1
|
Wang J, Chen T, Ruszaj DM, Mager DE, Straubinger RM. Integrated PK/PD Modeling Relates Smoothened Inhibitor Biomarkers to The Heterogeneous Intratumor Disposition of Cetuximab in Pancreatic Cancer Tumor Models. J Pharm Sci 2024; 113:72-84. [PMID: 37844759 DOI: 10.1016/j.xphs.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
Therapeutic antibodies have shown little efficacy in the treatment of pancreatic ductal adenocarcinomas (PDAC). Tumor desmoplasia, hypovascularity, and poor perfusion result in insufficient tumor cell exposure, contributing to treatment failure. Smoothened inhibitors of hedgehog signaling (sHHi) increase PDAC tumor permeability, perfusion, and drug delivery, and provide a tool to develop a quantitative, mechanistic understanding as to how the temporal dynamics of tumor priming can impact intratumor distribution of monoclonal antibodies (mAb). A linked pharmacokinetic (PK)/pharmacodynamic (PD) model was developed to integrate the plasma and tumor PK of a sHHi priming agent with its effects upon downstream stromal biomarkers Gli1, hyaluronic acid, and interstitial fluid pressure in PDAC patient-derived xenograft (PDX) tumors. In parallel, in situ tumor concentrations of cetuximab (CTX: anti-epidermal growth factor receptor; EGFR) were quantified as a marker for tumor delivery of mAb or antibody-drug conjugates. A minimal, physiologically-based pharmacokinetic (mPBPK) model was constructed to link sHHi effects upon mechanistic effectors of tumor barrier compromise with the intratumor distribution of CTX, and CTX occupancy of EGFR in tumors. Integration of the mPBPK model of mAb deposition and intratumor distribution with the PK/PD model of tumor responses to priming not only identified physiological parameters that are critical for tumor antibody distribution, but also provides insight into dosing regimens that could achieve maximal tumor disposition of therapeutic antibodies under conditions of transient PDAC tumor permeability barrier compromise that mechanistically-diverse tumor priming strategies may achieve.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Ting Chen
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Donna M Ruszaj
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Enhanced Pharmacodynamics, LLC, Buffalo, NY, USA
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Department of Cell Stress Biochemistry and Biophysics, Roswell Park Comprehenhsive Cancer Center, Buffalo, NY, USA; Department of Pharmacology and Therapeutics, Roswell Park Comprehenhsive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
2
|
Naoi S, Yamane M, Nemoto T, Kato M, Saito R, Tachibana T. Physiologically based pharmacokinetic (PBPK) model that describes enhanced FcRn-dependent distribution of monoclonal antibodies (mAbs) by pI-engineering in mice. Drug Metab Pharmacokinet 2023; 53:100506. [PMID: 38029470 DOI: 10.1016/j.dmpk.2023.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/22/2023] [Accepted: 03/15/2023] [Indexed: 12/01/2023]
Abstract
We previously reported that monoclonal antibodies (mAbs) with a high isoelectric point (pI) value tended to exhibit fast plasma clearance (CL) and large steady-state volume of distribution (Vdss) in mice. However, the positive correlation between pI, CL, and Vdss cannot be described by the reported physiologically based pharmacokinetic (PBPK) models, in which FcRn-mediated transcytosis of mAbs is set to be minimal compared to convection-mediated transport. To address this issue, physiological parameters (lymph flow rate, reflection coefficient, endothelial uptake clearance, and FcRn concentration) were optimized based on the pharmacokinetic profiles of mAbs with various pI values in wild type and FcRn-deficient (beta-2-microglobulin knockout [KO]) mice. Simulations using the PBPK model developed in this study showed a positive correlation between pI, CL and Vdss observed in wild-type mice. Therefore, this model successfully characterized our hypothetical mechanism that an electrostatic positive interaction between mAbs and the endothelial membrane enhances FcRn-mediated transcytosis of mAbs, resulting in large Vdss. We sought to determine the right contribution of the two pathways of antibody distribution to the interstitial space and established a new model that could effectively capture the effect of pI on FcRn-mediated distribution of mAbs in the body.
Collapse
Affiliation(s)
- Sotaro Naoi
- Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka, 412-8513, Japan.
| | - Mizuki Yamane
- Chugai Pharmaceutical Co., Ltd., 200, Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Takayuki Nemoto
- Chugai Pharmaceutical Co., Ltd., 200, Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Motohiro Kato
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20, Shinmachi, Nishitokyo, Tokyo, 202-8585, Japan
| | - Ryoichi Saito
- Chugai Pharmaceutical Co., Ltd., 200, Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Tatsuhiko Tachibana
- Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka, 412-8513, Japan
| |
Collapse
|
3
|
Han SY, Yu JE, You BH, Kim SY, Bae M, Chae HS, Chin YW, Hong SH, Lee JH, Jung SH, Choi YH. No Interference of H9 Extract on Trastuzumab Pharmacokinetics in Their Combinations. Int J Mol Sci 2023; 24:16677. [PMID: 38068999 PMCID: PMC10706748 DOI: 10.3390/ijms242316677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Trastuzumab is used to treat breast cancer patients overexpressing human epidermal growth factor receptor 2, but resistance and toxicity limit its uses, leading to attention to trastuzumab combinations. Recently, the synergistic effect of trastuzumab and H9 extract (H9) combination against breast cancer has been reported. Because drug exposure determines its efficacy and toxicity, the question of whether H9 changes trastuzumab exposure in the body has been raised. Therefore, this study aimed to characterize trastuzumab pharmacokinetics and elucidate the effect of H9 on trastuzumab pharmacokinetics at a combination dose that shows synergism in mice. As a result, trastuzumab showed linear pharmacokinetics after its intravenous administration from 1 to 10 mg/kg. In the combination of trastuzumab and H9, single and 2-week treatments of oral H9 (500 mg/kg) did not influence trastuzumab pharmacokinetics. In the multiple-combination treatments of trastuzumab and H9 showing their synergistic effect (3 weeks of trastuzumab with 2 weeks of H9), the pharmacokinetic profile of trastuzumab was comparable to that of 3 weeks of trastuzumab alone. In tissue distribution, the tissue to plasma ratios of trastuzumab below 1.0 indicated its limited distributions within the tissues, and these patterns were unaffected by H9. These results suggest that the systemic and local exposures of trastuzumab are unchanged by single and multiple-combination treatments of H9.
Collapse
Affiliation(s)
- Seung Yon Han
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| | - Jeong-Eun Yu
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| | - Byoung Hoon You
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| | - Seo-Yeon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| | - Mingoo Bae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| | - Hee-Sung Chae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea;
| | - Soo-Hwa Hong
- Department of Korean Internal Medicine, Dongguk University Bundang Korean Medicine Hospital, Seongnam-si 13601, Gyeonggi-do, Republic of Korea;
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Gyeongju-si 38066, Gyeongsangbuk-do, Republic of Korea; (J.-H.L.); (S.H.J.)
| | - Seung Hyun Jung
- College of Korean Medicine, Dongguk University, Gyeongju-si 38066, Gyeongsangbuk-do, Republic of Korea; (J.-H.L.); (S.H.J.)
| | - Young Hee Choi
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (S.Y.H.); (J.-E.Y.); (B.H.Y.); (S.-Y.K.); (M.B.); (H.-S.C.)
| |
Collapse
|
4
|
A triple enhanced permeable gold nanoraspberry designed for positive feedback interventional therapy. J Control Release 2022; 345:120-137. [PMID: 35276301 DOI: 10.1016/j.jconrel.2022.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/24/2022]
Abstract
Due to the unique microenvironment, nanoparticles cannot easily penetrate deeply into tumours, which decreases their therapeutic efficacy. Thus, new strategies should be developed to solve this problem and increase the efficacy of nanomedicine. In this study, gold nanoraspberries (GNRs) were constructed using ultrasmall gold nanospheres (UGNPs) with a matrix metalloproteinase (MMP)-2/9-sensitive peptide as a cross-linking agent. These UGNPs were then modified with trastuzumab (TRA) and mertansine derivatives (DM1) via the AuS bond. TRA targets the human epidermal growth factor receptor-2 (Her-2) which is overexpressed on Her-2+ breast cancer cells. The AuS bond in GNRs-DM1 can be replaced by the free sulfhydryl group of GSH, which could achieve GSH dependent redox responsive release of the drug. In the mouse model of Her-2+ breast cancer, a "positive feedback" triple enhanced penetration platform was construct to treat tumours. Firstly, near-infrared light-triggered photothermal conversion increased vascular permeability, resulting in nanoparticle penetration. Secondly, GNRs disintegrated into UGNPs in response to stimulation with MMPs. GNRs with larger particle sizes reached the tumour site through EPR effect and active targeting. Meanwhile, UGNPs with smaller particle sizes penetrated deeply into the tumour through diffusion. Thirdly, the UGNPs transformed activated cancer-associated fibroblasts to a quiescent state, which reduced intercellular pressure and promoted the penetration of the UGNPs into the interior of the tumour. In turn, an increase in the number of nanoparticles penetrating into the tumour led to a "positive feedback" loop of triple enhanced photothermal effects and further self-amplify the permeability in vivo. Interventional photothermal therapy (IPTT) was used to improve the therapeutic efficacy by reducing the laser power attenuation caused by percutaneous irradiation. The GNRs also showed excellent multimode imaging (computed tomography, photoacoustic imaging and photothermal imaging) capabilities and high anti-tumour efficacy due to efficient tumour targeting and triple enhanced deep penetration into the tumour site. Thus, these MMP-2/redox dual-responsive GNRs are promising carriers of drugs targeting human epidermal growth factor receptor 2+ breast cancer.
