Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Clin Aesthet Dermatol ; 14(12): 44-48, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35096254

ABSTRACT

BACKGROUND: It is often difficult to accurately predict how a melanoma will progress because melanomas can be so diverse in their genetic and histological makeup. OBJECTIVE: We sought to characterize the current state and progression of biomedical markers towards their utilization as prognostic indicators for patients with melanoma. METHODS: A literature search of the research repository databases PubMed and GoogleScholar was conducted using the following inclusion criteria: (1) published within the last 10 years, and (2) use of overall survival, disease progression, or clinical outcome as primary endpoints. Search terms included various permutations of "biomarkers," "prognostic," "immunologic," "serologic," "visual," and "melanoma." Results were evaluated for statistical power, results significance, and experimental design integrity. RESULTS: The prognostic capabilities of clinical tests for malignant melanoma have made great strides in the last few years, with several serologic and immunohistochemical biomarkers being preliminarily linked to various measures of clinical prognosis. While clinical feasibility of a single sensitive and specific biomarker remains unfeasible, use of select combinations of tested biomarkers remain viable. CONCLUSION: Diagnostic and prognostic genetic assays have begun to cross over from research to commercial application, giving physicians additional tools during the early stages of diagnosis to optimize and individualize treatments.

2.
PLoS One ; 10(7): e0133152, 2015.
Article in English | MEDLINE | ID: mdl-26173023

ABSTRACT

Patients with metastatic or recurrent and refractory sarcomas have a dismal prognosis. Therefore, new targeted therapies are urgently needed. This study was designed to evaluate chimeric antigen receptor (CAR) T cells targeting the type I insulin-like growth factor receptor (IGF1R) or tyrosine kinase-like orphan receptor 1 (ROR1) molecules for their therapeutic potential against sarcomas. Here, we report that IGF1R (15/15) and ROR1 (11/15) were highly expressed in sarcoma cell lines including Ewing sarcoma, osteosarcoma, alveolar or embryonal rhabdomyosarcoma, and fibrosarcoma. IGF1R and ROR1 CAR T cells derived from eight healthy donors using the Sleeping Beauty (SB) transposon system were cytotoxic against sarcoma cells and produced high levels of IFN-γ, TNF-α and IL-13 in an antigen-specific manner. IGF1R and ROR1 CAR T cells generated from three sarcoma patients released significant amounts of IFN-γ in response to sarcoma stimulation. The adoptive transfer of IGF1R and ROR1 CAR T cells derived from a sarcoma patient significantly reduced tumor growth in pre-established, systemically disseminated and localized osteosarcoma xenograft models in NSG mice. Infusion of IGF1R and ROR1 CAR T cells also prolonged animal survival in a localized sarcoma model using NOD/scid mice. Our data indicate that both IGF1R and ROR1 can be effectively targeted by SB modified CAR T cells and that such CAR T cells may be useful in the treatment of high risk sarcoma patients.


Subject(s)
Bone Neoplasms/immunology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Somatomedin/metabolism , Sarcoma/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/therapy , Cell Line, Tumor , DNA Transposable Elements/genetics , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , K562 Cells , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Receptor Tyrosine Kinase-like Orphan Receptors/immunology , Receptor, IGF Type 1 , Receptors, Antigen, T-Cell/immunology , Sarcoma/metabolism , Sarcoma/therapy , T-Lymphocytes/metabolism
3.
J Biomed Opt ; 18(12): 126014, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24346856

