Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Orbit ; 42(2): 117-123, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36214105

ABSTRACT

PURPOSE: Recently, several new therapies have emerged to address locally advanced cutaneous basal cell and squamous cell carcinomas. Given the constraints of the ocular adnexa and orbit, this review was designed to discuss the role of these modalities in this region. METHODS: A PubMed search was carried out to analyze the utility of United States Food and Drug Administration-approved therapies to address these malignancies. The data presented in the identified investigations were analyzed and abstracted. RESULTS: Multiple novel interventions may be useful in the management of periocular cutaneous basal cell and squamous cell carcinomas, including imiquimod, hedgehog inhibitors, and immunotherapy. While many of these treatments have not been specifically explored in the orbit and ocular adnexa, the literature generally shows favorable response rates. However, adverse events were common in these studies. CONCLUSIONS: Several novel treatments may address periorbital cutaneous malignancies, and these therapies may be particularly useful in patients with unresectable disease and those who are poor surgical candidates.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Skin Neoplasms , United States , Humans , Carcinoma, Basal Cell/surgery , Hedgehog Proteins/therapeutic use , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/therapy
2.
Sci Immunol ; 5(48)2020 06 05.
Article in English | MEDLINE | ID: mdl-32503877

ABSTRACT

Patients with severe COVID-19 have a hyperinflammatory immune response suggestive of macrophage activation. Bruton tyrosine kinase (BTK) regulates macrophage signaling and activation. Acalabrutinib, a selective BTK inhibitor, was administered off-label to 19 patients hospitalized with severe COVID-19 (11 on supplemental oxygen; 8 on mechanical ventilation), 18 of whom had increasing oxygen requirements at baseline. Over a 10-14 day treatment course, acalabrutinib improved oxygenation in a majority of patients, often within 1-3 days, and had no discernable toxicity. Measures of inflammation - C-reactive protein and IL-6 - normalized quickly in most patients, as did lymphopenia, in correlation with improved oxygenation. At the end of acalabrutinib treatment, 8/11 (72.7%) patients in the supplemental oxygen cohort had been discharged on room air, and 4/8 (50%) patients in the mechanical ventilation cohort had been successfully extubated, with 2/8 (25%) discharged on room air. Ex vivo analysis revealed significantly elevated BTK activity, as evidenced by autophosphorylation, and increased IL-6 production in blood monocytes from patients with severe COVID-19 compared with blood monocytes from healthy volunteers. These results suggest that targeting excessive host inflammation with a BTK inhibitor is a therapeutic strategy in severe COVID-19 and has led to a confirmatory international prospective randomized controlled clinical trial.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Benzamides/pharmacology , Benzamides/therapeutic use , Betacoronavirus , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Pyrazines/pharmacology , Pyrazines/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/metabolism , Aged , Aged, 80 and over , COVID-19 , Coronavirus Infections/virology , Critical Illness , Female , Follow-Up Studies , Humans , Inflammation/drug therapy , Inflammation/virology , Interleukin-6/metabolism , Male , Middle Aged , Monocytes/metabolism , Pandemics , Pneumonia, Viral/virology , Prospective Studies , Respiration, Artificial , SARS-CoV-2 , Treatment Outcome , COVID-19 Drug Treatment
3.
Exp Hematol Oncol ; 2(1): 8, 2013 Mar 09.
Article in English | MEDLINE | ID: mdl-23497451

ABSTRACT

Among the dendritic cell (DC) subsets, plasmacytoid DC's (pDC) are thought to be important in the generation of both antiviral and antitumor responses. While pDC may be useful in developing dendritic cell-based tumor vaccines, the low frequency of these cells in the peripheral blood has hampered attempts to understand their biology. To provide better insight into the biology of pDC, we isolated these unperturbed cells from the peripheral blood of healthy donors in order to further characterize their gene expression. Using gene array technology we compared the genetic profiles of these cells to those of CD14+ monocytes isolated from the same donors and found several immune related genes upregulated in this cell population. This is the first description, to our knowledge, of gene expression in this subset of DCs obtained from the peripheral blood of adult human donors without exposure in vitro to cytokine or growth factors. Understanding the natural genetic profiles of this dendritic cell subtype as well as others such as the BDCA-1 expressing myeloid DCs may enable us to manipulate these cells ex-vivo to generate enhanced DC-based tumor vaccines inducing more robust antitumor responses.

