share similar structures. Both molecules act as sensors for the immune system but recognise their respective antigens in different ways. Antibodies bind to a diverse set of antigenic shapes whilst TCRs only recognise linear peptides presented by a major histocompatibility complex (MHC). The antigen specificity and affinity of both receptors is determined primarily by the sequence and structure of their complementarity determining regions (CDRs). In antibodies the binding site is also known to be affected by the relative orientation of the variable domains, VH and VL. Here, the corresponding property for TCRs, the Vβ-Vα orientation, is investigated and compared with that of antibodies. We find that TCR and antibody orientations are distinct. General antibody orientations are found to be incompatible with binding to the MHC in a canonical TCR-like mode. Finally, factors that cause the orientation of TCRs and antibodies to be different are investigated. Packing of the long Vα CDR3 in the domain-domain interface is found to be influential. In antibodies, a similar packing affect can be achieved using a bulky residue at IMGT position 50 on the VH domain. Along with IMGT VH 50, other positions are identified that may help to promote a TCR-like orientation in antibodies. These positions should provide useful considerations in the engineering of therapeutic TCR-like antibodies. The immune system needs to be able to sense molecules that might be harmful to the organism. Such harmful molecules are known as antigens. Two classes of receptor proteins that mediate antigen recognition are antibodies and T-Cell receptors (TCRs). Antibodies are able to bind a diverse range of antigen shapes whilst TCRs are specialised to recognise a cell-surface protein, the pMHC. Antibodies that bind the pMHC are rarely created naturally. However, such TCR-like antibodies are of therapeutic importance. The binding regions of the TCR and the antibody have very similar three dimensional structures. Both consist of two independent units, domains, which associate and form the antigen binding site between them. This work examines how the two domains orientate with respect to one another in TCRs and antibodies. Our results show that the conformations that exist in TCRs and antibodies are distinct. Consequently it is difficult for an antibody to bind to a pMHC in the same way a TCR would. However, a similar conformation can be achieved in antibodies as in TCRs by the presence of certain amino-acids in the domain interface. This knowledge should aid the development of therapeutic TCR-like antibodies.
Collapse
Affiliation(s)
- James Dunbar
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Bernhard Knapp
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Angelika Fuchs
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Informatics, Penzberg, Germany
| | - Jiye Shi
- Informatics, UCB Pharma, Slough, United Kingdom
| | - Charlotte M. Deane
- Department of Statistics, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
167
|
King J, Alexander M, Byrne J, MacMillan K, Mollo A, Kirsa S, Green M. A review of the evidence for occupational exposure risks to novel anticancer agents – A focus on monoclonal antibodies. J Oncol Pharm Pract 2014; 22:121-34. [DOI: 10.1177/1078155214550729] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction Evidence of occupational exposure risks to novel anticancer agents is limited and yet to be formally evaluated from the Australian healthcare perspective. Methods From March to September 2013 medical databases, organizational policies, drug monographs, and the World Wide Web were searched for evidence relating to occupational exposure to monoclonal antibodies, fusion proteins, gene therapies, and other unclassified novel anticancer agents. Results Australian legislation, national and international guidelines, and drug company information excluded novel agents or provided inconsistent risk assessments and safe handling recommendations. Monoclonal antibody guidelines reported conflicting information and were often divergent with available evidence and pharmacologic rationale demonstrating minimal internalisation ability and occupational exposure risk. Despite similar physiochemical, pharmacologic, and internalisation properties to monoclonal antibodies, fusion proteins were included in only a minority of guidelines. Clinical directives for the safe handling of gene therapies and live vaccines were limited, where available focusing on prevention against exposure and cross-contamination. Although mechanistically different, novel small molecule agents (proteasome inhibitors), possess similar physiochemical and internalisation properties to traditional cytotoxic agents warranting cytotoxic classification and handling. Conclusion Novel agents are rapidly emerging into clinical practice, and healthcare personnel have few resources to evaluate risk and provide safety recommendations. Novel agents possess differing physical, molecular and pharmacological profiles compared to traditional cytotoxic anticancer agents. Evaluation of occupational exposure risk should consider both toxicity and internalisation. Evidence-based guidance able to direct safe handling practices for novel anticancer agents across a variety of clinical settings is urgently required.