Collapse
|
5
|
Hardiansyah D, Ng CM. Minimal Physiologically-based Pharmacokinetic Model to Investigate the Effect of Charge on the Pharmacokinetics of Humanized anti-HCV-E2 IgG Antibodies in Sprague-Dawley Rats. Pharm Res 2022; 39:481-496. [PMID: 35246757 DOI: 10.1007/s11095-022-03204-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 02/15/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To develop a minimal physiologically-based pharmacokinetic (mPBPK) model in quantifying the relationships between the charge and pharmacokinetics (PK) of therapeutic monoclonal IgG antibody (TMAb). METHODS PK data used in this study were native IgG and five humanized anti-HCVE2-IgG antibodies in rats. Different models that related the effect of charge on interstitial distribution, transcapillary transport, and cellular uptake for FcRn-mediated metabolism were tested. External validation was conducted to assess if the charge-parameter relationships derived from rats could be used to predict the PK of TMAbs in mice. The final mPBPK model was used to construct the relationships between the FcRn binding and charge on the PK of TMAbs. RESULTS Increasing the isoelectric point (pI) of IgG was associated with higher interstitial space distribution and cellular uptake. The transcapillary transport of IgG from plasma to interstitial space remains constant with pI values below 7.96 and then increased linearly with pI. The model-based simulation results suggested that improving the FcRn binding affinity can overcome the problems of low plasma/interstitial space exposures associated with TMAbs with higher pI values by reducing the FcRn-mediated metabolism and hence increasing drug exposure in the interstitial space that has close contact with many solid tumors. CONCLUSIONS The final mPBPK model was developed and used to construct complex quantitative relationships between the pI/FcRn binding affinity and PK of TMAbs and such relationships are useful to select the discovery of a "sweet spot" of designing future generation of TMAbs with optimal PK properties to achieve desirable plasma and tissue drug exposures.
Collapse
Affiliation(s)
- Deni Hardiansyah
- Physics Department, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16424, Indonesia
| | - Chee Meng Ng
- NewGround Pharmaceutical Consulting LLC, Foster City, CA, 94404, USA.
| |
Collapse
|
6
|
Stüber JC, Rechberger KF, Miladinović SM, Pöschinger T, Zimmermann T, Villenave R, Eigenmann MJ, Kraft TE, Shah DK, Kettenberger H, Richter WF. Impact of charge patches on tumor disposition and biodistribution of therapeutic antibodies. AAPS OPEN 2022. [DOI: 10.1186/s41120-021-00048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThis study explores the impact of antibody surface charge on tissue distribution into various tissues including tumor. Tumor-bearing mice were dosed intravenously with a mixture comprising three antibodies engineered to carry negative charge patches, a balanced charge distribution, or positive patches, respectively (cassette dosing). Tissue levels were analyzed with a specific LC-MS/MS method. In addition, the antibody mix was administered to non-tumor bearing mice. Muscle and skin interstitial fluid were obtained by centrifugation and analyzed by LC-MS/MS. An in vitro endothelium model was explored for its feasibility to mimic the observed distribution differences.A balanced charge distribution was optimal in terms of total tumor exposure, while in other tissues, negatively charged and balanced charged antibodies gave similar results. In contrast, positive charge patches generally resulted in increased serum clearance but markedly enhanced tumor and organ uptake, leading to higher tissue-to-serum ratios. The uptake and availability in the interstitial space were confirmed by specific assessment of antibody levels in the interstitial fluid of the muscle and skin, with similar charge impact as in total tissue. The in vitro model was able to differentiate the transport propensity of this series of antibody variants. In summary, our results show the differential effects of charge patches on an antibody surface on biodistribution and tumor uptake. These insights may help in the design of molecules with biodistribution properties tailored to their purpose, and an optimized safety profile.
Collapse
|
7
|
Pharmacokinetics and Pharmacodynamics of T-Cell Bispecifics in the Tumour Interstitial Fluid. Pharmaceutics 2021; 13:pharmaceutics13122105. [PMID: 34959386 PMCID: PMC8705663 DOI: 10.3390/pharmaceutics13122105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
The goal of this study is to investigate the pharmacokinetics in plasma and tumour interstitial fluid of two T-cell bispecifics (TCBs) with different binding affinities to the tumour target and to assess the subsequent cytokine release in a tumour-bearing humanised mouse model. Pharmacokinetics (PK) as well as cytokine data were collected in humanised mice after iv injection of cibisatamab and CEACAM5-TCB which are binding with different binding affinities to the tumour antigen carcinoembryonic antigen (CEA). The PK data were modelled and coupled to a previously published physiologically based PK model. Corresponding cytokine release profiles were compared to in vitro data. The PK model provided a good fit to the data and precise estimation of key PK parameters. High tumour interstitial concentrations were observed for both TCBs, influenced by their respective target binding affinities. In conclusion, we developed a tailored experimental method to measure PK and cytokine release in plasma and at the site of drug action, namely in the tumour. Integrating those data into a mathematical model enabled to investigate the impact of target affinity on tumour accumulation and can have implications for the PKPD assessment of the therapeutic antibodies.
Collapse
|
8
|
Li H, Lyu Y, Chen X, Li B, Hua Q, Ji F, Yin Y, Li H. Layers of interstitial fluid flow along a "slit-shaped" vascular adventitia. J Zhejiang Univ Sci B 2021; 22:647-663. [PMID: 34414700 DOI: 10.1631/jzus.b2000590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Interstitial fluid (ISF) flow through vascular adventitia has been discovered recently. However, its kinetic pattern was unclear. We used histological and topographical identification to observe ISF flow along venous vessels in rabbits. By magnetic resonance imaging (MRI) in live subjects, the inherent pathways of ISF flow from the ankle dermis through the legs, abdomen, and thorax were enhanced by paramagnetic contrast. By fluorescence stereomicroscopy and layer-by-layer dissection after the rabbits were sacrificed, the perivascular and adventitial connective tissues (PACTs) along the saphenous veins and inferior vena cava were found to be stained by sodium fluorescein from the ankle dermis, which coincided with the findings by MRI. The direction of ISF transport in a venous PACT pathway was the same as that of venous blood flow. By confocal microscopy and histological analysis, the stained PACT pathways were verified to be the fibrous connective tissues, consisting of longitudinally assembled fibers. Real-time observations by fluorescence stereomicroscopy revealed at least two types of spaces for ISF flow: one along adventitial fibers and another one between the vascular adventitia and its covering fascia. Using nanoparticles and surfactants, a PACT pathway was found to be accessible by a nanoparticle of <100 nm and contained two parts: a transport channel and an absorptive part. The calculated velocity of continuous ISF flow along fibers of the PACT pathway was 3.6‒15.6 mm/s. These data revealed that a PACT pathway was a "slit-shaped" porous biomaterial, comprising a longitudinal transport channel and an absorptive part for imbibition. The use of surfactants suggested that interfacial tension might play an essential role in layers of continuous ISF flow along vascular vessels. A hypothetical "gel pump" is proposed based on interfacial tension and interactions to regulate ISF flow. These experimental findings may inspire future studies to explore the physiological and pathophysiological functions of vascular ISF or interfacial fluid flow among interstitial connective tissues throughout the body.
Collapse
Affiliation(s)
- Hongyi Li
- Cardiology Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China. .,Cardiology Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - You Lyu
- Cardiology Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaoliang Chen
- Radiology Department, China-Japan Friendship Hospital, Beijing 100029, China
| | - Bei Li
- Cardiology Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qi Hua
- Cardiology Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China. ,
| | - Fusui Ji
- Cardiology Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yajun Yin
- Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Hua Li
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
9
|
Torres C, Dumas S, Palacio-Castañeda V, Descroix S, Brock R, Verdurmen WPR. A Computational Investigation of In Vivo Cytosolic Protein Delivery for Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13040562. [PMID: 33921165 PMCID: PMC8071550 DOI: 10.3390/pharmaceutics13040562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to specifically block or degrade cytosolic targets using therapeutic proteins would bring tremendous therapeutic opportunities in cancer therapy. Over the last few years, significant progress has been made with respect to tissue targeting, cytosolic delivery, and catalytic inactivation of targets, placing this aim within reach. Here, we developed a mathematical model specifically built for the evaluation of approaches towards cytosolic protein delivery, involving all steps from systemic administration to translocation into the cytosol and target engagement. Focusing on solid cancer tissues, we utilized the model to investigate the effects of microvascular permeability, receptor affinity, the cellular density of targeted receptors, as well as the mode of activity (blocking/degradation) on therapeutic potential. Our analyses provide guidance for the rational optimization of protein design for enhanced activity and highlight the importance of tuning the receptor affinity as a function of receptor density as well as the receptor internalization rate. Furthermore, we provide quantitative insights into how enzymatic cargoes can enhance the distribution, extent, and duration of therapeutic activity, already at very low catalytic rates. Our results illustrate that with current protein engineering approaches, the goal of delivery of cytosolic delivery of proteins for therapeutic effects is well within reach.