5.
Nat Mater ; 11(10): 895-905, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22797827

ABSTRACT

The tumour microenvironment thwarts conventional immunotherapy through multiple immunologic mechanisms, such as the secretion of the transforming growth factor-ß (TGF-ß), which stunts local tumour immune responses. Therefore, high doses of interleukin-2 (IL-2), a conventional cytokine for metastatic melanoma, induces only limited responses. To overcome the immunoinhibitory nature of the tumour microenvironment, we developed nanoscale liposomal polymeric gels (nanolipogels; nLGs) of drug-complexed cyclodextrins and cytokine-encapsulating biodegradable polymers that can deliver small hydrophobic molecular inhibitors and water-soluble protein cytokines in a sustained fashion to the tumour microenvironment. nLGs releasing TGF-ß inhibitor and IL-2 significantly delayed tumour growth, increased survival of tumour-bearing mice, and increased the activity of natural killer cells and of intratumoral-activated CD8(+) T-cell infiltration. We demonstrate that the efficacy of nLGs in tumour immunotherapy results from a crucial mechanism involving activation of both innate and adaptive immune responses.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotherapy/methods , Interleukin-2/administration & dosage , Nanostructures , Neoplasms, Experimental/therapy , Transforming Growth Factor beta/antagonists & inhibitors , Adaptive Immunity , Animals , Antineoplastic Agents/pharmacology , Cyclodextrins , Drug Compounding , Gels , Immunity, Innate , Interleukin-2/pharmacology , Killer Cells, Natural/metabolism , Liposomes , Mice , Mice, Inbred Strains , Neoplasms, Experimental/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects
6.
Clin Liver Dis ; 15(2): 423-41, vii-x, 2011 May.
Article in English | MEDLINE | ID: mdl-21689622

ABSTRACT

Many potential systemic therapies are being investigated for the treatment of hepatocellular carcinoma (HCC). The incidence of this malignancy is rising sharply and the vast majority of patients present at advanced stages. Although the earlier dismal results with cytotoxic chemotherapies made way for the development of locoregional therapies that provided improved overall survival, truly personalized therapy will require the selection of phenotypically similar stages of disease and populations, an understanding of the complex molecular and genetic pathways leading to HCC, and a keen understanding of the pathobiology of cirrhosis. Only then will we understand how to offer a particular patient at a specific stage of disease the appropriate therapy to truly prolong survival.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Animals , Clinical Trials as Topic , Humans , Liver Neoplasms, Experimental/drug therapy
7.
Nat Rev Immunol ; 10(8): 554-67, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616810

ABSTRACT

Transforming growth factor-beta (TGFbeta) is an immunosuppressive cytokine produced by tumour cells and immune cells that can polarize many components of the immune system. This Review covers the effects of TGFbeta on natural killer (NK) cells, dendritic cells, macrophages, neutrophils, CD8(+) and CD4(+) effector and regulatory T cells, and NKT cells in animal tumour models and in patients with cancer. Collectively, many recent studies favour the hypothesis that blocking TGFbeta-induced signalling in the tumour microenvironment enhances antitumour immunity and may be beneficial for cancer therapy. An overview of the current drugs and reagents available for inhibiting TGFbeta-induced signalling and their phase in clinical development is also provided.


