Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Expert Opin Investig Drugs ; 32(6): 479-493, 2023.
Article in English | MEDLINE | ID: mdl-37394970

ABSTRACT

INTRODUCTION: Diffuse large B-cell lymphoma (DLBCL) is an aggressive and clinically heterogeneous malignancy originating from B-cells with up to 40% of patients experiencing primary refractory disease or relapse after first-line treatment. However, the past 5 years have seen a flurry of new drug approvals for DLBCL anchored upon new immune therapies, including chimeric antigen receptor (CAR) T-cells and antibody-based therapies. AREAS COVERED: This article summarizes recent advances in the treatment of DLBCL, including in the first line and relapsed and refractory setting (second-line and beyond). A literature search was conducted for publications relevant to the immunotherapeutic approach to DLBCL from 2000 through March 2023 within PubMed and articles were reviewed. The search terms were immunotherapy, monoclonal antibodies, chimeric antigen receptor modified T-cell (CAR-T), and classification of DLBCL. Relevant clinical trials and pre-clinical studies exploring the strengths and weaknesses of current immune therapies against DLBCL were chosen. We additionally explored how intrinsic differences amongst DLBCL subtype biology and endogenous host immune recruitment contribute to variable therapeutic efficacy. EXPERT OPINION: Future treatments will minimize chemotherapy exposure and be chosen by underlying tumor biology, paving the way for the promise of chemotherapeutic free regimens and improved outcomes for poor-risk subgroups.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Immunotherapy , Antibodies, Monoclonal , Immunotherapy, Adoptive
2.
Leuk Lymphoma ; 64(4): 808-815, 2023 04.
Article in English | MEDLINE | ID: mdl-36891619

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 is the current standard of care for the treatment of relapsed refractory large B cell lymphoma, demonstrating impressive response rates in the second- and third-line setting. Despite these advances, this treatment strategy can result in significant toxicities, such as cytokine release syndrome or immune effector cell associated neurotoxicity syndrome. While the exact mechanisms of these immune-mediated toxicities are not clearly understood, emerging pre-clinical and clinical studies have revealed the pivotal role of myeloid cells, particularly macrophages, as key contributors to the efficacy of treatments and as crucial mediators of toxicity. In this review, we discuss the current understanding of how macrophages mediate these effects, highlighting specific mechanisms of macrophage biology relevant to CAR T-cell therapy activity and side effects. These findings are resulting in novel treatment strategies that target macrophages, and able to mitigate toxicity while preserving CAR T-cell therapy efficacy.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell , Lymphoma, Large B-Cell, Diffuse/etiology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , T-Lymphocytes , Antigens, CD19 , Macrophages
3.
Clin Cancer Res ; 27(15): 4195-4204, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33753453

ABSTRACT

PURPOSE: Neoadjuvant immunotherapy may improve the clinical outcome of regionally advanced operable melanoma and allows for rapid clinical and pathologic assessment of response. We examined neoadjuvant pembrolizumab and high-dose IFNα-2b (HDI) therapy in patients with resectable advanced melanoma. PATIENTS AND METHODS: Patients with resectable stage III/IV melanoma were treated with concurrent pembrolizumab 200 mg i.v. every 3 weeks and HDI 20 MU/m2/day i.v., 5 days per week for 4 weeks, then 10 MU/m2/day subcutaneously 3 days per week for 2 weeks. Definitive surgery followed, as did adjuvant combination immunotherapy, completing a year of treatment. Primary endpoint was safety of the combination. Secondary endpoints included overall response rate (ORR), pathologic complete response (pCR), recurrence-free survival (RFS), and overall survival (OS). Blood samples for correlative studies were collected throughout. Tumor tissue was assessed by IHC and flow cytometry at baseline and at surgery. RESULTS: A total of 31 patients were enrolled, and 30 were evaluable. At data cutoff (October 2, 2019), median follow-up for OS was 37.87 months (range, 33.2-43.47). Median OS and RFS were not reached. Radiographic ORR was 73.3% [95% confidence interval (CI): 55.5-85.8], with a 43% (95% CI: 27.3-60.1) pCR rate. None of the patients with a pCR have had a recurrence. HDI and pembrolizumab were discontinued in 73% and 43% of patients, respectively. Correlative analyses suggested that intratumoral PD-1/PD-L1 interaction and HLA-DR expression are associated with pCR (P = 0.002 and P = 0.008, respectively). CONCLUSIONS: Neoadjuvant concurrent HDI and pembrolizumab demonstrated promising clinical activity despite high rates of treatment discontinuation. pCR is a prognostic indicator.See related commentary by Menzies et al., p. 4133.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Interferon alpha-2/administration & dosage , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Drug Therapy, Combination , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Skin Neoplasms/pathology
4.
Ticks Tick Borne Dis ; 11(5): 101468, 2020 09.
Article in English | MEDLINE | ID: mdl-32723647