Collapse
Affiliation(s)
- Julie King
- Pharmacy Department, Western Health, Melbourne, Australia
| | - Marliese Alexander
- Pharmacy Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jenny Byrne
- Western and Central Melbourne Integrated Cancer Service, Melbourne, Australia
| | - Kent MacMillan
- Pharmacy Department, Western Health, Melbourne, Australia
| | | | - Sue Kirsa
- Pharmacy Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael Green
- Department of Cancer Services, Western Health, Melbourne, Australia
| |
Collapse
|
168
|
Vécsei L, Szok D, Csáti A, Tajti J. CGRP antagonists and antibodies for the treatment of migraine. Expert Opin Investig Drugs 2014; 24:31-41. [DOI: 10.1517/13543784.2015.960921] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
169
|
Widmer N, Bardin C, Chatelut E, Paci A, Beijnen J, Levêque D, Veal G, Astier A. Review of therapeutic drug monitoring of anticancer drugs part two – Targeted therapies. Eur J Cancer 2014; 50:2020-36. [DOI: 10.1016/j.ejca.2014.04.015] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/11/2014] [Indexed: 02/06/2023]
|
170
|
Li L, Gardner I, Dostalek M, Jamei M. Simulation of monoclonal antibody pharmacokinetics in humans using a minimal physiologically based model. AAPS JOURNAL 2014; 16:1097-109. [PMID: 25004823 DOI: 10.1208/s12248-014-9640-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 06/18/2014] [Indexed: 12/13/2022]
Abstract
Compared to small chemical molecules, monoclonal antibodies and Fc-containing derivatives (mAbs) have unique pharmacokinetic behaviour characterised by relatively poor cellular permeability, minimal renal filtration, binding to FcRn, target-mediated drug disposition, and disposition via lymph. A minimal physiologically based pharmacokinetic (PBPK) model to describe the pharmacokinetics of mAbs in humans was developed. Within the model, the body is divided into three physiological compartments; plasma, a single tissue compartment and lymph. The tissue compartment is further sub-divided into vascular, endothelial and interstitial spaces. The model simultaneously describes the levels of endogenous IgG and exogenous mAbs in each compartment and sub-compartment and, in particular, considers the competition of these two species for FcRn binding in the endothelial space. A Monte-Carlo sampling approach is used to simulate the concentrations of endogenous IgG and mAb in a human population. Existing targeted-mediated drug disposition (TMDD) models are coupled with the minimal PBPK model to provide a general platform for simulating the pharmacokinetics of therapeutic antibodies using primarily pre-clinical data inputs. The feasibility of utilising pre-clinical data to parameterise the model and to simulate the pharmacokinetics of adalimumab and an anti-ALK1 antibody (PF-03446962) in a population of individuals was investigated and results were compared to published clinical data.
Collapse
Affiliation(s)
- Linzhong Li
- Simcyp Limited, A Certara Company, Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK,
| | | | | | | |
Collapse
|
171
|
Bogen IL, Boix F, Nerem E, Mørland J, Andersen JM. A monoclonal antibody specific for 6-monoacetylmorphine reduces acute heroin effects in mice. J Pharmacol Exp Ther 2014; 349:568-76. [PMID: 24700886 DOI: 10.1124/jpet.113.212035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy against drugs of abuse is being studied as an alternative treatment option in addiction medicine and is based on antibodies sequestering the drug in the bloodstream and blocking its entry into the brain. Producing an efficient vaccine against heroin has been considered particularly challenging because of the rapid metabolism of heroin to multiple psychoactive molecules. We have previously reported that heroin's first metabolite, 6-monoacetylmorphine (6-MAM), is the predominant mediator for heroin's acute behavioral effects and that heroin is metabolized to 6-MAM primarily prior to brain entry. On this basis, we hypothesized that antibody sequestration of 6-MAM is sufficient to impair heroin-induced effects and therefore examined the effects of a monoclonal antibody (mAb) specific for 6-MAM. In vitro experiments in human and rat blood revealed that the antibody was able to bind 6-MAM and block the metabolism to morphine almost completely, whereas the conversion of heroin to 6-MAM remained unaffected. Mice pretreated with the mAb toward 6-MAM displayed a reduction in heroin-induced locomotor activity that corresponded closely to the reduction in brain 6-MAM levels. Intraperitoneal and intravenous administration of the anti-6-MAM mAb gave equivalent protection against heroin effects, and the mAb was estimated to have a functional half-life of 8 to 9 days in mice. Our study implies that an antibody against 6-MAM is effective in counteracting heroin effects.