Subject(s)
Cell Polarity/immunology , Neoplasms/immunology , Transforming Growth Factor beta/immunology , Animals , Antineoplastic Agents/pharmacology , Cytokines/immunology , Dendritic Cells/immunology , Hematopoiesis, Extramedullary , Humans , Immunosuppression Therapy , Killer Cells, Natural/immunology , Macrophages/immunology , Neoplasms/therapy , Neutrophils/immunology , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
8.
Expert Opin Biol Ther ; 9(4): 451-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19344282

ABSTRACT

BACKGROUND: Recent findings on T cells and dendritic cells have elucidated principles that can be used for a bottom-up approach to engineering artificial antigen presentation on synthetic substrates. OBJECTIVE/METHODS: To compare the latest artificial antigen-presenting cell (aAPC) technology, focussing on acellular systems because they offer advantages such as easy tunability and rapid point-of-care application compared with cellular systems. We review acellular aAPC performance and discuss their promise for clinical applications. RESULTS/CONCLUSION: Acellular aAPCs are a powerful alternative to natural-cell-based therapies, offering flexibility and modularity for incorporation oSf a variety of stimuli, hence increasing precision. Current technologies should adapt physiologically important signals within safe materials to more closely approximate their cellular counterparts. These constructs could be administered parenterally as APC replacements for active vaccines or used ex vivo for adoptive immunotherapy.


Subject(s)
Adoptive Transfer/methods , Antigen Presentation/immunology , Vaccines, Acellular/immunology , Adoptive Transfer/trends , Animals , Humans , Immunotherapy, Active/methods , Immunotherapy, Active/trends , Vaccines, Acellular/administration & dosage
9.
Hum Gene Ther ; 20(1): 51-61, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19025415

ABSTRACT

mRNA transfection is a useful approach for temporal cell reprogramming with minimal risk of transgene-mediated mutagenesis. We applied this to redirect lymphocyte cytotoxicity toward malignant cells. Using the chimeric immune receptor (CIR) constructs anti-CD19 CIR and 8H9 CIR, we achieved uniform expression of CIRs on virtually the entire population of lymphocytes. We reprogrammed CD3+ CD8+, CD3+ CD4+, and natural killer (NK ) cells toward autologous and allogeneic targets such as B cells, Daudi lymphoma, primary melanoma, breast ductal carcinoma, breast adenocarcinoma, and rhabdomyosarcoma. The reprogramming procedure is fast. Although most of the experiments were performed on lymphocytes obtained after 7-day activation, only 1-day activation of T cells with anti-CD3, anti-CD28 antibodies, and interleukin-2 is sufficient to develop both lymphocyte cytotoxicity and competence for mRNA transfer. The entire procedure, which includes lymphocyte activation and reprogramming, can be completed in 2 days. The efficiency of mRNA-modified human T cells was tested in a murine xenograft model. Human CD3+CD8+ lymphocytes expressing anti-CD19 CIR mRNA inhibited Daudi lymphoma growth in NOD=SCID mice. These results demonstrate that a mixed population of cytotoxic lymphocytes, including T cells together with NK cells, can be quickly and simultaneously reprogrammed by mRNA against autologous malignancies. With relatively minor modifications the described method of lymphocyte reprogramming can be scaled up for cancer therapy.


Subject(s)
Antigens, CD19/immunology , Cytotoxicity, Immunologic , Lymphoma/immunology , RNA, Messenger/metabolism , Receptors, Immunologic , Recombinant Fusion Proteins , T-Lymphocytes/immunology , Transfection , Animals , Female , Humans , Lymphocyte Activation , Lymphoma/genetics , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Transgenes , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 13(18 Pt 1): 5262-70, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875754

ABSTRACT

Immune homeostasis is a delicate balance between the immune defense against foreign pathogens and suppression of the immune system to maintain self-tolerance and prevent autoimmune disease. Maintenance of this balance involves several crucial networks of cytokines and various cell types. Among these regulators, transforming growth factor-beta (TGF-beta) is a potent cytokine with diverse effects on hematopoietic cells. Its pivotal function within the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis and activation of leukocytes in the periphery, including lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. Through its pleiotropic effects on these immune cells, TGF-beta prevents the development of autoimmune diseases without compromising immune responses to pathogens. However, overactivation of this pathway can lead to several immunopathologies under physiologic conditions including cancer progression, making it an attractive target for antitumor therapies. This review discusses the biological functions of TGF-beta and its effects on the immune system and addresses how immunosuppression by this cytokine can promote tumorigenesis, providing the rationale for evaluating the immune-enhancing and antitumor effects of inhibiting TGF-beta in cancer patients.