ABSTRACT

Anaplasmosis is an emerging infection in the United States and remains under-recognized in many areas including Pennsylvania. Presenting signs and symptoms are often nonspecific, but fulminant infection can occur in vulnerable populations. We present two cases of severe anaplasmosis that progressed to secondary hemophagocytic lymphohistiocytosis (HLH). This severe immune dysregulation syndrome has an extremely high mortality, but anaplasmosis represents one of the few treatable underlying etiologies. It is imperative for physicians to recognize this complication and start empiric doxycycline, as early treatment improves mortality. We also present a case of anaplasmosis-induced HLH successfully treated with a combination of doxycycline, steroids, and anakinra (an IL-1 receptor antagonist), highlighting that this primarily immune-mediated complication is amenable to treatment with both antibiotics and immune suppression.


Subject(s)
Anaplasmosis/complications , Anti-Bacterial Agents/therapeutic use , Doxycycline/therapeutic use , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Lymphohistiocytosis, Hemophagocytic/therapy , Steroids/therapeutic use , Aged , Aged, 80 and over , Humans , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Male , Pennsylvania , Treatment Outcome
5.
Acta Med Acad ; 46(2): 133-144, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29338277

ABSTRACT

OBJECTIVE: Patients are frequently prescribed multiple antipsychotic medications, leading to higher healthcare costs and increased risk for side effects. The efficacy of multiple versus single antipsychotics to prevent acute relapse, measured by incidence of inpatient readmission, is investigated in Arizona, USA. METHOD: A retrospective chart review compared socio-demographic and clinical data from 1,010 patients discharged on a single and 377 discharged on multiple antipsychotic medications. Case management records were reviewed for readmission within one year of discharge. RESULTS: Younger age, diagnosis of Schizophrenia or Schizoaffective Disorder, prescription of mood stabilizer, shorter length of stay, and discharge to residential treatment or crisis recovery unit were associated with multiple antipsychotics at discharge. Readmission rates of the single (13.7%) versus multiple (15.9%) antipsychotic groups were not statistically different (p=0.286). Logistic regression analysis established that only age (younger) and the prescription of a mood stabilizer at discharge were significant predictors for increased risk for readmission (p=0.010 and p=0.049, respectively). A Cox survival analysis supported these findings. CONCLUSIONS: Concomitant antipsychotic polypharmacy at discharge did not reduce readmission risk over a one-year period. Given the increased risk of side effects and financial costs of polypharmacy, this study did not provide evidence to support this practice. Strikingly, only two variables predicted readmission risk, younger age and prescription of mood stabilizer. Although practitioners should follow practice guidelines more closely to prevent unnecessary exposure to potentially lethal side effects of antipsychotic polypharmacy, further studies are needed to better identify patients at high risk for readmission.


