Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Immunother ; 47(5): 160-171, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38562119

ABSTRACT

SUMMARY: T-cell-directed cancer therapies such as T-cell-engaging bispecifics (TCBs) are commonly associated with cytokine release syndrome and associated clinical signs that can limit their tolerability and therapeutic benefit. Strategies for reducing cytokine release are therefore needed. Here, we report on studies performed in cynomolgus monkeys to test different approaches for mitigating cytokine release with TCBs. A "priming dose" as well as subcutaneous dosing reduced cytokine release compared with intravenous dosing but did not affect the intended T-cell response to the bispecific. As another strategy, cytokines or cytokine responses were blocked with an anti-IL-6 antibody, dexamethasone, or a JAK1/TYK2-selective inhibitor, and the effects on toxicity as well as T-cell responses to a TCB were evaluated. The JAK1/TYK2 inhibitor and dexamethasone prevented CRS-associated clinical signs on the day of TCB administration, but the anti-IL-6 had little effect. All interventions allowed for functional T-cell responses and expected damage to target-bearing tissues, but the JAK1/TYK2 inhibitor prevented the upregulation of activation markers on T cells, suggesting the potential for suppression of T-cell responses. Our results suggest that short-term prophylactic dexamethasone treatment may be an effective option for blocking cytokine responses without affecting desired T-cell responses to TCBs.


Subject(s)
Antibodies, Bispecific , Cytokines , Macaca fascicularis , T-Lymphocytes , Animals , Antibodies, Bispecific/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cytokines/metabolism , Dexamethasone/pharmacology , Humans , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Interleukin-6/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Neoplasms/immunology , Neoplasms/drug therapy
2.
Comp Med ; 73(5): 398-406, 2023 10 29.
Article in English | MEDLINE | ID: mdl-38087406

ABSTRACT

Cold agglutinin disease (CAD) is a condition involving anemia and its related symptoms; it is caused by autoantibodies that bind and agglutinate red blood cells in areas susceptible to hypothermia, such as extremities exposed to cold temperatures. CAD is rare, with 5 to 20 human cases per million individuals. In this report, we describe a case of CAD in a previously healthy and experimentally naïve adult Indian rhesus macaque that was housed indoors and presented with blood in the urine. After our observations of hemoglobinuria and anemia led us to suspect CAD, we demonstrated that the macaque's blood agglutinated at reduced temperatures. We also noticed that the provision of cold foraging treats triggered episodes of hemoglobinuria. Further investigation revealed that serum from the macaque agglutinated RBCs in vitro with high thermal amplitude (at or below 30 °C) and had an antibody titer of 8 to 32. The serum contained autoantibodies of the immunoglobulin M (IgM) isotype; agglutinins of the IgG isotype were not detected. The cold-dependent IgM autoantibodies in the serum from the affected macaque reacted against a common RBC antigen because RBCs collected from other macaques were bound and agglutinated by the affected animal's IgM under cold conditions. This in vitro binding activity was reversible when the test temperature was returned to normal body temperature (37 °C). These findings demonstrated cold-dependent RBC-specific IgM agglutinins and led us to a diagnosis of CAD. This is the first documented case of spontaneous CAD in a rhesus macaque.


Subject(s)
Anemia, Hemolytic, Autoimmune , Animals , Agglutinins , Anemia, Hemolytic, Autoimmune/veterinary , Autoantibodies , Cold Temperature , Hemoglobinuria , Immunoglobulin M , Macaca mulatta
3.
J Immunotoxicol ; 20(1): 2176952, 2023 12.
Article in English | MEDLINE | ID: mdl-36788724

ABSTRACT

Cytotoxic T-lymphocytes (CTL) are a subset of T-cells that play a critical role in protecting against intracellular infections and cancer, and have the ability to identify and kill infected or transformed cells expressing non-self peptides associated with major histocompatibility (MHC) Class I molecules. Conversely, aberrant CTL activity can contribute to immune-related pathology under conditions of overwhelming infection or autoimmunity. Disease-modifying therapeutics can have unintended effects on CTL, and a growing number of therapeutics are intended to either suppress or enhance CTL or their functions. The susceptibility of CTL to unintended effects from common therapeutic modalities underscores the need for a better understanding of the impact that such therapies have on CTL function and the associated safety implications. While there are reliable ways of quantifying CTL, notably via flow cytometric analysis of specific CTL markers, it has been a greater challenge to implement fit-for-purpose methods measuring CTL function in the context of safety studies of therapeutics. This review focuses on methods for measuring CTL responses in the context of drug safety and pharmacology testing, with the goals of informing the reader about current approaches, evaluating their pros and cons, and providing perspectives on the utility of these approaches for safety evaluation.


