Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nature ; 623(7989): 1070-1078, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968394

ABSTRACT

Three billion years of evolution has produced a tremendous diversity of protein molecules1, but the full potential of proteins is likely to be much greater. Accessing this potential has been challenging for both computation and experiments because the space of possible protein molecules is much larger than the space of those likely to have functions. Here we introduce Chroma, a generative model for proteins and protein complexes that can directly sample novel protein structures and sequences, and that can be conditioned to steer the generative process towards desired properties and functions. To enable this, we introduce a diffusion process that respects the conformational statistics of polymer ensembles, an efficient neural architecture for molecular systems that enables long-range reasoning with sub-quadratic scaling, layers for efficiently synthesizing three-dimensional structures of proteins from predicted inter-residue geometries and a general low-temperature sampling algorithm for diffusion models. Chroma achieves protein design as Bayesian inference under external constraints, which can involve symmetries, substructure, shape, semantics and even natural-language prompts. The experimental characterization of 310 proteins shows that sampling from Chroma results in proteins that are highly expressed, fold and have favourable biophysical properties. The crystal structures of two designed proteins exhibit atomistic agreement with Chroma samples (a backbone root-mean-square deviation of around 1.0 Å). With this unified approach to protein design, we hope to accelerate the programming of protein matter to benefit human health, materials science and synthetic biology.


Subject(s)
Algorithms , Computer Simulation , Protein Conformation , Proteins , Humans , Bayes Theorem , Directed Molecular Evolution , Machine Learning , Models, Molecular , Protein Folding , Proteins/chemistry , Proteins/metabolism , Semantics , Synthetic Biology/methods , Synthetic Biology/trends
2.
Sci Rep ; 13(1): 13408, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591971

ABSTRACT

The intestinal epithelial receptor Guanylyl Cyclase C (GUCY2C) is a tumor-associated cell surface antigen expressed across gastrointestinal malignancies that can serve as an efficacious target for colorectal cancer immunotherapy. Here, we describe a yeast surface-display approach combined with an orthogonal peptide-based mapping strategy to identify the GUCY2C binding epitope of a novel anti-GUCY2CxCD3 bispecific antibody (BsAb) that recently advanced into the clinic for the treatment of cancer. The target epitope was localized to the N-terminal helix H2 of human GUCY2C, which enabled the determination of the crystal structure of the minimal GUCY2C epitope in complex with the anti-GUCY2C antibody domain. To understand if this minimal epitope covers the entire antibody binding region and to investigate the impact of epitope position on the antibody's activity, we further determined the structure of this interaction in the context of the full-length extracellular domain (ECD) of GUCY2C. We found that this epitope is positioned on the protruding membrane-distal helical region of GUCY2C and that its specific location on the surface of GUCY2C dictates the close spatial proximity of the two antigen arms in a diabody arrangement essential to the tumor killing activity of GUCY2CxCD3 BsAb.


Subject(s)
Antibodies, Bispecific , Receptors, Enterotoxin , T-Lymphocytes , Humans , Epitopes , Recognition, Psychology
3.
West J Emerg Med ; 22(6): 1347-1354, 2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34787561

ABSTRACT

INTRODUCTION: Emergency departments (ED) use many medications with a range of therapeutic efficacy and potential significant side effects, and many medications have dosage adjustment recommendations based on the patient's specific genotype. How frequently medications with such pharmaco-genetic recommendations are used in United States (US) EDs has not been studied. METHODS: We conducted a cross-sectional analysis of the 2010-2015 National Hospital Ambulatory Medical Care Survey (NHAMCS). We reported the proportion of ED visits in which at least one medication with Clinical Pharmacogenetics Implementation Consortium (CPIC) recommendation of Level A or B evidence was ordered. Secondary comparisons included distributions and 95% confidence intervals of age, gender, race/ethnicity, ED disposition, geographical region, immediacy, and insurance status between all ED visits and those involving a CPIC medication. RESULTS: From 165,155 entries representing 805,726,000 US ED visits in the 2010-2015 NHAMCS, 148,243,000 ED visits (18.4%) led to orders of CPIC medications. The most common CPIC medication was tramadol (6.3%). Visits involving CPIC medications had higher proportions of patients who were female, had private insurance and self-pay, and were discharged from the ED. They also involved lower proportions of patients with Medicare and Medicaid. CONCLUSION: Almost one fifth of US ED visits involve a medication with a pharmacogenetic recommendation that may impact the efficacy and toxicity for individual patients. While direct application of genotyping is still in development, it is important for emergency care providers to understand and support this technology given its potential to improve individualized, patient-centered care.