Subject(s)
Antipsychotic Agents/therapeutic use , Patient Readmission , Polypharmacy , Psychotic Disorders/drug therapy , Schizophrenia/drug therapy , Adult , Age Factors , Arizona , Drug Prescriptions , Drug Therapy, Combination , Female , Hospitalization , Humans , Logistic Models , Male , Residential Treatment , Retrospective Studies , Risk
6.
Oncotarget ; 8(7): 10785-10808, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-27974697

ABSTRACT

Effective adoptive immunotherapy has proved elusive for many types of human cancer, often due to difficulties achieving robust expansion of natural tumor-specific T-cells from peripheral blood. We hypothesized that antigen-driven T-cell expansion might best be triggered in vitro by acute activation of innate immunity to mimic a life-threatening infection. Unfractionated peripheral blood mononuclear cells (PBMC) were subjected to a two-step culture, first synchronizing their exposure to exogenous antigens with aggressive surrogate activation of innate immunity, followed by γ-chain cytokine-modulated T-cell hyperexpansion. Step 1 exposure to GM-CSF plus paired Toll-like receptor agonists (resiquimod and LPS), stimulated abundant IL-12 and IL-23 secretion, as well as upregulated co-stimulatory molecules and CD11c expression within the myeloid (CD33+) subpopulation. Added synthetic long peptides (>20aa) derived from widely expressed oncoproteins (MUC1, HER2/neu and CMVpp65), were reliably presented to CD4+ T-cells and cross-presented to CD8+ T-cells. Both presentation and cross-presentation demonstrated proteasomal and Sec61 dependence that could bypass the endoplasmic reticulum. Step 2 exposure to exogenous IL-7 or IL-7+IL-2 produced selective and sustained expansion of both CD4+ and CD8+ peptide-specific T-cells with a predominant interferon-γ-producing T1-type, as well as the antigen-specific ability to lyse tumor targets. Other γ-chain cytokines and/or combinations were initially proliferogenic, but followed by a contractile phase not observed with IL-7 or IL-7+IL-2. Regulatory T-cells were minimally propagated under these culture conditions. This mechanistically rational culture sequence, effective even for unvaccinated donors, enables rapid preparation of T-cells recognizing tumor-associated antigens expressed by the majority of human cancers, including pancreatic cancers, breast cancers and glioblastomas.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-7/pharmacology , Mucin-1/immunology , Receptor, ErbB-2/immunology , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cross-Priming/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Imidazoles/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-7/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mucin-1/metabolism , Neoplasms/immunology , Neoplasms/therapy , Peptides/immunology , Peptides/pharmacology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
7.
PLoS One ; 11(1): e0145920, 2016.
Article in English | MEDLINE | ID: mdl-26788922

ABSTRACT

It remains challenging to produce decisive vaccines against MUC1, a tumor-associated antigen widely expressed by pancreas, breast and other tumors. Employing clinically relevant mouse models, we ruled out such causes as irreversible T-cell tolerance, inadequate avidity, and failure of T-cells to recognize aberrantly glycosylated tumor MUC1. Instead, every tested MUC1 preparation, even non-glycosylated synthetic 9mer peptides, induced interferon gamma-producing CD4(+) and CD8(+) T-cells that recognized glycosylated variants including tumor-associated MUC1. Vaccination with synthetic peptides conferred protection as long as vaccination was repeated post tumor challenge. Failure to revaccinate post challenge was associated with down-regulated tumor MUC1 and MHC molecules. Surprisingly, direct admixture of MUC1-expressing tumor with MUC1-hyperimmune T-cells could not prevent tumor outgrowth or MUC1 immunoediting, whereas ex vivo activation of the hyperimmune T-cells prior to tumor admixture rendered them curative. Therefore, surrogate T-cell preactivation outside the tumor bed, either in culture or by repetitive vaccination, can overcome tumor escape.