Subject(s)
Neoplasms , T-Lymphocytes, Cytotoxic , Animals , Primates , Neoplasms/therapy , Cytotoxicity, Immunologic
4.
Front Immunol ; 13: 845417, 2022.
Article in English | MEDLINE | ID: mdl-35493516

ABSTRACT

P-cadherin is a cell-cell adhesion molecule that is overexpressed in several solid tumors. PF-06671008 is a T-cell-redirecting bispecific antibody that engages both P-cadherin on tumors and CD3ϵ on T cells and induces antitumor activity in preclinical models. We conducted a phase 1, open-label, first-in-human, dose-escalation study to characterize the safety and tolerability of PF-06671008, towards determining the recommended phase 2 dose. Adult patients with treatment-refractory solid tumors received PF-06671008 (1.5-400 ng/kg) as a weekly intravenous (IV) infusion on a 21-day/3-week cycle. Parallel cohorts evaluated dosing via subcutaneous injection (SC) or an IV-prime dose. Of the 27 patients enrolled in the study, 24 received PF-06671008 IV in escalating doses, two received SC, and one IV-prime. A dose-limiting toxicity of cytokine release syndrome (CRS) occurred in the 400-ng/kg IV group, prompting evaluation of SC and IV-prime schedules. In all, 25/27 patients who received PF-06671008 reported at least one treatment-related adverse event (TRAE); the most common were CRS (21/27), decreased lymphocyte count (9/27), and hypophosphatemia (8/27). Seven patients permanently discontinued treatment due to adverse events and no treatment-related deaths occurred. Cytokine peak concentrations and CRS grade appeared to positively correlate with Cmax. Although the study was terminated due to limited antitumor activity, it provides important insights into understanding and managing immune-related adverse events resulting from this class of molecules. Clinical Trial Registration: URL: https://clinicaltrials.gov/ct2/show/NCT02659631, ClinicalTrials.gov Identifier: NCT02659631.


Subject(s)
Antibodies, Bispecific , Neoplasms , Adult , Antibodies, Bispecific/adverse effects , Cadherins , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Treatment Outcome
5.
Regul Toxicol Pharmacol ; 119: 104825, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33220389

ABSTRACT

With the growth of monoclonal antibodies and other proteins as major modalities in the pharmaceutical industry, there has been an increase in pharmacology and toxicity testing of biotherapeutics in animals. Animals frequently mount an immune response to human therapeutic proteins. This can result in asymptomatic anti-drug antibody formation, immune complexes that affect drug disposition and/or organ function such as kidney, cytokine release responses, fatal hypersensitivity, or a range of reactions in between. In addition, an increasing number of oncology therapeutics are being developed that enhance or directly stimulate immune responses by a variety of mechanisms, which could increase the risk of autoreactivity and an autoimmune-like syndrome in animals and humans. When evaluating the risk of biotherapeutics prior to entering the clinic, the nonclinical safety data may include any of these responses and it is critical to understand whether they represent a safety liability for humans. The DruSafe Leadership group of the IQ Consortium conducted a survey of industry to understand sponsors' experiences with these immune reactions in nonclinical studies related to both immunogenicity and pharmacologically-mediated immune perturbations. The survey covered what pathways were affected, how the immune responses were presented, how the company and health authorities interpreted the data and whether the immune responses were observed in the clinic. Additionally, the survey gathered information on association of these findings with anti-drug antibodies as well as sponsor's use of immunogenicity predictive tools. The data suggests that the ability of a biotherapeutic to activate the immune system, intended or not, plays a significant role on characteristics of the response and whether theys are translatable.


