Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cancers (Basel) ; 14(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35884422

ABSTRACT

Triple-negative breast cancers (TNBC) expressing PD-L1 qualify for checkpoint inhibitor immunotherapy. Cyclin E/CDK2 is a potential target axis in TNBC; however, small-molecule drugs at efficacious doses may be associated with toxicity, and treatment alongside immunotherapy requires investigation. We evaluated CDK inhibition at suboptimal levels and its anti-tumor and immunomodulatory effects. Transcriptomic analyses of primary breast cancers confirmed higher cyclin E/CDK2 expression in TNBC compared with non-TNBC. Out of the three CDK2-targeting inhibitors tested, the CDK 2, 7 and 9 inhibitor SNS-032 was the most potent in reducing TNBC cell viability and exerted cytotoxicity against all eight TNBC cell lines evaluated in vitro. Suboptimal SNS-032 dosing elevated cell surface PD-L1 expression in surviving TNBC cells. In mice engrafted with human immune cells and challenged with human MDA-MB-231 TNBC xenografts in mammary fat pads, suboptimal SNS-032 dosing partially restricted tumor growth, enhanced the tumor infiltration of human CD45+ immune cells and elevated cell surface PD-L1 expression in surviving cancer cells. In tumor-bearing mice engrafted with human immune cells, the anti-PD-L1 antibody avelumab, given sequentially following suboptimal SNS-032 dosing, reduced tumor growth compared with SNS-032 alone or with avelumab without prior SNS-032 priming. CDK inhibition at suboptimal doses promotes immune cell recruitment to tumors, PD-L1 expression by surviving TNBC cells and may complement immunotherapy.

2.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34112739

ABSTRACT

BACKGROUND: Cancer immunotherapy with monoclonal antibodies and chimeric antigen receptor (CAR) T cell therapies can benefit from selection of new targets with high levels of tumor specificity and from early assessments of efficacy and safety to derisk potential therapies. METHODS: Employing mass spectrometry, bioinformatics, immuno-mass spectrometry and CRISPR/Cas9 we identified the target of the tumor-specific SF-25 antibody. We engineered IgE and CAR T cell immunotherapies derived from the SF-25 clone and evaluated potential for cancer therapy. RESULTS: We identified the target of the SF-25 clone as the tumor-associated antigen SLC3A2, a cell surface protein with key roles in cancer metabolism. We generated IgE monoclonal antibody, and CAR T cell immunotherapies each recognizing SLC3A2. In concordance with preclinical and, more recently, clinical findings with the first-in-class IgE antibody MOv18 (recognizing the tumor-associated antigen Folate Receptor alpha), SF-25 IgE potentiated Fc-mediated effector functions against cancer cells in vitro and restricted human tumor xenograft growth in mice engrafted with human effector cells. The antibody did not trigger basophil activation in cancer patient blood ex vivo, suggesting failure to induce type I hypersensitivity, and supporting safe therapeutic administration. SLC3A2-specific CAR T cells demonstrated cytotoxicity against tumor cells, stimulated interferon-γ and interleukin-2 production in vitro. In vivo SLC3A2-specific CAR T cells significantly increased overall survival and reduced growth of subcutaneous PC3-LN3-luciferase xenografts. No weight loss, manifestations of cytokine release syndrome or graft-versus-host disease, were detected. CONCLUSIONS: These findings identify efficacious and potentially safe tumor-targeting of SLC3A2 with novel immune-activating antibody and genetically modified cell therapies.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/immunology , Immunoglobulin E/metabolism , Immunotherapy/methods , Receptors, Chimeric Antigen/immunology , Animals , Humans , Mice
3.
MAbs ; 12(1): 1685349, 2020.
Article in English | MEDLINE | ID: mdl-31769737

ABSTRACT

IgE monoclonal antibodies hold great potential for cancer therapy. Preclinical in vivo systems, particularly those in which the antibody recognizes the host species target antigen and binds to cognate Fc receptors, are often the closest approximation to human exposure and represent a key challenge for evaluating the safety of antibody-based therapies. We sought to develop an immunocompetent rat system to assess the safety of a rodent anti-tumor IgE, as a surrogate for the human therapeutic candidate. We generated a rat IgE against the human tumor-associated antigen chondroitin sulfate proteoglycan 4 (CSPG4) and cross-reactive for the rat antigen. We analyzed CSPG4 distribution in normal rat and human tissues and investigated the in vivo safety of the antibody by monitoring clinical signs and molecular biomarkers after systemic administration to immunocompetent rats. Human and rat CSPG4 expression in normal tissues were comparable. Animals receiving antibody exhibited transient mild to moderate adverse events accompanied by mild elevation of serum tryptase, but not of angiotensin II or cytokines implicated in allergic reactions or cytokine storm. In the long term, repeated antibody administration was well tolerated, with no changes in animal body weight, liver and kidney functions or blood cell counts. This model provides preclinical support for the safety profiling of IgE therapeutic antibodies. Due to the comparable antigen tissue distribution in human and rat, this model may also comprise an appropriate tool for proof-of-concept safety evaluations of different treatment approaches targeting CSPG4.