Subject(s)
Cancer Vaccines/therapeutic use , Mucin-1/genetics , Mucin-1/immunology , Neoplasms, Experimental/prevention & control , Peptides/therapeutic use , Animals , Antigens/chemistry , Antigens/immunology , Antigens/therapeutic use , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Glycosylation , Humans , Mice , Mice, Transgenic , Mucin-1/metabolism , Neoplasms, Experimental/immunology , Peptides/chemistry , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape
8.
Immunol Invest ; 41(6-7): 680-710, 2012.
Article in English | MEDLINE | ID: mdl-23017141

ABSTRACT

The receptor tyrosine kinase inhibitor, sunitinib, is astonishingly effective in its capacity to reduce MDSCs in peripheral tissues such as blood (human) and spleen (mouse), restoring responsiveness of bystander T lymphocytes to TcR stimulation. Sunitinib blocks proliferation of undifferentiated MDSCs and decreases survival of more differentiated neutrophilic MDSC (n-MDSC) progeny. Ironically, sunitinib's profound effects are observed even in a total absence of detectable anti-tumor therapeutic response. This is best explained by the presence of disparate MDSC-conditioning stimuli within individual body compartments, allowing sensitivity and resistance to sunitinib to coexist within the same mouse or patient. The presence or absence of GM-CSF is likely the major determinant in each compartment, given that GM-CSF's capacity to preempt STAT3-dependent with dominant STAT5-dependent hematopoietic programming confers sunitinib resistance and redirects differentiation from the n-MDSC lineage to the more versatile monocytoid (m-MDSC) lineage. The clinical sunitinib experience underscores that strategies for MDSC and Treg depletions must be mindful of disparities among body compartments to avoid sanctuary effects. Ironically, m-MDSCs manifesting resistance to sunitinib also have the greatest potential to differentiate into tumoricidal accessory cells, by virtue of their capacity to respond to T cell-secreted IFN-γ or to TLR agonists with nitric oxide and peroxynitrate production.


Subject(s)
Indoles/therapeutic use , Myeloid Progenitor Cells/drug effects , Neoplasms/pathology , Neoplasms/therapy , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes/drug effects , Tumor Escape , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immune Tolerance , Indoles/pharmacology , Mice , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Neoplasms/immunology , Neoplasms/metabolism , Organ Specificity , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Sunitinib , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
Cancer Res ; 70(22): 9041-52, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21045144

ABSTRACT

Little preclinical modeling currently exists to support the use of OX40 agonists as therapeutic agents in the setting of advanced cancers, as well as the mechanisms through which therapeutic efficacy is achieved. We show that treatment of mice bearing well-established day 17 sarcomas with a novel OX40 ligand-Fc fusion protein (OX40L-Fc) resulted in tumor regression or dormancy in the majority of treated animals. Unexpectedly, dendritic cells (DC) in the progressive tumor microenvironment (TME) acquire OX40 expression and bind fluorescently labeled OX40L-Fc. Furthermore, longitudinal analyses revealed that DCs become enriched in the tumor-draining lymph node (TDLN) of both wild-type and Rag-/- mice within 3 days after OX40L-Fc treatment. By day 7 after treatment, a significant expansion of CXCR3+ T effector cells was noted in the TDLN, and by day 10 after treatment, type 1 polarized T cells exhibiting a reactivated memory phenotype had accumulated in the tumors. High levels of CXCL9 (a CXCR3 ligand) and enhanced expression of VCAM-1 by vascular endothelial cells (VEC) were observed in the TME early after treatment with OX40L-Fc. Notably, these vascular alterations were maintained in Rag-/- mice, indicating that the OX40L-Fc-mediated activation of both DC and VEC occurs in a T-cell-independent manner. Collectively, these findings support a paradigm in which the stimulation of DC, T cells, and the tumor vasculature by an OX40 agonist dynamically orchestrates the activation, expansion, and recruitment of therapeutic T cells into established tumors.


Subject(s)
Dendritic Cells/immunology , Endothelial Cells/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Female , Flow Cytometry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, CCR7/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , T-Lymphocyte Subsets/metabolism , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...