Subject(s)
Biological Products/toxicity , Immune System/drug effects , Animals , Antibodies/immunology , Biological Products/immunology , Drug Evaluation, Preclinical , Drug Industry , Drug-Related Side Effects and Adverse Reactions , Macaca fascicularis , Mice , Rats , Surveys and Questionnaires , Toxicity Tests
6.
Mol Cancer Ther ; 19(10): 2105-2116, 2020 10.
Article in English | MEDLINE | ID: mdl-32847983

ABSTRACT

Development of antagonistic mAbs that specifically target the immune checkpoint receptor, programmed cell death protein-1 (PD-1), is of great interest for cancer immunotherapy. Here, we report the biophysical characteristics and nonclinical antagonistic activities of sasanlimab (PF-06801591), a humanized anti-PD-1 antibody of IgG4 isotype. We show that sasanlimab binds selectively and with similar high potency to human and cynomolgus monkey PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, with no detectable Fc-dependent effector function. The binding of sasanlimab to human and cynomolgus PD-1 is associated with the formation of a stable complex, which is likely to be the main driver of this high-affinity interaction. In vitro, sasanlimab significantly augmented T-cell proliferation and cytokine production in mixed lymphocyte reaction and superantigen stimulation assays. In vivo, sasanlimab accelerated the incidence of GvHD by enhancing T-cell proliferation and cytokine secretion in a xenogeneic model of acute GvHD and halted the growth of MC-38 colon adenocarcinoma tumors in human PD-1 knock-in mice. Pharmacokinetic and toxicokinetic findings from cynomolgus monkey showed that sasanlimab was active and well-tolerated. Taken together, the data presented here support the clinical development of sasanlimab for the treatment of patients with advanced cancers as a single agent or in combination with other immunotherapies.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Humans , Immune Checkpoint Inhibitors/pharmacology , Mice
7.
J Immunotoxicol ; 17(1): 67-85, 2020 12.
Article in English | MEDLINE | ID: mdl-32100588

ABSTRACT

Currently, there is a multitude of CD3 bispecifics with different molecular designs and binding properties in preclinical and clinical development for the treatment of liquid or solid tumors. The key safety concerns with CD3 bispecifics are excessive release of cytokines, which may translate to potentially life-threating cytokine release syndrome (CRS), target organ toxicity due to redirection of T-cells to normal tissues expressing the tumor-associated antigen (TAA) (off-tumor/on-target cytotoxicity), and, in some instances, neurotoxicity. Another key challenge is to arrive at a safe clinical starting dose and an efficient escalating strategy that allows patients in early dose cohorts the potential for clinical benefit in Phase 1 trials. To expand the therapeutic index and bring more treatment options to patients, there are intense efforts to overcome these challenges through improvements in molecular design, preclinical safety assessment strategies, and clinical management practices. A recent workshop at the U.S. Food and Drug Administration (FDA) with industry, academic, and regulatory agency representation was held to discuss the challenges and explore where such improvements to the development of CD3 bispecifics can be implemented. Here, the content of the presentations and the discussion that occurred during this workshop are summarized.


Subject(s)
Antibodies, Bispecific/toxicity , Antigens, Neoplasm/metabolism , Antineoplastic Agents/toxicity , CD3 Complex/antagonists & inhibitors , Cytokine Release Syndrome/prevention & control , Animals , Antibodies, Bispecific/administration & dosage , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , CD3 Complex/immunology , CD3 Complex/metabolism , Consensus , Consensus Development Conferences as Topic , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/immunology , Cytokines/metabolism , Drug Screening Assays, Antitumor/standards , Europe , Humans , Japan , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Translational Research, Biomedical/standards , United States , United States Food and Drug Administration
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.
Mol Cancer Ther ; 18(11): 2008-2020, 2019 11.
Article in English | MEDLINE | ID: mdl-31434693

ABSTRACT

The restricted expression pattern of B-cell maturation antigen (BCMA) makes it an ideal tumor-associated antigen (TAA) for the treatment of myeloma. BCMA has been targeted by both CD3 bispecific antibody and antibody-drug conjugate (ADC) modalities, but a true comparison of modalities has yet to be performed. Here we utilized a single BCMA antibody to develop and characterize both a CD3 bispecific and 2 ADC formats (cleavable and noncleavable) and compared activity both in vitro and in vivo with the aim of generating an optimal therapeutic. Antibody affinity, but not epitope was influential in drug activity and hence a high-affinity BCMA antibody was selected. Both the bispecific and ADCs were potent in vitro and in vivo, causing dose-dependent cell killing of myeloma cell lines and tumor regression in orthotopic myeloma xenograft models. Primary patient cells were effectively lysed by both CD3 bispecific and ADCs, with the bispecific demonstrating improved potency, maximal cell killing, and consistency across patients. Safety was evaluated in cynomolgus monkey toxicity studies and both modalities were active based on on-target elimination of B lineage cells. Distinct nonclinical toxicity profiles were seen for the bispecific and ADC modalities. When taken together, results from this comparison of BCMA CD3 bispecific and ADC modalities suggest better efficacy and an improved toxicity profile might be achieved with the bispecific modality. This led to the advancement of a bispecific candidate into phase I clinical trials.