Subject(s)
Medicare , Pharmacogenetics , Aged , Cross-Sectional Studies , Emergency Service, Hospital , Female , Health Care Surveys , Humans , Medicaid , United States
4.
Mol Imaging Biol ; 23(6): 941-951, 2021 12.
Article in English | MEDLINE | ID: mdl-34143379

ABSTRACT

PURPOSE: A sensitive and specific imaging biomarker to monitor immune activation and quantify pharmacodynamic responses would be useful for development of immunomodulating anti-cancer agents. PF-07062119 is a T cell engaging bispecific antibody that binds to CD3 and guanylyl cyclase C, a protein that is over-expressed by colorectal cancers. Here, we used 89Zr-Df-IAB22M2C (89Zr-Df-Crefmirlimab), a human CD8-specific minibody to monitor CD8+ T cell infiltration into tumors by positron emission tomography. We investigated the ability of 89Zr-Df-IAB22M2C to track anti-tumor activity induced by PF-07062119 in a human CRC adoptive transfer mouse model (with injected activated/expanded human T cells), as well as the correlation of tumor radiotracer uptake with CD8+ immunohistochemical staining. PROCEDURES: NOD SCID gamma mice bearing human CRC LS1034 tumors were treated with four different doses of PF-07062119, or a non-targeted CD3 BsAb control, and imaged with 89Zr-Df-IAB22M2C PET at days 4 and 9. Following PET/CT imaging, mice were euthanized and dissected for ex vivo distribution analysis of 89Zr-Df-IAB22M2C in tissues on days 4 and 9, with additional data collected on day 6 (supplementary). Data were analyzed and reported as standard uptake value and %ID/g for in vivo imaging and ex vivo tissue distribution. In addition, tumor tissues were evaluated by immunohistochemistry for CD8+ T cells. RESULTS: The results demonstrated substantial mean uptake of 89Zr-Df-IAB22M2C (%ID/g) in PF-07062119-treated tumors, with significant increases in comparison to non-targeted BsAb-treated controls, as well as PF-07062119 dose-dependent responses over time of treatment. A moderate correlation was observed between tumor tissue radioactivity uptake and CD8+ cell density, demonstrating the value of the imaging agent for non-invasive assessment of intra-tumoral CD8+ T cells and the mechanism of action for PF-07062119. CONCLUSION: Immune-imaging technologies for quantitative cellular measures would be a valuable biomarker in immunotherapeutic clinical development. We demonstrated a qualification of 89Zr-IAB22M2C PET to evaluate PD responses (mice) to a novel immunotherapeutic.


Subject(s)
Positron Emission Tomography Computed Tomography , Zirconium , Animals , Biomarkers , Cell Line, Tumor , Mice , Mice, SCID , Positron-Emission Tomography/methods , Receptors, Enterotoxin , T-Lymphocytes
5.
MAbs ; 13(1): 1850395, 2021.
Article in English | MEDLINE | ID: mdl-33459147

ABSTRACT

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Enterotoxin/immunology , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , Female , Humans , Hybridomas , Macaca fascicularis/immunology , Macaca fascicularis/metabolism , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Protein Engineering/methods , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Oncotarget ; 11(15): 1344-1357, 2020 Apr 14.
Article in English | MEDLINE | ID: mdl-32341754