ABSTRACT

To develop an indocyanine green (ICG) tracer with slower clearance kinetics, we explored ICG-encapsulating liposomes (Lip) in three different formulations: untargeted (Lip/ICG), targeted to vascular endothelial growth factor (VEGF) receptors (scVEGF-Lip/ICG) by the receptor-binding moiety single-chain VEGF (scVEGF), or decorated with inactivated scVEGF (inactive-Lip/ICG) that does not bind to VEGF receptors. Experiments were conducted with tumor-bearing mice that were placed in a scattering medium with tumors located at imaging depths of either 1.5 or 2.0 cm. Near-infrared fluorescence diffuse optical tomography that provides depth-resolved spatial distributions of fluorescence in tumor was used for the detection of postinjection fluorescent signals. All liposome-based tracers, as well as free ICG, were injected intravenously into mice in the amounts corresponding to 5 nmol of ICG/mouse, and the kinetics of increase and decrease of fluorescent signals in tumors were monitored. A signal from free ICG reached maximum at 15-min postinjection and then rapidly declined with t1/2 of ~20 min. The signals from untargeted Lip/ICG and inactive-Lip/ICG also reached maximum at 15-min postinjection, however, declined somewhat slower than free ICG with t1/2 of ~30 min. By contrast, a signal from targeted scVEGF-Lip/ICG grew slower than that of all other tracers, reaching maximum at 30-min postinjection and declined much slower than that of other tracers with t1/2 of ~90 min, providing a more extended observation window. Higher scVEGF-Lip/ICG tumor accumulation was further confirmed by the analysis of fluorescence on cryosections of tumors that were harvested from animals at 400 min after injection with different tracers.


Subject(s)
Fluorescent Dyes/metabolism , Indocyanine Green/metabolism , Liposomes/metabolism , Mammary Neoplasms, Experimental/pathology , Receptors, Vascular Endothelial Growth Factor/metabolism , Tomography, Optical/methods , Animals , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Humans , Indocyanine Green/chemistry , Liposomes/chemistry , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C
4.
J Nucl Med ; 52(10): 1630-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21890879

ABSTRACT

UNLABELLED: Tumor vessels abundantly express receptors for vascular endothelial growth factor (VEGF), despite treatment with conventional or antiangiogenic drugs. We wished to determine whether the high levels of VEGF receptor (VEGFR) within the tumor vasculature could be leveraged for intracellular delivery of therapeutically significant doses of scVEGF/(177)Lu, a novel radiopharmaceutical based on a recombinant single-chain (sc) derivative of VEGF, in orthotopic breast cancer models. METHODS: scVEGF-PEG (polyethylene gycol)-DOTA conjugates containing 2.0-, 3.4-, or 5.0-kDa PEG linkers site-specifically conjugated to a cysteine-containing tag (Cys-tag) in scVEGF were radiolabeled with (177)Lu (scVEGF/(177)Lu) for in vivo studies. Human MDA231luc and mouse 4T1luc cell lines were injected orthotopically to establish breast carcinoma tumors in immunodeficient and immunocompetent hosts, respectively. The effects of scVEGF/(177)Lu were defined by analysis of changes in tumor growth and immunohistochemical staining for the endothelial markers CD31 and VEGFR-2 and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining for intratumoral apoptosis. RESULTS: Biodistribution assays and dosimetric calculations established that scVEGF/(177)Lu with a 3.4-kDa PEG linker delivered the highest dose of radiation to tumors (69.9 cGy/MBq/g of tissue) and the lowest dose to the kidneys (33.3 cGy/MBq/organ). Total doses below 40 MBq/mouse of scVEGF/(177)Lu did not affect renal function, and 3 divided doses of 6.3 MBq/mouse or a bolus dose of 18.9 MBq/mouse induced only transient lymphopenia and weight loss (<10% baseline weight). In mice with orthotopic mammary breast carcinoma, intravenous injections of well-tolerated bolus and fractionated doses of scVEGF/(177)Lu in the range from 6.3 to 18.9 MBq/mouse (25-76 MBq/m(2)) resulted in dose-dependent tumor growth inhibition. Immunohistochemical analysis of tumors at 4-5 wk after single injections of scVEGF/(177)Lu indicated dose-dependent regression of tumor vasculature and widespread intratumoral apoptosis. A single dose of 7.4 MBq/mouse of scVEGF/(177)Lu given before a course of bevacizumab or sunitinib treatment enhanced the antiangiogenic effects of both drugs. CONCLUSION: Selective targeting of VEGFR in tumor vasculature with well-tolerated doses of scVEGF/(177)Lu is effective in orthotopic breast cancer models. As high levels of VEGFR expression in the tumor vasculature are a common feature in a variety of cancers, targeting tumor angiogenesis with scVEGF/(177)Lu warrants further exploration.