Subject(s)
Neoplasms/drug therapy , Neoplasms/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/physiology , Animals , Dendritic Cells/immunology , Humans , Killer Cells, Natural/immunology , T-Lymphocytes/immunology
11.
Anticancer Drugs ; 18(6): 721-4, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17762403

ABSTRACT

Since the results of the MOSAIC trial demonstrated an improved disease-free survival in stage III colorectal patients treated with oxaliplatin combined with 5-fluorouracil and folinic acid when they were compared with those treated with 5-fluorouracil and folinic acid alone, the addition of this organoplatin to 5-fluorouracil and folinic acid has become first-line adjuvant treatment for stage III colorectal cancer. Unfortunately, there is a small population of patients who develop grade III/IV hypersensitivity reactions to oxaliplatin which, until recently, have interfered with further treatment with oxaliplatin-containing regimens. Successful oxaliplatin desensitization protocols for patients having severe oxaliplatin hypersensitivity reactions have been reported. However, none of these protocols, have incorporated magnesium and calcium salts. Retrospective data has suggested that pretreating colorectal cancer patients with magnesium sulfate and calcium gluconate before the administration of oxaliplatin may reduce the incidence of neurotoxicities induced by this drug. Therefore, we modified a previously published oxaliplatin-desensitization protocol by incorporating intravenous calcium gluconate and magnesium sulfate, and report a patient with stage IIIc colorectal cancer and prior severe hypersensitivity reactions to oxaliplatin who underwent successful oxaliplatin desensitization using this protocol.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Calcium Gluconate/therapeutic use , Drug Hypersensitivity/prevention & control , Magnesium Sulfate/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Calcium Gluconate/administration & dosage , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/therapeutic use , Magnesium Sulfate/administration & dosage , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Sigmoid Neoplasms/drug therapy , Sigmoid Neoplasms/pathology , Treatment Outcome
12.
J Neurooncol ; 76(3): 277-81, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16244794

ABSTRACT

A 59-year-old male was transferred to our facility after initial workup for left eye blindness revealed multiple brain lesions. The patient presented with scant pedunculated skin lesions on his neck and arms, axillary freckling and bilateral subcutaneous ankle nodules suggestive of neurofibromatosis type 1 (NF-1). Neurological examination revealed left-sided blindness, diminished pupillary response to light, incomplete eyelid closure and facial droop. A CT series of the chest/abdomen/pelvis demonstrated several lesions in the lungs, liver and left kidney. CT-guided biopsy of the renal lesion was "consistent with metastatic melanoma" based on the presence of malignant cell staining for the melanocyte differentiation antigen MART-1, as well as the calcium binding protein S-100. A dermatology consultant confirmed neurofibromas and a café au lait macule. No dysplastic nevi or primary melanoma were identified. An ophthalmology consultant noted gray choroidal lesions on fundoscopic examination thought to be metastatic disease of unknown primary. Despite treatment with whole brain irradiation and intravenous steroids, the patient died on hospital day 10. A full autopsy showed diffuse neurofibromas and café au lait macules consistent with NF-1 and malignant melanocytic schwannomas with multiple metastases.


Subject(s)
Blindness/etiology , Neoplasms, Second Primary/pathology , Neurilemmoma/pathology , Neurofibromatosis 1/pathology , Optic Nerve Neoplasms/pathology , Cell Transformation, Neoplastic , Humans , Immunohistochemistry , Male , Melanocytes/pathology , Middle Aged , Neoplasms, Second Primary/metabolism , Neurilemmoma/complications , Neurilemmoma/metabolism , Neurofibromatosis 1/metabolism , Optic Chiasm/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...