ABSTRACT

P-cadherin-LP-DART is a bispecific antibody targeting P-cadherin expressed on the tumor cells and CD3 on the T-cells. Previously we demonstrated the development and efficacy of P-cadherin-LP-DART in in vitro and in vivo models. Here, we evaluated the three pillars: exposure, targeting specificity and pharmacodynamic modulation for P-cadherin-LP-DART using fluorescence molecular tomography (FMT). Bispecific antibodies and T-cells were conjugated with a near-infrared fluorophores: VivoTag®680XL (VT680) and CellVue®NIR815 (CV815), respectively. In vitro binding and cytotoxic T-lymphocyte assay demonstrated that P-cadherin-LP-DART significantly retained its properties after VT680 conjugation. In vivo FMT imaging was performed to determine the bispecific biodistribution and T-cell trafficking in HCT-116 xenograft model. Peak tumor exposure (2.71%ID) was observed at 96 hr post-injection with measurable quantity even at 240 hr (1.46%ID) (Pillar 1). P-cadherin-LP-DART accumulation in tumor was 20-25 fold higher compared to Control-LP-DART demonstrating the targeting specificity (Pillar 2). Imaging after engraftment of CV815 labeled T-cells showed P-cadherin-LP-DART mediated T-cell trafficking in tumors (Pillar 3). This study harnessed the multichannel capability of FMT and demonstrated the targeting of drug and trafficking of T cells to tumors, simultaneously. Our results show the impact of molecular imaging in demonstrating three pillars of pharmacology, longitudinally and non-invasively.

7.
Pharmacogenomics ; 21(6): 411-418, 2020 04.
Article in English | MEDLINE | ID: mdl-32308127

ABSTRACT

Aim: To investigate the current state of TPMT testing at a single-academic medical center. Methods: Single-center, retrospective chart review for patients newly prescribed a thiopurine. Data collection and evaluation included the prevalence and timing of TPMT testing, correct dosage adjustment if applicable, and incidence of myelosuppression. Results: 121 patients (71%) received TPMT testing. Out of the tested patients, 110 (90.9%) were designated as wild-type with normal metabolism. Dosing modification was appropriate in applicable patients. In unadjusted analysis, there was a lower incidence of myelosuppression among patients who were tested versus those who were not (16.5 vs 36.7%). Conclusion: Based on the study results, TPMT testing opportunities exist for nearly 30% of patients. Testing may reduce the incidence of myelosuppression.


Subject(s)
Delivery of Health Care/methods , Methyltransferases/genetics , Pharmacogenomic Testing/methods , Adult , Aged , Azathioprine/administration & dosage , Azathioprine/metabolism , Delivery of Health Care/trends , Female , Humans , Male , Mercaptopurine/administration & dosage , Mercaptopurine/metabolism , Middle Aged , North Carolina/epidemiology , Pharmacogenetics/methods , Pharmacogenetics/trends , Pharmacogenomic Testing/trends , Retrospective Studies
8.
Clin Cancer Res ; 26(9): 2188-2202, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31996389

ABSTRACT

PURPOSE: Gastrointestinal cancers remain areas of high unmet need despite advances in targeted and immunotherapies. Here, we demonstrate potent, tumor-selective efficacy with PF-07062119, a T-cell engaging CD3 bispecific targeting tumors expressing Guanylyl Cyclase C (GUCY2C), which is expressed widely across colorectal cancer and other gastrointestinal malignancies. In addition, to address immune evasion mechanisms, we explore combinations with immune checkpoint blockade agents and with antiangiogenesis therapy. EXPERIMENTAL DESIGN: PF-07062119 activity was evaluated in vitro in multiple tumor cell lines, and in vivo in established subcutaneous and orthotopic human colorectal cancer xenograft tumors with adoptive transfer of human T cells. Efficacy was also evaluated in mouse syngeneic tumors using human CD3ε transgenic mice. IHC and mass cytometry were performed to demonstrate drug biodistribution, recruitment of activated T cells, and to identify markers of immune evasion. Combination studies were performed with anti-PD-1/PD-L1 and anti-VEGF antibodies. Toxicity and pharmacokinetic studies were done in cynomolgus macaque. RESULTS: We demonstrate that GUCY2C-positive tumors can be targeted with an anti-GUCY2C/anti-CD3ε bispecific, with selective drug biodistribution to tumors. PF-07062119 showed potent T-cell-mediated in vitro activity and in vivo efficacy in multiple colorectal cancer human xenograft tumor models, including KRAS- and BRAF-mutant tumors, as well as in the immunocompetent mouse syngeneic tumor model. PF-07062119 activity was further enhanced when combined with anti-PD-1/PD-L1 treatment or in combination with antiangiogenic therapy. Toxicity studies in cynomolgus indicated a monitorable and manageable toxicity profile. CONCLUSIONS: These data highlight the potential for PF-07062119 to demonstrate efficacy and improve patient outcomes in colorectal cancer and other gastrointestinal malignancies.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/immunology , Colorectal Neoplasms/therapy , Gastrointestinal Neoplasms/therapy , Immunotherapy/methods , Receptors, Enterotoxin/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Antibodies, Bispecific/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Tissue Distribution
9.
J Pharm Technol ; 36(3): 110-113, 2020 Jun.
Article in English | MEDLINE | ID: mdl-37927308