Collapse
Affiliation(s)
- Camilo Torres
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (C.T.); (V.P.-C.); (R.B.)
| | - Simon Dumas
- Physico-Chemistry Curie, Institut Curie, PSL Research University, CNRS UMR168, Sorbonne University, 75005 Paris, France; (S.D.); (S.D.)
| | - Valentina Palacio-Castañeda
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (C.T.); (V.P.-C.); (R.B.)
| | - Stéphanie Descroix
- Physico-Chemistry Curie, Institut Curie, PSL Research University, CNRS UMR168, Sorbonne University, 75005 Paris, France; (S.D.); (S.D.)
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (C.T.); (V.P.-C.); (R.B.)
| | - Wouter P. R. Verdurmen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (C.T.); (V.P.-C.); (R.B.)
- Correspondence:
| |
Collapse
|
10
|
Case BA, Kruziki MA, Stern LA, Hackel BJ. Evaluation of affibody charge modification identified by synthetic consensus design in molecular PET imaging of epidermal growth factor receptor. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2018; 3:171-182. [PMID: 31467687 PMCID: PMC6715147 DOI: 10.1039/c7me00095b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tumor overexpression of epidermal growth factor receptor (EGFR) correlates to therapeutic response in select patient populations. Thus, molecular positron emission tomography (PET) imaging of EGFR could stratify responders versus non-responders. We previously demonstrated effectiveness of a "synthetic consensus" design principle to identify six neutralizing mutations within a 58-amino acid EGFR-targeted affibody domain. Herein, we extend the approach to identify additional neutralized variants that vary net charge from -2 to either -4 or +4 while retaining high affinity (1.6 ± 1.2 nM and 2.5 ± 0.7 nM), specific binding to EGFR, secondary structure, and stability (Tm = 68 °C and 59 °C). We radiolabeled the resultant collection of five charge variants with 64Cu and evaluated PET imaging performance in murine models with subcutaneously xenografted EGFRhigh and EGFRlow tumors. All variants exhibited good EGFRhigh tumor imaging as early as 1 h, with EA35S (+3/-5) achieving 7.7 ± 1.4 %ID/g tumor at 4 h with 1.5 ± 0.3%ID/g EGFRlow tumor, 34 ± 5 tumor:muscle and 12 ± 3 tumor:blood ratios. The positively charged EA62S mutant (+6/-2) exhibited 2.2-3.3-fold higher liver signal than the other variants (p<0.01). The EA68 variant with higher charge density was more stable to human and mouse serum than neutralized variants. In a comparison of radiometal chelators, 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA) exhibited superior physiological specificity to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). In total, these studies comparatively evaluated a set of EGFR-targeted affibodies varying in net charge and charge density, which revealed functional variations that are useful in engineering an ideal probe for translational studies.
Collapse
Affiliation(s)
- Brett A Case
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Max A Kruziki
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Lawrence A Stern
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
11
|
Abstract
Hydrogels have evolved into indispensable biomaterials in the fields of drug delivery and regenerative medicine. This minireview aims to highlight the recent advances in the hydrogel design for controlled release of bioactive proteins. The latest developments of enzyme-responsive and externally regulated drug delivery systems are summarized. The design strategies and applications of phase-separated hydrogel systems are also described. We expect that these emerging approaches will enable expanded use of hydrogels in biomedicine and healthcare.
Collapse
Affiliation(s)
- Ki Hyun Bae
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | | |
Collapse
|
12
|
Abstract
We review recent progress in cancer nanomedicine to overcome the delivery barriers in tumor microenvironment, including the understanding in the nanomedicine delivery process, stimulus-responsive delivery, and several new strategies to normalize tumor microenvironment. The application of nanomedicine in cancer immunotherapy, a renewed cancer therapy by recent breakthrough, is also highlighted.
Collapse
|
13
|
Hyaluronidase-incorporated hyaluronic acid–tyramine hydrogels for the sustained release of trastuzumab. J Control Release 2015; 216:47-55. [DOI: 10.1016/j.jconrel.2015.08.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/24/2015] [Accepted: 08/06/2015] [Indexed: 02/07/2023]
|
14
|
Golkar N, Tamaddon AM, Samani SM. Effect of lipid composition on incorporation of trastuzumab-PEG-lipid into nanoliposomes by post-insertion method: physicochemical and cellular characterization. J Liposome Res 2015; 26:113-25. [DOI: 10.3109/08982104.2015.1048692] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Vasalou C, Helmlinger G, Gomes B. A mechanistic tumor penetration model to guide antibody drug conjugate design. PLoS One 2015; 10:e0118977. [PMID: 25786126 PMCID: PMC4364906 DOI: 10.1371/journal.pone.0118977] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/27/2015] [Indexed: 11/19/2022] Open
Abstract
Antibody drug conjugates (ADCs) represent novel anti-cancer modalities engineered to specifically target and kill tumor cells expressing corresponding antigens. Due to their large size and their complex kinetics, these therapeutic agents often face heterogeneous distributions in tumors, leading to large untargeted regions that escape therapy. We present a modeling framework which includes the systemic distribution, vascular permeability, interstitial transport, as well as binding and payload release kinetics of ADC-therapeutic agents in mouse xenografts. We focused, in particular, on receptor dynamics such as endocytic trafficking mechanisms within cancer cells, to simulate their impact on tumor mass shrinkage upon ADC administration. Our model identified undesirable tumor properties that can impair ADC tissue homogeneity, further compromising ADC success, and explored ADC design optimization scenarios to counteract upon such unfavorable intrinsic tumor tissue attributes. We further demonstrated the profound impact of cytotoxic payload release mechanisms and the role of bystander killing effects on tumor shrinkage. This model platform affords a customizable simulation environment which can aid with experimental data interpretation and the design of ADC therapeutic treatments.
Collapse
Affiliation(s)
- Christina Vasalou
- Advanced Quantitative Sciences, Novartis, Cambridge, MA, United States of America
| | - Gabriel Helmlinger
- Advanced Quantitative Sciences, Novartis, Cambridge, MA, United States of America
| | - Bruce Gomes
- Advanced Quantitative Sciences, Novartis, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
16
|
Haslene-Hox H, Oveland E, Woie K, Salvesen HB, Tenstad O, Wiig H. Distribution volumes of macromolecules in human ovarian and endometrial cancers--effects of extracellular matrix structure. Am J Physiol Heart Circ Physiol 2014; 308:H18-28. [PMID: 25380817 DOI: 10.1152/ajpheart.00672.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Elements of the extracellular matrix (ECM), notably collagen and glucosaminoglycans, will restrict part of the space available for soluble macromolecules simply because the molecules cannot occupy the same space. This phenomenon may influence macromolecular drug uptake. To study the influence of steric and charge effects of the ECM on the distribution volumes of macromolecules in human healthy and malignant gynecologic tissues we used as probes 15 abundant plasma proteins quantified by high-resolution mass spectrometry. The available distribution volume (VA) of albumin was increased in ovarian carcinoma compared with healthy ovarian tissue. Furthermore, VA of plasma proteins between 40 and 190 kDa decreased with size for endometrial carcinoma and healthy ovarian tissue, but was independent of molecular weight for the ovarian carcinomas. An effect of charge on distribution volume was only found in healthy ovaries, which had lower hydration and high collagen content, indicating that a condensed interstitium increases the influence of negative charges. A number of earlier suggested biomarker candidates were detected in increased amounts in malignant tissue, e.g., stathmin and spindlin-1, showing that interstitial fluid, even when unfractionated, can be a valuable source for tissue-specific proteins. We demonstrate that the distribution of abundant plasma proteins in the interstitium can be elucidated by mass spectrometry methods and depends markedly on hydration and ECM structure. Our data can be used in modeling of drug uptake, and give indications on ECM components to be targeted to increase the uptake of macromolecular substances.