Subject(s)
Antibodies, Bispecific/administration & dosage , B-Cell Maturation Antigen/metabolism , CD3 Complex/immunology , Immunoconjugates/administration & dosage , Multiple Myeloma/drug therapy , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacology , Antibody Affinity , B-Cell Maturation Antigen/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacology , Mice , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
J Immunotoxicol ; 16(1): 164-172, 2019 12.
Article in English | MEDLINE | ID: mdl-31464151

ABSTRACT

The current era of drug discovery has been marked by a significant increase in the development of immune modulating agents to address a range of diseases such as cancer, chronic inflammation, and other conditions of dysregulated immunity. Non-clinical evaluation of these agents in animal models can be challenging, as the presence of an active immune state is often required in order to detect the effects of the test agent. Modulation of interleukin (IL)-10 signaling represents this type of situation in that altering IL-10 action in vivo can be difficult to appreciate in the absence of an ongoing immune response. The study presented here reports on the use of lipopolysaccharide (LPS) challenge in cynomolgus macaques to induce predictable inflammatory cytokine responses. The results showed that IL-10 receptor (IL-10R) blockade with an antagonist monoclonal antibody (mAb) dramatically enhanced the LPS-induced cytokine response, thus demonstrating in vivo pharmacologic activity of this immunomodulatory antibody. We submit that this approach could be applied to other cases where the intent of a candidate therapeutic is to modulate components of inflammatory cytokine responses.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Interleukin-10 Receptor alpha Subunit/antagonists & inhibitors , Lipopolysaccharides/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Drug Evaluation, Preclinical/methods , Immunologic Factors/therapeutic use , Injections, Intravenous , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-10 Receptor alpha Subunit/immunology , Interleukin-10 Receptor alpha Subunit/metabolism , Lipopolysaccharides/administration & dosage , Macaca fascicularis , Male
11.
Clin Transl Sci ; 12(6): 600-608, 2019 11.
Article in English | MEDLINE | ID: mdl-31268236

ABSTRACT

T-cell-engaging bispecific antibodies (T-BsAbs) are an important class of antibody therapeutics in immuno-oncology. T-BsAbs simultaneously bind to CD3 on T cells and a tumor-associated antigen on tumor cells, activate T cells, and redirect T cells' cytotoxicity against tumor cells. Cytokine release syndrome (CRS), a common dose-limiting adverse event for T-BsAbs, is associated with T-cell activation. A "priming" dose strategy (i.e., a lower initial dose followed by a higher maintenance dose) has been implemented in the clinic to mitigate CRS and to achieve efficacious doses with T-BsAbs. So far, the selection of the optimal priming dosing regimen is largely empirical. A "fit-for-purpose" semimechanistic pharmacokinetic/pharmacodynamic model was developed to characterize the cytokine release profiles upon T-BsAb treatment, including the priming effect observed with repeated dosing. This model can be utilized to simulate cytokine profiles following various dosing regimens and may assist the design of clinical dosing strategies for T-BsAbs programs.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Cytokines/metabolism , Models, Biological , T-Lymphocytes/drug effects , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Cytokines/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Lymphocyte Activation/drug effects , Macaca fascicularis , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
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.

13.
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
14.
Virology ; 488: 28-36, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26590795

ABSTRACT

Lymphocryptoviruses such as Epstein-Barr virus (EBV) cause persistent infections in human and non-human primates, and suppression of the immune system can increase the risk of lymphocryptovirus (LCV)-associated tumor development in both human and non-human primates. To enable LCV infection as a non-clinical model to study effects of therapeutics on EBV immunity, we determined the genomic DNA sequence of the LCV from cynomolgus macaque, a species commonly used for non-clinical testing. Comparison to rhesus macaque LCV and human EBV sequences indicates that LCV from the cynomolgus macaque has the same genomic arrangement and a high degree of similarity in most genes, especially with rhesus macaque LCV. Genes showing lower similarity were those encoding proteins involved in latency and/or tumor promotion or immune evasion. The genomic sequence of LCV from cynomolgus macaque should aid the development of non-clinical tools for identifying therapeutics that impact LCV immunity and carry potential lymphoma risk.