ABSTRACT

Objective: To report the utilization of emicizumab in a patient with severe hemophilia A with inducible inhibitors and the reduction of drug costs related to decreased on-demand recombinant factor VIIa use. Case Summary: A 65-year-old African American man with established hemophilia A with an inducible factor VIII inhibitor presented with a bleeding hematoma from the right posterior thigh. The patient was historically managed on frequent administration of recombinant factor VIIa to achieve hemostasis and was started on every 2-hour dosing during this admission. Emicizumab, a new therapy for hemophilia A, became available during this admission, and the patient discontinued recombinant factor VIIa and transitioned to weekly emicizumab injections. The patient did not require any recombinant factor VIIa during the following 12 months resulting in substantial drug cost savings. Discussion: After initiation of emicizumab therapy, the patient no longer required on-demand treatment with recombinant factor VIIa for bleeds. Through this reduction in recombinant factor VIIa, there was a large decrease in inpatient drug costs and inpatient admissions for bleeding events. Conclusion: The potential reduction in drug costs and inpatient admissions should be considered when determining if emicizumab therapy is appropriate for hemophilia A patients with inhibitors. Further research is needed to confirm that continued long-term use of emicizumab remains associated with a reduction in on-demand treatment.

10.
Hosp Pharm ; 54(5): 309-313, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31555006

ABSTRACT

Purpose: The purpose of this quality-improvement project was to assess risk evaluation and mitigation strategies (REMS) program compliance for pulmonary arterial hypertension (PAH) drugs following the initiation of more rigid protocols and informatics changes. The primary objective of the study was to determine the effects of these changes on overall compliance of the REMS program requirements. Method: This was a single-center, retrospective evaluation of protocols and informatics updates that were developed to increase compliance with REMS programs for four drugs used to treat PAH. Two separate time periods were examined for comparison: the preinformatics period, January 2015 to February 2016, and the postinformatics period, October 2016 to April 2017. To be included in the study, patients must have been at least 18 years of age and have been ordered one of the following agents: riociguat, macitentan, bosentan, or ambrisentan. Results: Overall, 94 patients were evaluated with 50 in the preinformatics group and 44 in the postinformatics group. The overall mean age of included patients was 55 years, 57.9% of patients were white, 69.1% were female, and 43.6% were prescribed ambrisentan during the study period. The primary composite endpoint of adherence to REMS protocol (pregnancy tests performed within 30 days of medication initiation for female patients of childbearing potential, liver function tests [LFTs] ordered within 30 days of bosentan initiation, and initiation of therapy order documented by an attending provider enrolled in the REMS program) showed an overall improvement in the postinformatics period, 95% vs 71% (P = .07).There was a statistically significant increase in pregnancy tests performed within 30 days of medication order in the postinformatics period (36.4% vs 100%; P = .01). Furthermore, during the postinformatics period, the number of documented interventions (iVents) performed by a pharmacist was 90.9%. Conclusion: Initiation of more rigid ordering protocols for the endothelin receptor antagonists (macitentan, bosentan, or ambrisentan) and riociguat improved pharmacist and physician compliance with REMS requirements.

11.
AAPS J ; 21(4): 73, 2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31165304

ABSTRACT

Typesetting error occurred and Figure 1a and Figure 1b were altered during the uploading process. The original article has been corrected.