Collapse
Affiliation(s)
| | - Eystein Oveland
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kathrine Woie
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway; and
| | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway; and Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway;
| |
Collapse
|
17
|
Sagstad SJ, Oveland E, Karlsen TV, Haslene-Hox H, Tenstad O, Wiig H. Age-related changes in rat dermal extracellular matrix composition affect the distribution of plasma proteins as a function of size and charge. Am J Physiol Heart Circ Physiol 2014; 308:H29-38. [PMID: 25362136 DOI: 10.1152/ajpheart.00545.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Collagen and glycosaminoglycans (GAGs) constituting the ECM may limit the space available and thus exclude macromolecules from a fraction of the interstitial fluid (IF) phase. This exclusion phenomenon is of importance for transcapillary fluid and solute exchange. The purpose of the study was to examine the range of interstitial exclusion in rat skin by using probes within a span of molecular weights and electrical charge and also to test if a change in interstitial composition, occurring as a consequence of aging, affected exclusion. To this end, we used a novel approach, involving the exact determination of albumin concentration and mass in IF and tissue eluate by HPLC and thereafter, expressing the corresponding numbers relative to albumin for a set of probe proteins assessed by quantitative proteomics. Albumin was excluded from 55±4% (n=8) of the extracellular fluid phase. There was a highly significant, positive correlation between probe Stokes-Einstein (SE) radius and fractional excluded volume (VEF), described by VEF=0.078·SE radius+0.269 (P<0.001), and oppositely, a negative correlation between probe isoelectric point (pI) and exclusion for proteins with comparable size, VEF=-0.036·pI+0.719 (P=0.04). Aging resulted in a significant reduction in skin hydration and sulfated GAGs, a moderate increase in hyaluronan, and a corresponding, reduced VEF for albumin and the other macromolecular probes. Our findings suggest that the changes in the ECM in aged skin may result in delayed adjustments of fluid perturbations and reduced ability for salt storage.
Collapse
Affiliation(s)
| | - Eystein Oveland
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
18
|
Haslene-Hox H, Madani A, Berg KCG, Woie K, Salvesen HB, Wiig H, Tenstad O. Quantification of the concentration gradient of biomarkers between ovarian carcinoma interstitial fluid and blood. BBA CLINICAL 2014; 2:18-23. [PMID: 26673827 PMCID: PMC4633919 DOI: 10.1016/j.bbacli.2014.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/25/2022]
Abstract
Background Tumor interstitial fluid (TIF) rather than plasma should be used in cancer biomarker discovery because of the anticipated higher concentration of locally produced proteins in the tumor microenvironment. Nevertheless, the actual TIF-to-plasma gradient of tumor specific proteins has not been quantified. We present the proof-of-concept for the quantification of the postulated gradient between TIF and plasma. Methods TIF was collected by centrifugation from serous (n = 19), endometrioid (n = 9) and clear cell (n = 3) ovarian carcinomas with early (n = 15) and late stage (n = 16) disease in grades 1 (n = 2), 2 (n = 8) and 3 (n = 17), and ELISA was used for the determination of CA-125, osteopontin and VEGF-A. Results All three markers were significantly up-regulated in TIF compared with plasma (p < 0.0001). The TIF-to-plasma ratio of the ovarian cancer biomarker CA-125 ranged from 1.4 to 24,300 (median = 194) and was inversely correlated to stage (p = 0.0006). The cancer related osteopontin and VEGF-A had TIF-to-plasma ratios ranging from 1 to 62 (median = 15) and 2 to 1040 (median = 59), respectively. The ratios were not affected by tumor stage, indicative of more widespread protein expression. Conclusion We present absolute quantitative data on the TIF-to-plasma gradient of selected proteins in the tumor microenvironment, and demonstrate a substantial and stage dependent gradient for CA-125 between TIF and plasma, suggesting a relation between total tumor burden and tissue-to-plasma gradient. General significance We present novel quantitative data on biomarker concentration in the tumor microenvironment, and a new strategy for biomarker selection, applicable in future biomarker studies. Quantification of CA-125, VEGF and osteopontin in tumor interstitial fluid (TIF) A large TIF-to-plasma gradient was observed for CA-125, the highest in early stage. Lower VEGF and osteopontin gradient indicate more widespread protein expression.
Collapse
Affiliation(s)
- Hanne Haslene-Hox
- Department of Biomedicine, University of Bergen, Norway ; Department of Bioprocess Technology, SINTEF Materials and Chemistry, Trondheim, Norway
| | - Amina Madani
- Department of Biomedicine, University of Bergen, Norway
| | - Kaja C G Berg
- Department of Biomedicine, University of Bergen, Norway
| | - Kathrine Woie
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway ; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Norway
| |
Collapse
|
19
|
Øien AH, Justad SR, Tenstad O, Wiig H. Effects of hydration on steric and electric charge-induced interstitial volume exclusion--a model. Biophys J 2014; 105:1276-84. [PMID: 24010671 DOI: 10.1016/j.bpj.2013.07.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/26/2013] [Accepted: 07/24/2013] [Indexed: 11/17/2022] Open
Abstract
The presence of collagen and charged macromolecules like glycosaminoglycans (GAGs) in the interstitial space limits the space available for plasma proteins and other macromolecules. This phenomenon, known as interstitial exclusion, is of importance for interstitial fluid volume regulation. Physical/mathematical models are presented for calculating the exclusion of electrically charged and neutral macromolecules that equilibrate in the interstitium under various degrees of hydration. Here, a central hypothesis is that the swelling of highly electrically charged GAGs with increased hydration shields parts of the neutral collagen of the interstitial matrix from interacting with electrically charged macromolecules, such that exclusion of charged macromolecules exhibits change due to steric and charge effects. GAGs are also thought to allow relatively small neutral, but also charged macromolecules neutralized by a very high ionic strength, diffuse into the interior of GAGs, whereas larger macromolecules may not. Thus, in the model, relatively small electrically charged macromolecules, such as human serum albumin, and larger neutral macromolecules such as IgG, will have quite similar total volume exclusion properties in the interstitium. Our results are in agreement with ex vivo and in vivo experiments, and suggest that the charge of GAGs or macromolecular drugs may be targeted to increase the tissue uptake of macromolecular therapeutic agents.
Collapse
Key Words
- 1-particles
- 2-particles
- 3-particles
- A
- A [as subscript]
- DCL(z)
- Debye length
- GAGs, glycosaminoclycans [also as subscript]
- H(z)
- L
- N1in
- O
- Q(1), Q(2)
- T
- VO
- VT
- albumin- or macromolecules in general [also as subscript]
- available
- available volume of charged particle
- available volume of small neutral macroparticle
- available volume per unit volume for large neutral macroparticles
- average volume density of macromolecules, as albumin, in cell
- axial length of equilibration cell
- charge part per unit volume of excluded volume
- circular cross-section area of equilibration cell
- collagen particles [also as subscript]
- core radius of GAG
- d1, d2 = 2H(z), d3
- dCL
- diameters of 1-, 2-, and 3-particles
- distance between two side-by-side GAGs- or collagen particle centers
- distance of closest approach in GAG-albumin interactions
- distance of closest approach in albumin-albumin interactions
- effective density of collagen particles
- electrical charge on particle I
- excluded volume per unit volume for large neutral macroparticles
- excluded volume per unit volume of charged macroparticle
- excluded volume per unit volume of small neutral macroparticle
- highest thickness value
- hydration
- inside cell tissue [also as subscript]
- l
- lowest thickness value
- n1T(z), n1O(z)
- n1in
- n2, n3
- n3eff(z)
- number densities of 2- and 3-particles
- number of macromolecules, as albumin, poured into cell
- outside cell tissue [also as subscript]
- outside tissue-cell volume
- p1
- pressure of 1-particles
- r
- radial distance
- radius of GAGs as function of tissue thickness
- rmin
- thickness of tissue sample
- tissue-cell volume
- volume density of 1-particles in tissue, outside tissue
- volume of collagen-, GAG- and macroparticle material per unit volume of tissue
- volume of water per unit volume of tissue (water content)
- z
- z1
- z2
- ζ
- λD
- νA
- νA0G
- νAO
- νE
- νE00
- νE0G
- νEO
- νH2O
- νQ
Collapse
Affiliation(s)
- Alf H Øien
- Department of Mathematics, University of Bergen, Bergen, Norway
| | | | | | | |
Collapse
|
20
|
Han H, Davis ME. Targeted nanoparticles assembled via complexation of boronic-acid-containing targeting moieties to diol-containing polymers. Bioconjug Chem 2013; 24:669-77. [PMID: 23461746 DOI: 10.1021/bc300640j] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The delivery of therapeutics via nanoscaled vehicles for solid cancer treatment can be enhanced by the incorporation of a targeting capability. Here, we describe a new method for assembling a targeted nanoparticle that utilizes the reversible covalent complexation between boronic acids and diols to achieve a targeted nanoparticle for the delivery of the anticancer drug camptothecin (CPT). CPT is conjugated to a biocompatible, hydrophilic copolymer of mucic acid and PEG (MAP). When this polymer-drug conjugate is placed in water, it self-assembles into MAP-CPT nanoparticles of ca. 30 nm (diameter) and slightly negative zeta potential. The antibody Herceptin is attached to a boronic acid via a polyethylene glycol (PEG) spacer, and this boronic acid-containing targeting moiety is complexed with the diol-containing MAP to form a targeted MAP-CPT nanoparticle. The addition of Herceptin targeting agent to the MAP-CPT nanoparticles yields targeted MAP-CPT nanoparticles with increased nanoparticle size to ca. 40 nm (diameter). The main mechanisms of CPT release from MAP-CPT nanoparticles are found by in vitro analysis to be hydrolysis and nanoparticle disruption by fat. Cellular uptake of nanoparticles is enhanced by 70% compared to nontargeted version by the incorporation of a single Herceptin antibody targeting agent per nanoparticle. This single Herceptin antibody targeted MAP-CPT nanoparticle system carries ca. 60 CPT molecules per nanoparticle and shows prolonged plasma circulation with an elimination half-life of 21.2 h and AUC value of 2766 μg.h/mL at a 10 mg CPT/kg tail vein injection in mice.