Subject(s)
Breast Neoplasms/radiotherapy , Lutetium/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Apoptosis/radiation effects , Bevacizumab , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/radiation effects , Combined Modality Therapy , Endothelial Cells/radiation effects , Female , Humans , Indoles/therapeutic use , Lutetium/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, SCID , Neovascularization, Pathologic/radiotherapy , Pyrroles/therapeutic use , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/radiation effects , Sunitinib , Vascular Endothelial Growth Factor A/pharmacokinetics
5.
J Nucl Med ; 51(6): 959-66, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484434

ABSTRACT

UNLABELLED: Several drugs targeting vascular endothelial growth factor (VEGF) and its receptors (VEGFRs) are approved for cancer treatment. However, these drugs induce relatively modest and frequently unpredictable tumor responses. In this work, we explored whether noninvasive imaging of VEGFR, a direct target of antiangiogenic drugs, can provide real-time information on responses to the treatment with sunitinib, a small-molecule VEGFR inhibitor approved by the Food and Drug Administration. METHODS: We imaged VEGFR in an orthotopic mammary tumor model during the course of treatment with sunitinib using a recently developed SPECT tracer, a (99m)Tc-labeled single-chain VEGF (scVEGF), that binds to and is internalized by VEGFR. Tumors from imaged mice were harvested and cryosectioned, and alternating sections were analyzed by autoradiography and immunohistochemistry to determine the expression of endothelial cell markers VEGFR-2 and CD31. RESULTS: In vitro assays with endothelial cells overexpressing VEGFR-2 established that sunitinib does not inhibit VEGFR-2-mediated uptake of scVEGF-based tracers. SPECT and autoradiography with (99m)Tc-scVEGF of tumor cryosections revealed a 2.2- to 2.6-fold decrease in tracer uptake after 4 daily doses of sunitinib. However, once treatment was discontinued, tracer uptake rapidly (3 d) increased, particularly at the tumor edges. Immunohistochemical analysis of VEGFR-2 and CD31 supported SPECT and autoradiographic imaging findings, revealing the corresponding depletion of VEGFR-2- and CD31-positive endothelial cells from tumor vasculature during therapy and the rapid reemergence of VEGFR-2- and CD31-positive vasculature at the tumor edges after discontinuation of treatment. CONCLUSION: Our findings suggest that imaging with (99m)Tc-scVEGF might be useful for monitoring VEGFR responses to antiangiogenic treatment regimens.


Subject(s)
Breast Neoplasms/metabolism , Indoles/pharmacology , Molecular Imaging/methods , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Biological Transport/drug effects , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Drug Administration Schedule , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Indoles/administration & dosage , Male , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Engineering , Pyrroles/administration & dosage , Sunitinib , Technetium , Time Factors , Tomography, Emission-Computed, Single-Photon , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Toxins (Basel) ; 2(9): 2242-57, 2010 09.
Article in English | MEDLINE | ID: mdl-22069683