12.
AAPS J ; 21(4): 66, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31119428

ABSTRACT

CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , CD3 Complex/immunology , Cadherins/metabolism , Models, Biological , Animals , Antibodies, Bispecific/blood , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , HCT116 Cells , Humans , Immunotherapy , Lymphocyte Activation , Macaca fascicularis , Mice, SCID , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Translational Research, Biomedical , Xenograft Model Antitumor Assays
13.
Vox Sang ; 114(4): 374-385, 2019 May.
Article in English | MEDLINE | ID: mdl-30937927

ABSTRACT

BACKGROUND/OBJECTIVES: Prothrombin complex concentrates (PCC) are increasingly administered off-label in the United States to treat bleeding in cardiovascular surgical patients and carry the potential risk for acquired thromboembolic side-effects after surgery. Therefore, we hypothesized that the use of low-dose 3-factor (3F) PCC (20-30 IU/kg), as part of a transfusion algorithm, reduces bleeding without increasing postoperative thrombotic/thromboembolic complications. MATERIALS/METHODS: After IRB approval, we retrospectively analysed 114 consecutive, complex cardiovascular surgical patients (age > 18 years), between February 2014 and June 2015, that received low-dose 3F-PCC (Profilnine® ), of which seven patients met established exclusion criteria. PCC was dosed according to an institutional perioperative algorithm. Allogeneic transfusions were recorded before and after PCC administration (n = 107). The incidence of postoperative thromboembolic events was determined within 30 days of surgery, and Factor II levels were measured in a subset of patients (n = 20) as a quality control measure to avoid excessive PCC dosing. RESULTS: Total allogeneic blood product transfusion reached a mean of 12·4 ± 9·9 units before PCC and 5·0 ± 6·3 units after PCC administration (P < 0·001). The mean PCC dose was 15·8 ± 7·1 IU/kg. Four patients (3·8%) each experienced an ischaemic stroke on postoperative day 1, 2, 4 and 27. Seven patients (6·5%) had acquired venous thromboembolic disease within 10 days of surgery. Median factor II level after transfusion algorithm adherence and PCC administration was 87%. CONCLUSIONS: 3F-PCC use for refractory bleeding after cardiovascular surgery resulted in reduced transfusion of allogeneic blood and blood products. Adherence to this algorithmic approach was associated with an acceptable incidence of postoperative thrombotic/thromboembolic complications.


Subject(s)
Blood Coagulation Factors/chemistry , Blood Coagulation , Hemorrhage/therapy , Hemostasis , Thromboembolism/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Algorithms , Blood Coagulation Tests , Blood Platelets/cytology , Blood Transfusion , Cardiopulmonary Bypass , Female , Fibrinogen/chemistry , Humans , Male , Middle Aged , Postoperative Period , Retrospective Studies , United States , Young Adult
14.
Transfusion ; 59(5): 1870-1879, 2019 05.
Article in English | MEDLINE | ID: mdl-30762882

ABSTRACT

Therapeutic plasma exchange (TPE) removes coagulation proteins, but its impact on therapeutic anticoagulation is unknown. We performed a systematic review of the literature to determine the coagulation effects of TPE in patients receiving systemic anticoagulation. We searched MEDLINE, CINAHL, EMBASE, and Web of Science until June 2018 for studies combining controlled vocabulary and keywords related to therapeutic plasma exchange, plasmapheresis, anticoagulants, and therapy. The primary outcome was the effect of TPE on anti-Xa activity, activated partial thromboplastin time (aPTT), or international normalized ratio (INR). The secondary outcome was reports of post-TPE bleeding or thrombosis. A total of 1830 references were screened and eight studies identified. Our selected studies (five case reports and three case series) involved 23 patients and evaluated the effects of seven anticoagulants. Six studies of unfractionated heparin, low-molecular-weight heparins, and direct oral anticoagulants demonstrated an anti-Xa level decline. Two studies of unfractionated heparin and low-molecular-weight heparins showed an aPTT increase. One study of warfarin showed a post-TPE INR increase. Reports of post-TPE bleeding occurred in two patients and thrombosis in one. In patients receiving therapeutic anticoagulation, TPE is associated with anti-Xa activity decline and aPTT and INR increase. These coagulation changes do not appear to significantly increase bleeding or thrombotic risk. Our data suggest the need for prospective studies to investigate the true clinical impact of TPE on therapeutic anticoagulation.