Collapse
Affiliation(s)
- Inger Lise Bogen
- Department of Drug Abuse Research and Method Development, Division of Forensic Sciences, Norwegian Institute of Public Health, Oslo, Norway
| | | | | | | | | |
Collapse
|
172
|
Dollery CT. Lost in Translation (LiT): IUPHAR Review 6. Br J Pharmacol 2014; 171:2269-90. [PMID: 24428732 PMCID: PMC3997269 DOI: 10.1111/bph.12580] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/20/2013] [Accepted: 12/18/2013] [Indexed: 12/14/2022] Open
Abstract
Translational medicine is a roller coaster with occasional brilliant successes and a large majority of failures. Lost in Translation 1 ('LiT1'), beginning in the 1950s, was a golden era built upon earlier advances in experimental physiology, biochemistry and pharmacology, with a dash of serendipity, that led to the discovery of many new drugs for serious illnesses. LiT2 saw the large-scale industrialization of drug discovery using high-throughput screens and assays based on affinity for the target molecule. The links between drug development and university sciences and medicine weakened, but there were still some brilliant successes. In LiT3, the coverage of translational medicine expanded from molecular biology to drug budgets, with much greater emphasis on safety and official regulation. Compared with R&D expenditure, the number of breakthrough discoveries in LiT3 was disappointing, but monoclonal antibodies for immunity and inflammation brought in a new golden era and kinase inhibitors such as imatinib were breakthroughs in cancer. The pharmaceutical industry is trying to revive the LiT1 approach by using phenotypic assays and closer links with academia. LiT4 faces a data explosion generated by the genome project, GWAS, ENCODE and the 'omics' that is in danger of leaving LiT4 in a computerized cloud. Industrial laboratories are filled with masses of automated machinery while the scientists sit in a separate room viewing the results on their computers. Big Data will need Big Thinking in LiT4 but with so many unmet medical needs and so many new opportunities being revealed there are high hopes that the roller coaster will ride high again.
Collapse
|
173
|
Sailstad JM, Amaravadi L, Clements-Egan A, Gorovits B, Myler HA, Pillutla RC, Pursuhothama S, Putman M, Rose MK, Sonehara K, Tang L, Wustner JT. A white paper--consensus and recommendations of a global harmonization team on assessing the impact of immunogenicity on pharmacokinetic measurements. AAPS J 2014; 16:488-98. [PMID: 24682765 PMCID: PMC4012055 DOI: 10.1208/s12248-014-9582-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/20/2014] [Indexed: 11/30/2022] Open
Abstract
The Global Bioanalysis Consortium (GBC) set up an international team to explore the impact of immunogenicity on pharmacokinetic (PK) assessments. The intent of this paper is to define the field and propose best practices when developing PK assays for biotherapeutics. We focus on the impact of anti-drug antibodies (ADA) on the performance of PK assay leading to the impact on the reported drug concentration and exposure. The manuscript describes strategies to assess whether the observed change in the drug concentration is due to the ADA impact on drug clearance rates or is a consequence of ADA interference in the bioanalytical method applied to measure drug concentration. This paper provides the bioanalytical scientist guidance for developing ADA-tolerant PK methods. It is essential that the data generated in the PK, ADA, pharmacodynamic and efficacy/toxicity evaluations are viewed together. Therefore, the extent for the investigation of the PK sensitivity to the presence of ADA should be driven by the project needs and risk based.
Collapse
Affiliation(s)
- J M Sailstad
- Sailstad and Associates Inc., Durham, North Carolina, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Target-mediated clearance and bio-distribution of a monoclonal antibody against the Kunitz–type protease inhibitor 2 domain of Tissue Factor Pathway Inhibitor. Thromb Res 2014; 133:464-71. [DOI: 10.1016/j.thromres.2013.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/29/2013] [Accepted: 12/11/2013] [Indexed: 01/08/2023]
|
175
|
Immunogenicity and PK/PD evaluation in biotherapeutic drug development: scientific considerations for bioanalytical methods and data analysis. Bioanalysis 2014; 6:79-87. [DOI: 10.4155/bio.13.302] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
With the advent of novel technologies, considerable advances have been made in the evaluation of the relationship between PK and PD. Ligand-binding assays have been the primary assay format supporting PK and immunogenicity assessments. Critical and in-depth characterizations of the ligand-binding assay of interest can provide valuable understanding of the limitations, for interpreting PK/PD and immunogenicity results. This review illustrates key challenges with regard to understanding the relationship between anti-drug antibody and PK/PD, including confounding factors associated with the development and validation of ligand-binding assays, mechanisms by which anti-drug antibody impacts PK/PD, factors to consider during data analyses and interpretation, and a perspective on integrating immunogenicity data into the well-established quantitative modeling approach. Through recognizing these challenges, we propose some opportunities for improvements in the development and validation of fit-for-purpose bioanalytical methods.