ABSTRACT

SLT-VEGF is a recombinant cytotoxin comprised of Shiga-like toxin (SLT) subunit A fused to human vascular endothelial growth factor (VEGF). It is highly cytotoxic to tumor endothelial cells overexpressing VEGF receptor-2 (VEGFR-2/KDR/Flk1) and inhibits the growth of primary tumors in subcutaneous models of breast and prostate cancer and inhibits metastatic dissemination in orthotopic models of pancreatic cancer. We examined the efficacy of SLT-VEGF in limiting tumor growth and metastasis in an orthotopic melanoma model, using NCR athymic nude mice inoculated with highly metastatic Line IV Cl 1 cultured human melanoma cells. Twice weekly injections of SLT-VEGF were started when tumors became palpable at one week after intradermal injection of 1 × 10(6) cells/mouse. Despite selective depletion of VEGFR-2 overexpressing endothelial cells from the tumor vasculature, SLT-VEGF treatment did not affect tumor growth. However, after primary tumors were removed, continued SLT-VEGF treatment led to fewer tumor recurrences (p = 0.007), reduced the incidence of lung metastasis (p = 0.038), and improved survival (p = 0.002). These results suggest that SLT-VEGF is effective at the very early stages of tumor development, when selective killing of VEGFR-2 overexpressing endothelial cells can still prevent further progression. We hypothesize that SLT-VEGF could be a promising adjuvant therapy to inhibit or prevent outgrowth of metastatic foci after excision of aggressive primary melanoma lesions.


Subject(s)
Lung Neoplasms/drug therapy , Melanoma/drug therapy , Neoplasm Recurrence, Local/prevention & control , Receptors, Vascular Endothelial Growth Factor/pharmacology , Shiga Toxin/pharmacology , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Disease Progression , Humans , Longevity/drug effects , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Melanoma/mortality , Melanoma/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Recombinant Fusion Proteins/pharmacology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate , Xenograft Model Antitumor Assays
7.
Neoplasia ; 11(11): 1165-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19881952

ABSTRACT

Diverse physiological and therapeutic insults that increase the amount of unfolded or misfolded proteins in the endoplasmic reticulum (ER) induce the unfolded protein response, an evolutionarily conserved protective mechanism that manages ER stress. Glucose-regulated protein 78/immunoglobulin heavy-chain binding protein (GRP78/BiP) is an ER-resident protein that plays a central role in the ER stress response and is the only known substrate of the proteolytic A subunit (SubA) of a novel bacterial AB(5) toxin. Here, we report that an engineered fusion protein, epidermal growth factor (EGF)-SubA, combining EGF and SubA, is highly toxic to growing and confluent epidermal growth factor receptor-expressing cancer cells, and its cytotoxicity is mediated by a remarkably rapid cleavage of GRP78/BiP. Systemic delivery of EGF-SubA results in a significant inhibition of human breast and prostate tumor xenografts in mouse models. Furthermore, EGF-SubA dramatically increases the sensitivity of cancer cells to the ER stress-inducing drug thapsigargin, and vice versa, demonstrating the first example of mechanism-based synergism in the action of a cytotoxin and an ER-targeting drug.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Epidermal Growth Factor/pharmacology , Neoplasms, Experimental/drug therapy , Recombinant Fusion Proteins/pharmacology , Stress, Physiological/drug effects , Animals , Blotting, Western , Breast Neoplasms/drug therapy , Drug Synergism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/drug effects , Humans , Immunohistochemistry , Male , Mice , Microscopy, Fluorescence , Prostatic Neoplasms/drug therapy , Protein Folding/drug effects , Xenograft Model Antitumor Assays
8.
Adv Genet ; 67: 1-27, 2009.
Article in English | MEDLINE | ID: mdl-19914448

ABSTRACT

Neovascularization takes place in a large number of pathologies, including cancer. Significant effort has been invested in the development of agents that can inhibit this process, and an increasing number of such agents, known as antiangiogenic drugs, are entering clinical trials or being approved for clinical use. The key players involved in the development and maintenance of tumor neovasculature are vascular endothelial growth factor (VEGF) and its receptors (VEGFRs), and therefore VEGF/VEGFR signaling pathways have been a focus of anticancer therapies for several decades. This review focuses on two main approaches designed to selectively target VEGFRs, inhibiting VEGFR with small molecule inhibitors of receptor tyrosine kinase activity and inhibiting the binding of VEGF to VEGFRs with specific antibodies or soluble decoy VEGF receptors. The major problem with these strategies is that they appeared to be effective only in relatively small and unpredictable subsets of patients. An alternative approach would be to subvert VEGFR for intracellular delivery of cytotoxic molecules. We describe here one such molecule, SLT-VEGF, a fusion protein containing VEGF121 and the highly cytotoxic catalytic subunit of Shiga-like toxin.