Subject(s)
Plasma Exchange/methods , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Female , Humans , Male , Partial Thromboplastin Time
15.
Cancer Immunol Immunother ; 67(2): 247-259, 2018 02.
Article in English | MEDLINE | ID: mdl-29067496

ABSTRACT

Strong evidence exists supporting the important role T cells play in the immune response against tumors. Still, the ability to initiate tumor-specific immune responses remains a challenge. Recent clinical trials suggest that bispecific antibody-mediated retargeted T cells are a promising therapeutic approach to eliminate hematopoietic tumors. However, this approach has not been validated in solid tumors. PF-06671008 is a dual-affinity retargeting (DART®)-bispecific protein engineered with enhanced pharmacokinetic properties to extend in vivo half-life, and designed to engage and activate endogenous polyclonal T cell populations via the CD3 complex in the presence of solid tumors expressing P-cadherin. This bispecific molecule elicited potent P-cadherin expression-dependent cytotoxic T cell activity across a range of tumor indications in vitro, and in vivo in tumor-bearing mice. Regression of established tumors in vivo was observed in both cell line and patient-derived xenograft models engrafted with circulating human T lymphocytes. Measurement of in vivo pharmacodynamic markers demonstrates PF-06671008-mediated T cell activation, infiltration and killing as the mechanism of tumor inhibition.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Cadherins/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , CD3 Complex/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Female , HCT116 Cells , HT29 Cells , Humans , Mice , Xenograft Model Antitumor Assays
16.
Antibodies (Basel) ; 5(1)2016 Mar 04.
Article in English | MEDLINE | ID: mdl-31557987

ABSTRACT

Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (-4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.

17.
FEBS J ; 276(17): 4909-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19664054

ABSTRACT

LOX-1 is a scavenger receptor that functions as the primary receptor for oxidized low-density lipoprotein (OxLDL) in endothelial cells. The binding of OxLDL to LOX-1 is believed to lead to endothelial activation, dysfunction, and injury, which constitute early atherogenic events. Because of its potential pathological role in atherosclerosis, LOX-1 has been proposed as a therapeutic target for the treatment of this disease. In order to antagonize the ligand-binding function of cell surface LOX-1, we generated a series of recombinant human LOX-1-crystallizable fragment (Fc) fusion proteins and subsequently characterized their biochemical properties and ligand-binding activities in vitro. Consistent with the notion that oligomerization of cell surface LOX-1 is required for high-avidity binding of ligands, we found that LOX-1-Fc fusion protein containing four ligand-binding domains per Fc dimer, but not the one containing two ligand-binding domains, exhibited ligand-binding activity. Optimal ligand-binding activity could be achieved via crosslinking of LOX-1-Fc fusion proteins with a polyclonal antibody against Fc. The crosslinked LOX-1-Fc protein also effectively inhibited the binding and internalization of OxLDL by cell surface LOX-1. These findings demonstrate that functional oligomerization is required for recombinant LOX-1-Fc to function as an effective antagonist.


Subject(s)
Cell Membrane/metabolism , Lipoproteins, LDL/metabolism , Models, Molecular , Recombinant Fusion Proteins/pharmacology , Scavenger Receptors, Class E/metabolism , Amino Acid Sequence , Animals , Antibodies/chemistry , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Cross-Linking Reagents/chemistry , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics
18.
Arthritis Rheum ; 60(3): 840-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19248108

ABSTRACT

OBJECTIVE: Lubricin, also referred to as superficial zone protein and PRG4, is a synovial glycoprotein that supplies a friction-resistant, antiadhesive coating to the surfaces of articular cartilage, thereby protecting against arthritis-associated tissue wear and degradation. This study was undertaken to generate and characterize a novel recombinant lubricin protein construct, LUB:1, and to evaluate its therapeutic efficacy following intraarticular delivery in a rat model of osteoarthritis (OA). METHODS: Binding and localization of LUB:1 to cartilage surfaces was assessed by immunohistochemistry. The cartilage-lubricating properties of LUB:1 were determined using a custom friction testing apparatus. A cell-binding assay was performed to quantify the ability of LUB:1 to prevent cell adhesion. Efficacy studies were conducted in a rat meniscal tear model of OA. One week after the surgical induction of OA, LUB:1 or phosphate buffered saline vehicle was administered by intraarticular injection for 4 weeks, with dosing intervals of either once per week or 3 times per week. OA pathology scores were determined by histologic analysis. RESULTS: LUB:1 was shown to bind effectively to cartilage surfaces, and facilitated both cartilage boundary lubrication and inhibition of synovial cell adhesion. Treatment of rat knee joints with LUB:1 resulted in significant disease-modifying, chondroprotective effects during the progression of OA, by markedly reducing cartilage degeneration and structural damage. CONCLUSION: Our findings demonstrate the potential use of recombinant lubricin molecules in novel biotherapeutic approaches to the treatment of OA and associated cartilage abnormalities.