Subject(s)
DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Lymphocryptovirus/genetics , Lymphocryptovirus/isolation & purification , Macaca fascicularis/virology , Animals , Gene Order , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Synteny
15.
Reprod Toxicol ; 53: 105-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25461901

ABSTRACT

Two intravenous studies with tanezumab, an anti-nerve growth factor monoclonal antibody, were conducted in pregnant cynomolgus monkeys to assess potential effects on pregnancy and pre- and postnatal development. Study 1 evaluated infants up to 12 months of age following weekly maternal dosing (0, 0.5, 4 or 30 mg/kg; 18 per group) from gestation day (GD) 20 through parturition. Study 2 evaluated infants 2 months postnatally following weekly maternal dosing (0, 0.5 or 30 mg/kg; 20-21 per group) from GD 20 through 48. In the absence of maternal toxicity, tanezumab increased stillbirth and post-birth infant mortality/morbidity, decreased infant growth and resulted in microscopic changes in the peripheral sympathetic and sensory nervous system of the infants at all doses. Decreased primary antibody responses and increased incidences in skin changes in infants were also observed. The no-observed-adverse-effect-level for maternal toxicity was 30 mg/kg and <0.5 mg/kg for developmental toxicity.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Animals , Birth Weight/drug effects , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Macaca fascicularis , Male , Maternal-Fetal Exchange , No-Observed-Adverse-Effect Level , Pregnancy , Prenatal Exposure Delayed Effects , Receptor, Nerve Growth Factor/antagonists & inhibitors , Skin/drug effects , Skin/pathology , Stillbirth
16.
J Virol Methods ; 207: 220-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25064358

ABSTRACT

Lymphocryptoviruses such as Epstein-Barr virus (EBV) are important pathogens in both human and non-human primates, particularly during immunosuppression. Immunomodulatory molecules that may suppress antiviral immunity are commonly tested in the cynomolgus macaque. To enable the study of lymphocryptovirus (LCV) in this non-clinical model, PCR-based assays were developed to measure LCV viral load, as well as transcripts for the lytic phase LCV gene, BALF-2. Results from studies employing these assays showed that LCV genome was detected in the oropharyngeal epithelium of all cynomolgus monkeys tested, and the majority had viral genome in peripheral blood mononuclear cells (PBMCs). The results also revealed LCV lytic phase gene expression not only in the oropharynx of most monkeys, but also in PBMCs of approximately one half of monkeys tested. This unexpected finding suggests that initiation of the lytic gene expression cascade occurs often in the peripheral blood cells of healthy monkeys.


Subject(s)
Gene Expression , Herpesviridae Infections/veterinary , Leukocytes, Mononuclear/virology , Lymphocryptovirus/isolation & purification , Primate Diseases/virology , Animals , Epithelium/virology , Genes, Viral , Herpesviridae Infections/virology , Lymphocryptovirus/genetics , Macaca fascicularis , Oropharynx/virology , Prevalence , Real-Time Polymerase Chain Reaction
17.
J Immunotoxicol ; 11(1): 35-43, 2014.
Article in English | MEDLINE | ID: mdl-23461640