Collapse
Affiliation(s)
- Han Han
- Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | |
Collapse
|
21
|
Hasinoff BB, Patel D, Wu X. The Dual-Targeted HER1/HER2 Tyrosine Kinase Inhibitor Lapatinib Strongly Potentiates the Cardiac Myocyte-Damaging Effects of Doxorubicin. Cardiovasc Toxicol 2012; 13:33-47. [DOI: 10.1007/s12012-012-9183-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
22
|
Wiig H, Swartz MA. Interstitial Fluid and Lymph Formation and Transport: Physiological Regulation and Roles in Inflammation and Cancer. Physiol Rev 2012; 92:1005-60. [PMID: 22811424 DOI: 10.1152/physrev.00037.2011] [Citation(s) in RCA: 456] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The interstitium describes the fluid, proteins, solutes, and the extracellular matrix (ECM) that comprise the cellular microenvironment in tissues. Its alterations are fundamental to changes in cell function in inflammation, pathogenesis, and cancer. Interstitial fluid (IF) is created by transcapillary filtration and cleared by lymphatic vessels. Herein we discuss the biophysical, biomechanical, and functional implications of IF in normal and pathological tissue states from both fluid balance and cell function perspectives. We also discuss analysis methods to access IF, which enables quantification of the cellular microenvironment; such methods have demonstrated, for example, that there can be dramatic gradients from tissue to plasma during inflammation and that tumor IF is hypoxic and acidic compared with subcutaneous IF and plasma. Accumulated recent data show that IF and its convection through the interstitium and delivery to the lymph nodes have many and diverse biological effects, including in ECM reorganization, cell migration, and capillary morphogenesis as well as in immunity and peripheral tolerance. This review integrates the biophysical, biomechanical, and biological aspects of interstitial and lymph fluid and its transport in tissue physiology, pathophysiology, and immune regulation.
Collapse
Affiliation(s)
- Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway; and Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Melody A. Swartz
- Department of Biomedicine, University of Bergen, Bergen, Norway; and Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
23
|
Boswell CA, Bumbaca D, Fielder PJ, Khawli LA. Compartmental tissue distribution of antibody therapeutics: experimental approaches and interpretations. AAPS JOURNAL 2012; 14:612-8. [PMID: 22648903 DOI: 10.1208/s12248-012-9374-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/16/2012] [Indexed: 01/12/2023]
Abstract
Monoclonal antibodies have provided many validated and potential new therapeutic candidates for various diseases encompassing the realms of neurology, ophthalmology, immunology, and especially oncology. The mechanism of action for these biological molecules typically involves specific binding to a soluble ligand or cell surface protein in order to block or alter a molecular pathway, induce a desired cellular response, or deplete a target cell. Many antigens reside within the interstitial space, the fluid-filled compartment that lies between the outer endothelial vessel wall and the plasma membranes of cells. This mini-review examines the concepts relevant to the kinetics and behavior of antibodies within the interstitium with a special emphasis on radiometric measurement of quantitative pharmacology. Molecular probes are discussed to outline chemical techniques, selection criteria, data interpretation, and relevance to the study of antibody pharmacokinetics. The importance of studying the tissue uptake of antibodies at a compartmental level is highlighted, including a brief overview of receptor occupancy and its interpretation in radiotracer studies. Experimental methods for measuring the spatial composition of tissues are examined in terms of relative vascular, interstitial, and cellular volumes using solid tumors as a representative example. Experimental methods and physiologically based pharmacokinetic modeling are introduced as distinct approaches to distinguish between free and bound fractions of interstitial antibody. Overall, the review outlines the available methods for pharmacokinetic measurements of antibodies and physiological measurements of the compartments that they occupy, while emphasizing that such approaches may not fully capture the complexities of dynamic, heterogeneous tumors and other tissues.
Collapse
Affiliation(s)
- C Andrew Boswell
- Genentech Research and Early Development, South San Francisco, California 94080, USA.
| | | | | | | |
Collapse
|
24
|
Zhao J, Mi Y, Liu Y, Feng SS. Quantitative control of targeting effect of anticancer drugs formulated by ligand-conjugated nanoparticles of biodegradable copolymer blend. Biomaterials 2011; 33:1948-58. [PMID: 22153869 DOI: 10.1016/j.biomaterials.2011.11.051] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/20/2011] [Indexed: 02/05/2023]
Abstract
There have been two strategies developed in the recent literature for quantitative control of the targeting effects for drug delivery by ligand-conjugated nanoparticles of biodegradable copolymer blend such as PLGA/PLGA-PEG, i.e. the pre-conjugation strategy and the post-conjugation strategy, in which the ligand conjugation was made before and after the nanoparticle formulation respectively. This research developed another drug delivery system of the PLA-TPGS/TPGS-COOH copolymer blend and further improved the post-conjugation strategy to precisely control the targeting effects by two ways: one is to adjust the PLA-TPGS:TPGS-COOH copolymer blend ratio in the nanoparticle formulation process, which provides a way for coarse control, and another is to control the feeding concentration of the ligand in the herceptin conjugation process, which further provides a fine control. Herceptin conjugation was visualized by the FETEM with immumogold labeling and further quantified by the two techniques, i.e. the Bradford assay and the flow cytometry to confirm each other. The positive correlation between the surface density of the ligand and the cellular internalization as well as the cytotoxicity of the nanoparticle formulations was assessed, which demonstrated that the strategy developed in this research is simple and feasible, which can precisely control the targeting effects of the nanoparticles of biodegradable polymers as well as other nanocarriers such as micelles and liposomes.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Bioengineering, National University of Singapore, Singapore 117576, Singapore
| | | | | | | |
Collapse
|
25
|
Thurber GM, Weissleder R. Quantitating antibody uptake in vivo: conditional dependence on antigen expression levels. Mol Imaging Biol 2011; 13:623-32. [PMID: 20809210 DOI: 10.1007/s11307-010-0397-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE Antibodies form an important class of cancer therapeutics, and there is intense interest in using them for imaging applications in diagnosis and monitoring of cancer treatment. Despite the expanding body of knowledge describing pharmacokinetic and pharmacodynamic interactions of antibodies in vivo, discrepancies remain over the effect of antigen expression level on tumoral uptake with some reports indicating a relationship between uptake and expression and others showing no correlation. PROCEDURES Using a cell line with high epithelial cell adhesion molecule expression and moderate epidermal growth factor receptor expression, fluorescent antibodies with similar plasma clearance were imaged in vivo. A mathematical model and mouse xenograft experiments were used to describe the effect of antigen expression on uptake of these high-affinity antibodies. RESULTS As predicted by the theoretical model, under subsaturating conditions, uptake of the antibodies in such tumors is similar because localization of both probes is limited by delivery from the vasculature. In a separate experiment, when the tumor is saturated, the uptake becomes dependent on the number of available binding sites. In addition, targeting of small micrometastases is shown to be higher than larger vascularized tumors. CONCLUSIONS These results are consistent with the prediction that high affinity antibody uptake is dependent on antigen expression levels for saturating doses and delivery for subsaturating doses. It is imperative for any probe to understand whether quantitative uptake is a measure of biomarker expression or transport to the region of interest. The data provide support for a predictive theoretical model of antibody uptake, enabling it to be used as a starting point for the design of more efficacious therapies and timely quantitative imaging probes.
Collapse
Affiliation(s)
- Greg M Thurber
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | |
Collapse
|
26
|
Wasik AM, Christensson B, Sander B. The role of cannabinoid receptors and the endocannabinoid system in mantle cell lymphoma and other non-Hodgkin lymphomas. Semin Cancer Biol 2011; 21:313-21. [PMID: 22024769 DOI: 10.1016/j.semcancer.2011.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The initiating oncogenic event in mantle cell lymphoma (MCL) is the translocation of cyclin D1, t(11;14)(q13;q32). However, other genetic aberrations are necessary for an overt lymphoma to arise. Like other B cell lymphomas, MCL at some points during the oncogenesis is dependent on interactions with other cells and factors in the microenvironment. The G protein coupled receptors cannabinoid receptors 1 and 2 (CB1 and CB2) are expressed at low levels on non-malignant lymphocytes and at higher levels in MCL and other lymphoma subtypes. In this review we give an overview of what is known on the role of the cannabinoid receptors and their ligands in lymphoma as compared to non-malignant T and B lymphocytes. In MCL cannabinoids mainly reduce cell proliferation and induce cell death. Importantly, our recent findings demonstrate that cannabinoids may induce either apoptosis or another type of programmed cell death, cytoplasmic vacuolation/paraptosis in MCL. The signalling to death has been partly characterized. Even though cannabinoid receptors seem to be expressed in many other types of B cell lymphoma, the functional role of cannabinoid receptor targeting is yet largely unknown. In non-malignant B and T lymphocytes, cannabinoid receptors are up-regulated in response to antigen receptor signalling or CD40. For T lymphocytes IL-4 has also a crucial role in transcriptional regulation of CB1. In lymphocytes, cannabinoid act in several ways - by affecting cell migration, cytokine response, at high doses inhibit cell proliferation and inducing cell death. The possible role for the endocannabinoid system in the immune microenvironment of lymphoma is discussed.