Collapse
|
176
|
Dodds M, Chow V, Markus R, Pérez-Ruixo JJ, Shen D, Gibbs M. The use of pharmacometrics to optimize biosimilar development. J Pharm Sci 2013; 102:3908-14. [PMID: 24027111 DOI: 10.1002/jps.23697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 06/07/2013] [Accepted: 07/24/2013] [Indexed: 12/27/2022]
Abstract
Pharmacometric approaches can assist in biosimilar development by leveraging quantitative knowledge of the originator product characteristics such as dose-exposure and exposure-response information to support a targeted approach to clinical studies. The degree to which these approaches can be applied relies on the level of information known about the originator and information that supports application of the originator model to the biosimilar. A model-based approach testing the hypothesis that the biosimilar PK and/or PK/PD profile is similar to the originator in the target patient population is aligned with the central comparability exercise required for the biosimilar approval. This Commentary details the key opportunities in study design and study analysis where pharmacometrics approaches can aid biosimilar development.
Collapse
Affiliation(s)
- Mike Dodds
- Pharmacokinetics & Drug Metabolism, Amgen Inc., Seattle, Washington, 98119
| | | | | | | | | | | |
Collapse
|
177
|
Gupta R, Levin E, Wu JJ, Koo J, Liao W. An update on drug–drug interactions with biologics for the treatment of moderate-to-severe psoriasis. J DERMATOL TREAT 2013; 25:87-9. [DOI: 10.3109/09546634.2013.825041] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
178
|
Golay J, Semenzato G, Rambaldi A, Foà R, Gaidano G, Gamba E, Pane F, Pinto A, Specchia G, Zaja F, Regazzi M. Lessons for the clinic from rituximab pharmacokinetics and pharmacodynamics. MAbs 2013; 5:826-37. [PMID: 23933992 DOI: 10.4161/mabs.26008] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The anti-CD20 antibody rituximab (RTX; Rituxan®, MabThera®) was the first anti-cancer antibody approved by the US Food and Drug Administration in 1997 and it is now the most-studied unconjugated therapeutic antibody. The knowledge gained over the past 15 y on the pharmacodynamics (PD) of this antibody has led to the development of a new generation of anti-CD20 antibodies with enhanced efficacy in vitro. Studies on the pharmacokinetics (PK) properties and the effect of factors such as tumor load and localization, antibody concentration in the circulation and gender on both PK and clinical response has allowed the design of optimized schedules and novel routes of RTX administration. Although clinical results using newer anti-CD20 antibodies, such as ofatumumab and obinutuzumab, and novel administration schedules for RTX are still being evaluated, the knowledge gained so far on RTX PK and PD should also be relevant for other unconjugated monoclonal antibody therapeutics, and will be critically reviewed here.
Collapse
Affiliation(s)
- Josée Golay
- Division of Hematology; Ospedale Papa Giovanni XXIII; Bergamo, Italy
| | - Gianpietro Semenzato
- Padua University School of Medicine; Hematology Branch; Department of Medicine; Padua, Italy
| | | | - Robin Foà
- Division of Hematology; Department of Cellular Biotechnologies and Hematology; University "Sapienza"; Rome, Italy
| | - Gianluca Gaidano
- Division of Hematology; Department of Translational Medicine; Amedeo Avogadro University of Eastern Piedmont; Novara, Italy
| | | | - Fabrizio Pane
- Dipartimento di Medicina Clinica e Chirurgia; Università di Napoli Federico II and Ceinge-Biotecnologie Avanzate; Naples, Italy
| | - Antonello Pinto
- Hematology-Oncology and Stem Cell Transplantation Unit; Istituto Nazionale Tumori; Fondazione 'G.Pascale'; IRCCS; Naples, Italy
| | | | - Francesco Zaja
- Clinica Ematologica; DISM, AOUD S.M. Misericordia; Udine, Italy
| | | |
Collapse
|