Subject(s)
Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , Chondroitin Sulfate Proteoglycans/immunology , Immunoglobulin E/administration & dosage , Membrane Proteins/immunology , Recombinant Fusion Proteins/administration & dosage , Animals , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/adverse effects , Cell Line, Tumor , Cross Reactions , Female , Humans , Immunization, Secondary , Immunocompetence , Immunoglobulin E/adverse effects , Mice , Rats , Recombinant Fusion Proteins/adverse effects
4.
Front Immunol ; 10: 453, 2019.
Article in English | MEDLINE | ID: mdl-30941125

ABSTRACT

The immune system employs several checkpoint pathways to regulate responses, maintain homeostasis and prevent self-reactivity and autoimmunity. Tumor cells can hijack these protective mechanisms to enable immune escape, cancer survival and proliferation. Blocking antibodies, designed to interfere with checkpoint molecules CTLA-4 and PD-1/PD-L1 and counteract these immune suppressive mechanisms, have shown significant success in promoting immune responses against cancer and can result in tumor regression in many patients. While inhibitors to CTLA-4 and the PD-1/PD-L1 axis are well-established for the clinical management of melanoma, many patients do not respond or develop resistance to these interventions. Concerted efforts have focused on combinations of approved therapies aiming to further augment positive outcomes and survival. While CTLA-4 and PD-1 are the most-extensively researched targets, results from pre-clinical studies and clinical trials indicate that novel agents, specific for checkpoints such as A2AR, LAG-3, IDO and others, may further contribute to the improvement of patient outcomes, most likely in combinations with anti-CTLA-4 or anti-PD-1 blockade. This review discusses the rationale for, and results to date of, the development of inhibitory immune checkpoint blockade combination therapies in melanoma. The clinical potential of new pipeline therapeutics, and possible future therapy design and directions that hold promise to significantly improve clinical prognosis compared with monotherapy, are discussed.


Subject(s)
Antibodies, Monoclonal/immunology , Immune System/drug effects , Immune System/immunology , Melanoma/immunology , Melanoma/therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Humans , Immunotherapy/methods
5.
J Allergy Clin Immunol ; 144(1): 236-253, 2019 07.
Article in English | MEDLINE | ID: mdl-30738173

ABSTRACT

BACKGROUND: Gain-of-function (GOF) mutations in PIK3CD cause a primary immunodeficiency characterized by recurrent respiratory tract infections, susceptibility to herpesvirus infections, and impaired antibody responses. Previous work revealed defects in CD8+ T and B cells that contribute to this clinical phenotype, but less is understood about the role of CD4+ T cells in disease pathogenesis. OBJECTIVE: We sought to dissect the effects of increased phosphoinositide 3-kinase (PI3K) signaling on CD4+ T-cell function. METHODS: We performed detailed ex vivo, in vivo, and in vitro phenotypic and functional analyses of patients' CD4+ T cells and a novel murine disease model caused by overactive PI3K signaling. RESULTS: PI3K overactivation caused substantial increases in numbers of memory and follicular helper T (TFH) cells and dramatic changes in cytokine production in both patients and mice. Furthermore, PIK3CD GOF human TFH cells had dysregulated phenotype and function characterized by increased programmed cell death protein 1, CXCR3, and IFN-γ expression, the phenotype of a TFH cell subset with impaired B-helper function. This was confirmed in vivo in which Pik3cd GOF CD4+ T cells also acquired an aberrant TFH phenotype and provided poor help to support germinal center reactions and humoral immune responses by antigen-specific wild-type B cells. The increase in numbers of both memory and TFH cells was largely CD4+ T-cell extrinsic, whereas changes in cytokine production and TFH cell function were cell intrinsic. CONCLUSION: Our studies reveal that CD4+ T cells with overactive PI3K have aberrant activation and differentiation, thereby providing mechanistic insight into dysfunctional antibody responses in patients with PIK3CD GOF mutations.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Differentiation , Phosphatidylinositol 3-Kinases/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Gain of Function Mutation , Humans , Mice , Phenotype
6.
J Exp Med ; 215(8): 2073-2095, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30018075