Collapse
Affiliation(s)
- Agata M Wasik
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital Huddinge, SE 141 86 Stockholm, Sweden.
| | | | | |
Collapse
|
27
|
Silvestrini M, Schiavuta P, Scopece P, Pecchielan G, Moretto L, Ugo P. Modification of nanoelectrode ensembles by thiols and disulfides to prevent non specific adsorption of proteins. Electrochim Acta 2011. [DOI: 10.1016/j.electacta.2011.06.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
28
|
Mi Y, Li K, Liu Y, Pu KY, Liu B, Feng SS. Herceptin functionalized polyhedral oligomeric silsesquioxane - conjugated oligomers - silica/iron oxide nanoparticles for tumor cell sorting and detection. Biomaterials 2011; 32:8226-33. [PMID: 21816464 DOI: 10.1016/j.biomaterials.2011.07.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 07/13/2011] [Indexed: 11/30/2022]
Abstract
Sorting and detection of circulating tumor cells (CTC) in peripheral blood as an efficient and non-invasive method to diagnose cancer have recently attracted much attention. In this article, we developed a multiply-engineered nanoparticle system for CTC sorting and detection, which consists of (1) conjugated oligomer (CO) as fluorescence signal source, (2) polyhedral oligomeric silsesquioxanes (POSS) scaffold for CO localization for better fluorescent effects, (3) silica nanoparticles (SiNPs) as formulation matrix of the POSS containing CO, (4) iron oxide (IO) layer on the silica nanoparticles (IO-SiNPs) for magnetic collection, and (5) herceptin surface functionalization of the IO-SiNPs to target cancer cells of HER2 overexpression. Such a multiply-engineered structure can be used for either traditional immunomagnetic methods or microfluidic devices for CTC sorting and detection.
Collapse
Affiliation(s)
- Yu Mi
- Department of Chemical & Biomolecular Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| | | | | | | | | | | |
Collapse
|
29
|
Petronzelli F, Anastasi AM, Pelliccia A, Santapaola D, Albertoni C, Rosi A, Leoni B, Ferrari LE, Paganelli G, Gramiccioli G, Pesce D, Alfano AM, Stasi MA, De Santis R. Preclinical pharmacology and safety of a novel avidin derivative for tissue-targeted delivery of radiolabelled biotin. Basic Clin Pharmacol Toxicol 2011; 109:145-55. [PMID: 21426491 DOI: 10.1111/j.1742-7843.2011.00701.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We recently described an oxidized avidin variant, named AvidinOX(®) , which is a product that chemically links to tissue proteins while maintaining the capacity to uptake intravenously administered biotin. Such product proved to be successful in targeting radionuclide therapy in a mouse model of inoperable breast cancer. Here, we show that the uptake of a single or multiple doses of biotin (up to five times), by the tissue-bound AvidinOX(®) , is stable for 2 weeks. Taking into account that oxidized avidin is the first chemically reactive protein to be proposed for clinical use, we evaluated its tolerability, immunogenicity and mutagenicity. Present in vitro data indicate that AvidinOX(®) (up to 10 μg/5 × 10(5) cells) does not affect cell viability or proliferation of PC3 human prostate cancer or 3T3 mouse fibroblast cell lines as well as primary mouse spleen cells. Safety pharmacology and toxicology studies were conducted using AvidinOX(®) up to the highest concentration compatible with its solubility (about 12 mg/mL), representing four times the product concentration intended for human use, and in the maximum administrable volume compatible with each study system. The intramuscular administration in rat and monkey induced a moderate to strong inflammatory response particularly after a second administration and consistently with the induction of an immune response. Interestingly, the intramuscular administration of AvidinOX(®) to rodents and monkeys exhibiting very high anti-avidin antibody titres was well tolerated with no systemic symptoms of any kind. Intravenous administration of AvidinOX(®) , performed to mimic an accidental injection of the dose intended for a local administration (15 μL of 3.3 mg/mL solution), showed significant localization of the product into the spleen not associated with uptake of the radiolabelled biotin intravenously injected after 24 hr, thus suggesting rapid inactivation. No mutagenic activity was induced by oxidized avidin in prokaryotic and eukaryotic cells. Overall, the present data indicate that AvidinOX(®) is well tolerated in rodents and non-human primates, thus supporting its clinical use within protocols of radionuclide therapy of inoperable tumour lesions.
Collapse
|
30
|
Ludwig DB, Trotter JT, Gabrielson JP, Carpenter JF, Randolph TW. Flow cytometry: a promising technique for the study of silicone oil-induced particulate formation in protein formulations. Anal Biochem 2010; 410:191-9. [PMID: 21146492 DOI: 10.1016/j.ab.2010.12.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/30/2010] [Accepted: 12/06/2010] [Indexed: 11/30/2022]
Abstract
Subvisible particles in formulations intended for parenteral administration are of concern in the biopharmaceutical industry. However, monitoring and control of subvisible particulates can be complicated by formulation components, such as the silicone oil used for the lubrication of prefilled syringes, and it is difficult to differentiate microdroplets of silicone oil from particles formed by aggregated protein. In this study, we demonstrate the ability of flow cytometry to resolve mixtures comprising subvisible bovine serum albumin (BSA) aggregate particles and silicone oil emulsion droplets with adsorbed BSA. Flow cytometry was also used to investigate the effects of silicone oil emulsions on the stability of BSA, lysozyme, abatacept, and trastuzumab formulations containing surfactant, sodium chloride, or sucrose. To aid in particle characterization, the fluorescence detection capabilities of flow cytometry were exploited by staining silicone oil with BODIPY 493/503 and model proteins with Alexa Fluor 647. Flow cytometric analyses revealed that silicone oil emulsions induced the loss of soluble protein via protein adsorption onto the silicone oil droplet surface. The addition of surfactant prevented protein from adsorbing onto the surface of silicone oil droplets. There was minimal formation of homogeneous protein aggregates due to exposure to silicone oil droplets, although oil droplets with surface-adsorbed trastuzumab exhibited flocculation. The results of this study demonstrate the utility of flow cytometry as an analytical tool for monitoring the effects of subvisible silicone oil droplets on the stability of protein formulations.
Collapse
Affiliation(s)
- D Brett Ludwig
- University of Colorado Center for Pharmaceutical Biotechnology, Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
| | | | | | | | | |
Collapse
|
31
|
Boswell CA, Tesar DB, Mukhyala K, Theil FP, Fielder PJ, Khawli LA. Effects of Charge on Antibody Tissue Distribution and Pharmacokinetics. Bioconjug Chem 2010; 21:2153-63. [DOI: 10.1021/bc100261d] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- C. Andrew Boswell
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| | - Devin B. Tesar
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| | - Kiran Mukhyala
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| | - Frank-Peter Theil
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| | - Paul J. Fielder
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| | - Leslie A. Khawli
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Department of Antibody Engineering, and Department of Bioinformatics, Genentech Research and Early Development, South San Francisco, California 94080, United States
| |
Collapse
|
32
|
Andrew JS, Anglin EJ, Wu EC, Chen MY, Cheng L, Freeman WR, Sailor MJ. Sustained Release of a Monoclonal Antibody from Electrochemically Prepared Mesoporous Silicon Oxide. ADVANCED FUNCTIONAL MATERIALS 2010; 20:4168-4174. [PMID: 21274422 PMCID: PMC3026353 DOI: 10.1002/adfm.201000907] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanostructured mesoporous silica (SiO(2)) films are used to load and release the monoclonal antibody bevacizumab (Avastin) in vitro. A biocompatible and biodegradable form of mesoporous SiO(2) is prepared by electrochemical etching of single crystalline Si, followed by thermal oxidation in air at 800 °C. Porous SiO(2) exhibits a negative surface charge at physiological pH (7.4), allowing it to spontaneously adsorb the positively charged antibody from an aqueous phosphate buffered saline solution. This electrostatic adsorption allows bevacizumab to be concentrated by >100× (300 mg bevacziumab per gram of porous SiO(2) when loaded from a 1 mg mL(-1) solution of bevacziumab). Drug loading is monitored by optical interferometric measurements of the thin porous film. A two-component Bruggeman effective medium model is employed to calculate percent porosity and film thickness, and is further used to determine the extent of drug loading into the porous SiO(2) film. In vitro drug release profiles are characterized by an enzyme-linked immunosorbent assay (ELISA), which confirms that the antibody is released in its active, VEGF-binding form. The nanostructured delivery system described here provides a sustained release of the monoclonal antibody where approximately 98% of drug is released over a period of one month.