Subject(s)
Blood Vessels/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction , Animals , Humans , Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
Clin Diagn Lab Immunol ; 12(7): 801-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16002626

ABSTRACT

Diagnostic peptides previously isolated from phage-displayed libraries by affinity selection with serum antibodies from patients with Lyme disease were found to give reproducible serum reactivity patterns when tested in two different enzyme-linked immunosorbent assay formats. In addition, the hypothetical possibility that peptides selected by this type of "epitope discovery" technique might identify the original antigens eliciting antibody responses was tested by searching for sequence similarities in bacterial protein databases. In support of this hypothesis, our search uncovered similarities between peptides representing two different sequence motifs and sequences in the VlsE and BBA61 antigens of Borrelia burgdorferi. Utilizing synthetic peptides, we verified that the sequence KAASKETPPALNK, located at the C terminus of the VlsE antigen, had the same reactivity pattern to sera from patients with extracutaneous Lyme disease as the diagnostic peptide SKEKPPSLNWPA, with which it shared a 7-amino-acid-residue match (consensus residues are underlined). A peptide with conservative mutations of five of the consensus residues was nonreactive, strongly suggesting that the VlsE sequence represents the epitope that originally elicited antibody responses in these patients. The diagnostic sensitivity of this new VlsE epitope was relatively low (30%) compared to that (100%) of the well-documented C6 diagnostic peptide of VlsE when tested in our small cohort of 10 patients with Lyme disease. Nonetheless, the identification of this previously unknown epitope serves as a proof of the principle of the hypothetical ability of "epitope discovery" techniques to detect specific microbial antigens with diagnostic relevance in infectious diseases.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Borrelia burgdorferi/immunology , Epitopes/immunology , Lyme Disease/immunology , Peptide Library , Amino Acid Sequence , Antigens, Bacterial/genetics , Borrelia burgdorferi/genetics , Epitopes/genetics , Humans , Lyme Disease/diagnosis , Peptides/genetics , Peptides/immunology , Sensitivity and Specificity
10.
Biochem Biophys Res Commun ; 334(1): 223-30, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-15993843

ABSTRACT

Resveratrol (trans-3,4',5-trihydroxystilbene) is a grape-derived polyphenol under intensive study for its potential in cancer prevention. In the case of cultured human melanoma cells, no one to our knowledge has investigated whether resveratrol exerts similar anti-proliferative activities in cells with different metastatic potential. Therefore, we examined the effects of this polyphenol on the growth of weakly metastatic Line IV clone 3 and on autologous, highly metastatic Line IV clone 1 cultured melanoma cells. Comparable inhibition of growth and colony formation resulted from treatment by resveratrol in both cell lines. Flow cytometric analysis revealed that resveratrol-treated clone 1 cells had a dose-dependent increase in S phase and a concomitant reduction in the G(1) phase. No detectable change in cell cycle phase distribution was found in similarly treated clone 3 cells. Western blots demonstrated a significant increase in the expression of the tumor suppressor gene p53, without a commensurate change in p21 and several other cell cycle regulatory proteins in both cell types. Chromatography of Line IV clone 3 and clone 1 cell extracts on resveratrol affinity columns revealed that the basal expression of dihydronicotinamide riboside quinone reductase 2 (NQO2) was higher in Line IV clone 1 than clone 3 cells. Levels of NQO2 but not its structural analog NQO1 were dose-dependently increased by resveratrol in both cell lines. We propose that induction of NQO2 may relate to the observed increased expression of p53 that, in turn, contributes to the observed suppression of cell growth in both melanoma cell lines.


Subject(s)
Melanoma/enzymology , Melanoma/pathology , Quinone Reductases/metabolism , Stilbenes/administration & dosage , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Resveratrol , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...