ABSTRACT

Gain-of-function (GOF) mutations in PIK3CD, encoding the p110δ subunit of phosphatidylinositide 3-kinase (PI3K), cause a primary immunodeficiency. Affected individuals display impaired humoral immune responses following infection or immunization. To establish mechanisms underlying these immune defects, we studied a large cohort of patients with PIK3CD GOF mutations and established a novel mouse model using CRISPR/Cas9-mediated gene editing to introduce a common pathogenic mutation in Pik3cd In both species, hyperactive PI3K severely affected B cell development and differentiation in the bone marrow and the periphery. Furthermore, PI3K GOF B cells exhibited intrinsic defects in class-switch recombination (CSR) due to impaired induction of activation-induced cytidine deaminase (AID) and failure to acquire a plasmablast gene signature and phenotype. Importantly, defects in CSR, AID expression, and Ig secretion were restored by leniolisib, a specific p110δ inhibitor. Our findings reveal key roles for balanced PI3K signaling in B cell development and long-lived humoral immunity and memory and establish the validity of treating affected individuals with p110δ inhibitors.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Class I Phosphatidylinositol 3-Kinases/genetics , Germ-Line Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Animals , Antibody Affinity/immunology , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Child , Gain of Function Mutation/genetics , Humans , Immunoglobulin Class Switching , Immunoglobulins/metabolism , Interleukins/pharmacology , Mice , Models, Animal , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Plasma Cells/metabolism , Signal Transduction
7.
Arch Psychiatr Nurs ; 29(4): 249-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26165981

ABSTRACT

Maternal stress during pregnancy has been associated with numerous adverse pregnancy, birth, and health outcomes. Pregnant African American women have been reported to have higher levels of stress compared to other ethnic or racial groups underscoring the need for effective interventions to reduce stress in this population. The purpose of this study was to gain an in-depth understanding of the perceptions of guided imagery (GI) as a technique for stress management in a cohort of pregnant African American women who participated in a GI intervention as part of a larger mixed methods randomized controlled trial. The 12week intervention was a professionally recorded compact disc with four tracks developed and sequenced to reduce stress and associated symptoms. The findings from this descriptive phenomenologic study were derived from daily logs and interviews from 36 participants randomized to the GI group. Participants described the stressful nature of their lives. Results demonstrated pregnant African American women perceived the intervention as beneficial in reducing stress and the associated symptoms. The emergent themes suggested the intervention offered a respite from their stressful lives, reduced the negative emotional responses to stress and enhanced well-being, benefited other areas of their daily life, and provided an opportunity to connect with their baby. The study results support the perceived efficacy of GI as a stress coping intervention. GI is an economic as well as easy to implement, access and use technique that has potential stress coping benefits as perceived by pregnant African American women.


Subject(s)
Black or African American/psychology , Imagery, Psychotherapy/methods , Pregnancy Complications/psychology , Stress, Psychological/therapy , Adolescent , Adult , Female , Humans , Patient Satisfaction , Pregnancy , Pregnancy Complications/therapy , Treatment Outcome , Young Adult
8.
Article in English | MEDLINE | ID: mdl-24719646

ABSTRACT

The purpose of this study was to evaluate the efficacy of a guided imagery (GI) intervention for stress reduction in pregnant African American women beginning early in the second trimester. This prospective longitudinal study of 72 women used a randomized controlled experimental design with two groups conducted over 12 weeks. The intervention was a CD with 4 professionally recorded tracts designed and sequenced to influence study variables. Participants in both GI and usual care (UC) completed measures and donated 5 cc of blood at baseline, 8 weeks and 12 weeks. Participants also completed a daily stress scale. A mixed-effects linear model tested for differences between groups for self-reported measures of stress, anxiety, and fatigue as well as corticotrophin releasing hormone (CRH), a biologic marker of stress. Significant differences in perceived stress daily scores and at week 8 but not week 12 were found in the GI group compared to UC group. The GI group reported significantly less fatigue and anxiety than the UC group at week 8 but not week 12. There were no significant differences in CRH levels between groups. Results suggest that GI intervention may be effective in reducing perceived stress, anxiety, and fatigue measures among pregnant African American women.

SELECTION OF CITATIONS
SEARCH DETAIL
...