Collapse
Affiliation(s)
- Jennifer S. Andrew
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500, Gilman Dr, La Jolla, CA 92093 (USA)
| | - Emily J. Anglin
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500, Gilman Dr, La Jolla, CA 92093 (USA)
| | - Elizabeth C. Wu
- Bioengineering Department, University of California, San Diego, CA 92093-0412 (USA)
| | - Michelle Y. Chen
- Bioengineering Department, University of California, San Diego, CA 92093-0412 (USA)
| | - Lingyun Cheng
- Jacobs Retina Center at Shiley Eye Center, University of California, San Diego, San Diego, CA 92092 (USA)
| | - William R. Freeman
- Jacobs Retina Center at Shiley Eye Center, University of California, San Diego, San Diego, CA 92092 (USA)
| | | |
Collapse
|
33
|
A Time-Dependent Electrodiffusion-Convection Model for Charged Macromolecule Transport Across the Microvessel Wall and in the Interstitial Space. Cell Mol Bioeng 2009. [DOI: 10.1007/s12195-009-0074-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
34
|
Eikenberry S. A tumor cord model for doxorubicin delivery and dose optimization in solid tumors. Theor Biol Med Model 2009; 6:16. [PMID: 19664243 PMCID: PMC2736154 DOI: 10.1186/1742-4682-6-16] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 08/09/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Doxorubicin is a common anticancer agent used in the treatment of a number of neoplasms, with the lifetime dose limited due to the potential for cardiotoxocity. This has motivated efforts to develop optimal dosage regimes that maximize anti-tumor activity while minimizing cardiac toxicity, which is correlated with peak plasma concentration. Doxorubicin is characterized by poor penetration from tumoral vessels into the tumor mass, due to the highly irregular tumor vasculature. I model the delivery of a soluble drug from the vasculature to a solid tumor using a tumor cord model and examine the penetration of doxorubicin under different dosage regimes and tumor microenvironments. METHODS A coupled ODE-PDE model is employed where drug is transported from the vasculature into a tumor cord domain according to the principle of solute transport. Within the tumor cord, extracellular drug diffuses and saturable pharmacokinetics govern uptake and efflux by cancer cells. Cancer cell death is also determined as a function of peak intracellular drug concentration. RESULTS The model predicts that transport to the tumor cord from the vasculature is dominated by diffusive transport of free drug during the initial plasma drug distribution phase. I characterize the effect of all parameters describing the tumor microenvironment on drug delivery, and large intercapillary distance is predicted to be a major barrier to drug delivery. Comparing continuous drug infusion with bolus injection shows that the optimum infusion time depends upon the drug dose, with bolus injection best for low-dose therapy but short infusions better for high doses. Simulations of multiple treatments suggest that additional treatments have similar efficacy in terms of cell mortality, but drug penetration is limited. Moreover, fractionating a single large dose into several smaller doses slightly improves anti-tumor efficacy. CONCLUSION Drug infusion time has a significant effect on the spatial profile of cell mortality within tumor cord systems. Therefore, extending infusion times (up to 2 hours) and fractionating large doses are two strategies that may preserve or increase anti-tumor activity and reduce cardiotoxicity by decreasing peak plasma concentration. However, even under optimal conditions, doxorubicin may have limited delivery into advanced solid tumors.
Collapse
Affiliation(s)
- Steffen Eikenberry
- Department of Mathematics and Statistics, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
35
|
Abstract
Over many decades, several attempts have been made to treat hypertension using a vaccination strategy to inhibit the renin-angiotensin system. Renin vaccination successfully inhibited renin activity and reduced blood pressure in animal models, but caused autoimmune disease of the kidney. By contrast, most previous studies of angiotensin vaccination failed to reduce blood pressure in animal models, despite producing high titers of antibodies that prevented the pressor response to exogenous angiotensins. Angiotensin vaccination is being revisited as a strategy for treating hypertension, in the hope that new conjugates of angiotensin I and angiotensin II will produce antibodies of sufficient titer and affinity to reduce blood pressure in patients with hypertension. A recent clinical study of angiotensin II vaccination provided some cause for optimism, but many hurdles remain before this strategy can compete with current oral medications for hypertension.
Collapse
|
36
|
Mucelli SP, Zamuner M, Tormen M, Stanta G, Ugo P. Nanoelectrode ensembles as recognition platform for electrochemical immunosensors. Biosens Bioelectron 2008; 23:1900-3. [DOI: 10.1016/j.bios.2008.02.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 02/26/2008] [Accepted: 02/29/2008] [Indexed: 10/22/2022]
|
37
|
Baker JHE, Lindquist KE, Huxham LA, Kyle AH, Sy JT, Minchinton AI. Direct visualization of heterogeneous extravascular distribution of trastuzumab in human epidermal growth factor receptor type 2 overexpressing xenografts. Clin Cancer Res 2008; 14:2171-9. [PMID: 18381959 DOI: 10.1158/1078-0432.ccr-07-4465] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The high molecular weight and binding affinity of trastuzumab, a monoclonal antibody in use for treatment of breast cancers overexpressing human epidermal growth factor receptor type 2 (HER2), in combination with microenvironmental factors, may limit its distribution and efficacy. We assessed and mapped the distribution of systemically given, unlabeled trastuzumab at micrometer resolution in tumor xenografts using immunohistochemistry. EXPERIMENTAL DESIGN Mice bearing MDA-435/LCC6(HER2) xenografts were given single doses of 4 or 20 mg/kg unlabeled trastuzumab with tumor harvest at various time points thereafter; bound trastuzumab was imaged directly in tumor cryosections using fluorescently tagged antihuman secondary antibodies. Combinations of additional markers, including HER2, 5-bromo-2-deoxyuridine, CD31, DioC(7)(3), desmin, and collagen IV were also mapped on the same tumor sections. RESULTS Distribution of trastuzumab in MDA-435/LCC6(HER2) tumors is found to be heterogeneous, with tumor margins saturating more thoroughly in doses and times analyzed. Considerable intervessel heterogeneity is also seen. For example, in unsaturated tissues, there remain perfused vessels without any trastuzumab in addition to vessels with a few layers of positively stained perivascular cells, in addition to vessels with bound drug up to 150 microm away. This heterogeneity is independent of HER2 expression, microvessel density, and perfusion. A slightly greater proportion of vessels were associated with pericytes in sections with greater trastuzumab saturation, but this would not adequately account for observed heterogeneous trastuzumab distribution. CONCLUSIONS Complete penetration of trastuzumab in tumor tissue was not seen in our study, leaving the possibility that inadequate distribution may represent a mechanism for resistance to trastuzumab.
Collapse
Affiliation(s)
- Jennifer H E Baker
- Medical Biophysics Department, British Columbia Cancer Research Center, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
38
|
Semaeva E, Tenstad O, Bletsa A, Gjerde EAB, Wiig H. Isolation of rat trachea interstitial fluid and demonstration of local cytokine production in lipopolysaccharide-induced systemic inflammation. J Appl Physiol (1985) 2008; 104:809-20. [PMID: 18187613 DOI: 10.1152/japplphysiol.00846.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Access to interstitial fluid from trachea is important for understanding tracheal microcirculation and pathophysiology. We tested whether a centrifugation method could be applied to isolate this fluid in rats by exposing excised trachea to G forces up to 609 g. The ratio between the concentration of the equilibrated extracellular tracer 51Cr-labeled EDTA in fluid isolated at 239 g and plasma averaged 0.94 +/- 0.03 (n = 14), suggesting that contamination from the intracellular fluid phase was negligible. The protein pattern of the isolated fluid resembled plasma closely and had a protein concentration 83% of that in plasma. The colloid osmotic pressure in the centrifugate in controls (n = 5) was 18.8 +/- 0.6 mmHg with a corresponding pressure in plasma of 22 +/- 1.5 mmHg, whereas after overhydration (n = 5) these pressures fell to 9.8 +/- 0.4 and 11.9 +/- 0.4 mmHg, respectively. We measured inflammatory cytokine concentration in serum, interstitial fluid, and bronchoalveolar lavage fluid in LPS-induced inflammation. In control animals, low levels of IL-1 beta, IL-6, and TNF-alpha in serum, trachea interstitial fluid, and bronchoalveolar lavage fluid were detected. LPS resulted in a significantly higher concentration in IL-1 beta and IL-6 in interstitial fluid than in serum, showing a local production. To conclude, we have shown that interstitial fluid can be isolated from trachea by centrifugation and that trachea interstitial fluid has a high protein concentration and colloid osmotic pressure relative to plasma. Trachea interstitial fluid may also reflect lower airways and thus be of importance for understanding, e.g., inflammatory-induced airway obstruction.
Collapse
Affiliation(s)
- Elvira Semaeva
- Dept. of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | | | | | | | | |
Collapse
|
39
|
The endocannabinoid system in cancer-potential therapeutic target? Semin Cancer Biol 2007; 18:176-89. [PMID: 18249558 DOI: 10.1016/j.semcancer.2007.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Accepted: 12/05/2007] [Indexed: 01/13/2023]
Abstract
Endogenous arachidonic acid metabolites with properties similar to compounds of Cannabis sativa Linnaeus, the so-called endocannabinoids, have effects on various types of cancer. Although endocannabinoids and synthetic cannabinoids may have pro-proliferative effects, predominantly inhibitory effects on tumor growth, angiogenesis, migration and metastasis have been described. Remarkably, these effects may be selective for the cancer cells, while normal cells and tissues are spared. Such apparent tumor cell selectivity makes the endocannabinoid system an attractive potential target for cancer therapy. In this review we discuss various means by which the endocannabinoid system may be targeted in cancer and the current knowledge considering the regulation of the endocannabinoid system in malignancy.