Subject(s)
Cartilage, Articular/pathology , Glycoproteins/therapeutic use , Osteoarthritis/pathology , Osteoarthritis/prevention & control , Recombinant Proteins/therapeutic use , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/injuries , Cell Adhesion/drug effects , Disease Models, Animal , Disease Progression , Glycoproteins/administration & dosage , Glycoproteins/pharmacology , Injections, Intra-Articular , Male , Random Allocation , Rats , Rats, Inbred Lew , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/pathology , Treatment Outcome
19.
Endocrinology ; 146(8): 3351-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15878963

ABSTRACT

A full-length transcript encoding a functional type II GnRH receptor was cloned from the pituitary of the sea lamprey, Petromyzon marinus. The current study is the first to identify a pituitary GnRH receptor transcript in an agnathan, which is the oldest vertebrate lineage. The cloned receptor retains the conserved structural features and amino acid motifs of other known GnRH receptors and notably includes a C-terminal intracellular tail of approximately 120 amino acids, the longest C-terminal tail of any vertebrate GnRH receptor identified to date. The lamprey GnRH receptor was shown to activate the inositol phosphate (IP) signaling system; stimulation with either lamprey GnRH-I or lamprey GnRH-III led to dose-dependent responses in transiently transfected COS7 cells. Furthermore, analyses of serially truncated lamprey GnRH receptor mutants indicate perturbations of the C-terminal tail disrupts IP accumulation, however, the tailless lamprey GnRH receptor was not only functional but was also capable of stimulating IP levels equal to wild type. Expression of the receptor transcript was demonstrated in the pituitary and testes using RT-PCR, whereas in situ hybridization showed expression and localization of the transcript in the proximal pars distalis of the pituitary. The phylogenetic placement and structural and functional features of this GnRH receptor suggest that it is representative of an ancestral GnRH receptor. In addition to having an important role in lamprey reproductive processes, the extensive C-terminal tail of this lamprey GnRH receptor may have great significance for understanding the evolutionary change of this vital structural feature within the GnRH receptor family.


Subject(s)
Lampreys/genetics , Receptors, LHRH/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Consensus Sequence , DNA Primers , Humans , Lampreys/classification , Molecular Sequence Data , Phylogeny , Receptors, LHRH/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Vertebrates
20.
Gen Comp Endocrinol ; 141(1): 76-83, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15707605

ABSTRACT

Gonadotropin releasing hormone (GnRH) is the key hypothalamic neurohormone that is critical in its role of reproduction in all vertebrates. There are currently twenty-four known forms of GnRH that have been identified, 14 in vertebrates and 10 in invertebrates. In tunicates, the primary structure of nine forms have been identified, all of which have been shown to stimulate gamete release. However, the distribution and function of the various GnRH peptides in tunicates have not been fully examined. Therefore, the objective of this study was to determine tissue specific expression of Ci-gnrh-1 and Ci-gnrh-2 in an adult tunicate, Ciona intestinalis, using reverse transcriptase-polymerase chain reaction (RT-PCR) and in situ hybridization. To examine the expression of the two GnRH genes, total RNA and genomic DNA were isolated from whole animals. Total RNA from neural tissue (cerebral ganglion and neural gland), testis, ovary, heart, and hepatic organ were also isolated. Results from RT-PCR indicated both forms are only expressed in the neural tissue. We extended these studies using fluorescent dual label in situ hybridization. GnRH expression was confirmed to be in the cerebral ganglion bordering the neural gland. These current data along with previous studies suggest that GnRH may be involved in reproduction in the protochordate.


Subject(s)
Gene Expression Profiling , Gonadotropin-Releasing Hormone/analysis , Gonadotropin-Releasing Hormone/genetics , Urochordata/genetics , Urochordata/physiology , Animals , Brain/physiology , In Situ Hybridization , Reproduction/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...