ABSTRACT

A number of immunomodulatory therapeutics increase the risk of disease associated with latent herpesviruses such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV), a member of the lymphocryptovirus (LCV) family that infects humans. The diseases associated with loss of immunity to these viruses can have major impacts on patients as well as on the commercial viability of the immunomodulatory therapeutics. In an effort to develop non-clinical methods for measuring effects on anti-viral immunity, we have developed an interferon (IFN)-γ enzyme-linked immunosorbent spot (ELISPOT) assay to quantify the number of CMV or LCV-reactive T-cells in peripheral blood of cynomolgus macaques. After optimization of various parameters, the IFN-γ ELISPOT assay was characterized for specificity, intra-assay, monkey-to-monkey, and longitudinal variability and sensitivity to immunosuppression. The results show that nearly all animals have detectable responses against both CMV and LCV and responses were derived from T-cells specific to the virus of interest. Analyses of variability show assay reproducibility (≤23% CV), and that variability over time in anti-viral responses in individual animals (larger for LCV than for CMV) was ∼2-fold in most animals over a 3-month time period, which is predicted to allow for detection of drug-induced changes when using group sizes typical of non-clinical studies. In addition, the IFN-γ ELISPOT assay was capable of detecting decreases in the numbers of CMV and LCV reactive T-cells induced by immunosuppressive drugs in vitro. This assay may allow for non-clinical assessment of the effects of immunomodulatory therapeutics on anti-viral T-cell immunity in monkeys, and may help determine if therapeutics increase the risk of reactivating latent viral infections.


Subject(s)
Cytomegalovirus/immunology , Enzyme-Linked Immunospot Assay/methods , Herpesviridae Infections/immunology , Immunotherapy/methods , Lymphocryptovirus/immunology , T-Lymphocytes/immunology , Animals , Antigens, Viral/immunology , Drug Evaluation, Preclinical/methods , Humans , Immunity , Interferon-gamma/metabolism , Lymphocyte Activation , Macaca fascicularis , Observer Variation , Sensitivity and Specificity
18.
Cancer Immunol Immunother ; 61(10): 1721-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22406983

ABSTRACT

4-1BB (CD137, TNFRSF9) is a costimulatory receptor expressed on several subsets of activated immune cells. Numerous studies of mouse and human T cells indicate that 4-1BB promotes cellular proliferation, survival, and cytokine production. 4-1BB agonist mAbs have demonstrated efficacy in prophylactic and therapeutic settings in both monotherapy and combination therapy tumor models and have established durable anti-tumor protective T-cell memory responses. PF-05082566 is a fully human IgG2 that binds to the extracellular domain of human 4-1BB with high affinity and specificity. In preclinical studies, this agonist antibody demonstrated its ability to activate NF-κB and induce downstream cytokine production, promote leukocyte proliferation, and inhibit tumor growth in a human PBMC xenograft tumor model. The mechanism of action and robust anti-tumor efficacy of PF-05082566 support its clinical development for the treatment of a broad spectrum of human malignancies.


Subject(s)
4-1BB Ligand/agonists , Antibodies, Monoclonal/therapeutic use , Immunoglobulin G/therapeutic use , T-Lymphocytes/immunology , 4-1BB Ligand/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca fascicularis , Male , Mice , NF-kappa B/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
20.
J Immunotoxicol ; 9(1): 121-7, 2012.
Article in English | MEDLINE | ID: mdl-22136193

ABSTRACT

Epstein-Barr virus (EBV)-associated lymphomas are a known risk for immunosuppressed individuals. Non-clinical methods to determine the potential of new immunomodulatory compounds to produce EBV-associated lymphomas (hazard identification) have not been developed. Since lymphocryptovirus (LCV) in non-human primates (NHP) has similar characteristics to EBV in humans, a Roundtable meeting was held in October 2010 to explore how the potential for EBV-related lymphomas in humans can be assessed by using surrogate biomarkers for lymphoma risk in NHP toxicity studies. Stakeholders from regulatory agencies, academia, and industry came together to determine the research gaps and potential benefits and considerations of such an approach given the current state-of-the-science. Key conclusions from the discussion included considerations raised about the potential usefulness of LCV-related biomarkers from NHP studies since there is significant controversy over the reliability of using EBV viral load or EBV-specific T-lymphocytes to predict for lymphoproliferative disorders in transplant patients. In addition, there are technical challenges that need to be further addressed in order to develop methods to measure LCV viral load and LCV-specific T-lymphocytes from cynomolgus monkeys.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesviridae Infections/virology , Immunologic Factors/toxicity , Lymphocryptovirus/pathogenicity , Lymphoma/etiology , Primates , Toxicity Tests , Tumor Virus Infections/virology , Animals , Biomarkers/analysis , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/immunology , Herpesviridae Infections/diagnosis , Herpesviridae Infections/immunology , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/virology , Lymphoma/diagnosis , Lymphoma/immunology , Lymphoma/virology , Models, Animal , Risk Assessment , Risk Factors , Tumor Virus Infections/diagnosis , Tumor Virus Infections/immunology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...