Collapse
|
40
|
Immunotoxin and Taxol synergy results from a decrease in shed mesothelin levels in the extracellular space of tumors. Proc Natl Acad Sci U S A 2007; 104:17099-104. [PMID: 17940013 DOI: 10.1073/pnas.0708101104] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recombinant immunotoxins are chimeric proteins composed of the Fv portion of a tumor-specific antibody fused to a toxin. SS1P (CAT-5001) is an immunotoxin composed of an antimesothelin Fv fused to a 38-kDa portion of Pseudomonas exotoxin A. Immunotoxins have been shown to be active in lymphomas and leukemias, but are much less active against solid tumors. We recently reported that Taxol and other chemotherapeutic agents show striking synergistic antitumor activity in mice when immunotoxin SS1P, which targets the mesothelin antigen on solid tumors, is given with Taxol. Using a pair of Taxol-sensitive and Taxol-resistant KB tumors equally sensitive to immunotoxin SS1P, we examined the mechanism of synergy. We show that synergy is only observed with Taxol-sensitive tumors, ruling out an effect of Taxol on endothelial cells. We also show that the KB tumors have high levels of shed mesothelin in their extracellular space; these levels increase with tumor size and, after Taxol treatment, dramatically fall in the drug-sensitive but not the drug-resistant tumors. Because the mesothelin levels in the tumor exceed the levels of SS1P in the tumor, and because shed mesothelin is being continuously released into the circulation at a high rate, we propose that synergy is due to the Taxol-induced fall in shed antigen levels.
Collapse
|
41
|
Ambühl PM, Tissot AC, Fulurija A, Maurer P, Nussberger J, Sabat R, Nief V, Schellekens C, Sladko K, Roubicek K, Pfister T, Rettenbacher M, Volk HD, Wagner F, Müller P, Jennings GT, Bachmann MF. A vaccine for hypertension based on virus-like particles: preclinical efficacy and phase I safety and immunogenicity. J Hypertens 2007; 25:63-72. [PMID: 17143175 DOI: 10.1097/hjh.0b013e32800ff5d6] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Despite the availability of efficacious drugs, the success of treating hypertension is limited by patients' inconsistent drug intake. Immunization against angiotensin II may offer a valuable alternative to conventional drugs for the treatment of hypertension, because vaccines induce relatively long-lasting effects and do not require daily dosing. Here we describe the preclinical development and the phase I clinical trial testing of a virus-like particle (VLP)-based antihypertensive vaccine. METHODS AND RESULTS An angiotensin II-derived peptide was conjugated to the VLP Qbeta (AngQb). AngQb was highly immunogenic in mice and rats. To test for efficacy, spontaneously hypertensive rats (SHR) were immunized with 400 microg AngQb or VLP alone. Group mean systolic blood pressure (SBP) was reduced by up to 21 mmHg (159 +/- 2 versus 180 +/- 5 mmHg, P < 0.001), and total angiotensin II levels (antibody-bound and free) were increased ninefold (85 +/- 20 versus 9 +/- 1 pmol/l, P = 0.002) compared with VLP controls. SHR treated with the angiotensin-converting enzyme (ACE) inhibitor ramipril (1 mg/kg per day by mouth) reached an SBP of 155 +/- 2 mmHg. Twelve healthy volunteers of a placebo-controlled randomized phase I trial were injected once with 100 microg AngQb. Angiotensin II-specific antibodies were raised in all subjects (100% responder rate) and AngQb was well tolerated. CONCLUSIONS AngQb reduces blood pressure in SHR to levels obtained with an ACE inhibitor, and is immunogenic and well tolerated in humans. Therefore, vaccination against angiotensin II has the potential to become a useful antihypertensive treatment providing long-lasting effects and improving patient compliance.
Collapse
|
42
|
Choi J, Credit K, Henderson K, Deverkadra R, He Z, Wiig H, Vanpelt H, Flessner MF. Intraperitoneal immunotherapy for metastatic ovarian carcinoma: Resistance of intratumoral collagen to antibody penetration. Clin Cancer Res 2006; 12:1906-12. [PMID: 16551876 DOI: 10.1158/1078-0432.ccr-05-2141] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Convective transport of macromolecules from the peritoneal cavity into tumor is determined by its hydraulic permeability and the pressure gradient. Previous studies showed that establishing a pressure gradient into the tumor failed to result in significant penetration. This study addresses the hypothesis that the extracellular matrix is the major resistance to the penetration of an i.p. injected antibody. EXPERIMENTAL DESIGN Human ovarian tumors (SKOV-3 and OVCAR-3) were established in the abdominal wall of athymic rats. After anesthesia, the tumor serosal surface was treated for 2 hours with Krebs solution (control), collagenase (37.5 unit/mL), or hyaluronidase (10 unit/mL) followed by 3 hours of convective delivery of radiolabeled IgG. Transport of antibody into the tumor was measured with quantitative autoradiography along with the tumor interstitial pressure, concentration of collagen and hyaluronic acid, and IgG volume of distribution. RESULTS Antibody was excluded from 42% to 53% of tumor extracellular volume. Exposure of tumors to hyaluronidase did not enhance IgG transport despite removal of 90% of the hyaluronan from the exposed tumor. In contrast, collagenase reduced collagen content, lowered tumor interstitial pressure, and markedly enhanced antibody penetration. CONCLUSIONS Reduction of collagen, but not hyaluronan, in the matrix of ovarian xenografts enhanced the transport of i.p. injected antibody. Although high interstitial pressure is a deterrent to convective transport of macromolecules into the tumor parenchyma, the structure of the interstitial matrix provides an inherent resistance, which must be overcome before effective delivery of an antibody.
Collapse
Affiliation(s)
- Jaehwa Choi
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Thiagarajah JR, Kim JK, Magzoub M, Verkman AS. Slowed diffusion in tumors revealed by microfiberoptic epifluorescence photobleaching. Nat Methods 2006; 3:275-80. [PMID: 16554832 DOI: 10.1038/nmeth863] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 02/09/2006] [Indexed: 11/09/2022]
Abstract
It has not been possible to measure diffusion deep in solid tissues such as tumors because of the limited light penetration of conventional optical techniques. Here we report a microfiberoptic epifluorescence photobleaching (MFEP) method in which photobleaching is done by laser epi-illumination through a multimode fiberoptic whose micron-sized tip can be introduced deep into tissues. We applied MFEP to measure the diffusion of fluorescent macromolecules in tumors in living mice, at depths well beyond those accessible by surface optical measurements. Macromolecule diffusion was slowed about twofold within 200 microm of the surface of a solid tumor, but was slowed greater than tenfold beyond 500 microm. Our results reveal a remarkable and previously unrecognized slowing of diffusion deep in tumors, which correlated with the differing tissue architectures of tumor periphery versus core, and with altered tumor vasculature produced by aquaporin-1 deletion. MFEP should have wide applications for measuring diffusion in organs, solid tumors and other light-inaccessible tissue masses.
Collapse
Affiliation(s)
- Jay R Thiagarajah
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
44
|
Wiig H, Tenstad O, Bert JL. Effect of hydration on interstitial distribution of charged albumin in rat dermis in vitro. J Physiol 2005; 569:631-41. [PMID: 16210353 PMCID: PMC1464246 DOI: 10.1113/jphysiol.2005.096792] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
At physiological pH, negatively charged glycosaminoglycans in the extracellular matrix may influence distribution volume of macromolecular probes, a phenomenon of importance for hydration of the interstitium and therefore for body fluid balance. We hypothesized that such charge effect was dependent on hydration. Human serum albumin (HSA) (the pH value for the isoelectric point (pI) = 4.9) was made neutral by cationization (cHSA) (pI = 7.6). Rat dermis was studied in vitro in a specially designed equilibration cell allowing control of hydration. Using a buffer containing labelled native HSA and cHSA, the distribution volumes were calculated relative to that of 51Cr-EDTA, an extracellular tracer. During changes in hydration (H), defined as (wet weight - dry weight) (dry weight)(-1)), the slope of the equation describing the relationship between extracellular fluid volume (V(x)) (in g H2O (g dry weight)(-1)) and H (V(x) = 0.925 H + 0.105) differed significantly from that for available volumes of cHSA (V(a,cHSA) = 0.624 H - 0.538) and HSA (V(a,HSA) = 0.518 H - 0.518). A gradual reduction in H led to a reduction in difference between available volumes for the two albumin species. Screening the fixed charges by 1 m NaCl resulted in similar available and excluded volumes of native HSA and neutral cHSA. We conclude that during gradual dehydration, there is a reduced effect of fixed negative charges on interstitial exclusion of charged macromolecules. This effect may be explained by a reduced hydration domain surrounding tissue and probe macromolecules in conditions of increased electrostatic interactions. Furthermore, screening of negative charges suggested that hyaluronan associated with collagen may influence intrafibrillar volume of collagen and thereby available and excluded volume fraction.
Collapse
Affiliation(s)
- Helge Wiig
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| | | | | |
Collapse
|