Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Am Acad Dermatol ; 86(4): 846-853, 2022 04.
Article in English | MEDLINE | ID: mdl-34808324

ABSTRACT

BACKGROUND: There is a need to improve prognostic accuracy for patients with cutaneous melanoma. A 31-gene expression profile (31-GEP) test uses the molecular biology of primary tumors to identify individual patient metastatic risk. OBJECTIVE: Develop a nomogram incorporating 31-GEP with relevant clinical factors to improve prognostic accuracy. METHODS: In an IRB-approved study, 1124 patients from 9 Mohs micrographic surgery centers were prospectively enrolled, treated with Mohs micrographic surgery, and underwent 31-GEP testing. Data from 684 of those patients with at least 1-year follow-up or a metastatic event were included in nomogram development to predict metastatic risk. RESULTS: Logistic regression modeling of 31-GEP results and T stage provided the simplest nomogram with the lowest Bayesian information criteria score. Validation in an archival cohort (n = 901) demonstrated a significant linear correlation between observed and nomogram-predicted risk of metastasis. The resulting nomogram more accurately predicts the risk for cutaneous melanoma metastasis than T stage or 31-GEP alone. LIMITATIONS: The patient population is representative of Mohs micrographic surgery centers. Sentinel lymph node biopsy was not performed for most patients and could not be used in the nomogram. CONCLUSIONS: Integration of 31-GEP and T stage can gain clinically useful prognostic information from data obtained noninvasively.


Subject(s)
Melanoma , Skin Neoplasms , Bayes Theorem , Gene Expression Profiling/methods , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/surgery , Mohs Surgery , Nomograms , Prognosis , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Melanoma, Cutaneous Malignant
2.
J Am Acad Dermatol ; 82(1): 139-148, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31279037

ABSTRACT

BACKGROUND: Outcomes for patients with cutaneous squamous cell carcinoma (CSCC) treated with Mohs micrographic surgery (MS) in the United States have never been prospectively defined. Risk factors as they relate to outcomes are primarily derived from single-institution, retrospective data without regard for treatment modality. The American Joint Committee on Cancer Staging Manual, Eighth Edition, and the Brigham and Women's Hospital T staging systems have not been prospectively validated. OBJECTIVE: To prospectively quantify outcomes by T stage and verify historically high-risk features as they pertain to outcomes in MS-treated CSCC. METHODS: A 5-year, prospective, multicenter analysis of patients undergoing MS for invasive CSCC was conducted. RESULTS: The study enrolled 647 patients with 745 tumors. The 5-year local recurrence (LR)-free survival, nodal metastasis (NM)-free survival, and disease-specific survival were 99.3%, 99.2%, and 99.4%, respectively. Both staging systems were predictive of NM, disease-specific death, and all-cause death; neither was predictive of LR. Although Breslow depth was statistically associated with LR, NM, and disease-specific death, incidental perineural invasion was not. LIMITATIONS: The Brigham and Women's Hospital and the American Joint Committee on Cancer Staging Manual, Eighth Edition T staging systems were published after study enrollment, therefore T stages were retrospectively applied using the prospectively collected data. CONCLUSION: MS is a highly effective treatment for CSCC and may mitigate factors typically considered high risk. Uniform reporting of Breslow depth should be considered in CSCC. The American Joint Committee on Cancer Staging Manual, Eighth Edition, and the Brigham and Women's Hospital staging system are useful prognosticators but are not predictive of LR after MS.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Mohs Surgery , Neoplasm Recurrence, Local/pathology , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prospective Studies , Risk Factors , Survival Rate , Time Factors
3.
J Am Acad Dermatol ; 70(4): 630-636, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24433872

ABSTRACT

BACKGROUND: The prognosis and management of incidental perineural invasion (PNI) in patients with cutaneous squamous cell carcinoma (CSCC) has not been well defined. OBJECTIVE: We sought to investigate the clinical, histologic, and treatment characteristics associated with incidental PNI, histologic PNI extending beyond the tumor bulk, in patients with CSCC. METHODS: We conducted a multicenter prospective analysis of patients with CSCC undergoing Mohs micrographic surgery. RESULTS: The incidence of PNI was 4.6% in 753 CSCC cases. PNI was significantly associated with tumors of the head and neck (P = .039), larger tumor diameter (P < .001), presence of clinically palpable lymphadenopathy (P = .012), and recurrent (P < .001) and painful (P < .001) tumors. Further, PNI was significantly associated with poor tumor differentiation (P < .001), greater tumor thickness (P < .001), a greater number of Mohs stages (P < .001), and larger estimated maximum Mohs margin (P < .001) required to clear the tumor. LIMITATIONS: The low numbers of patients demonstrating incidental PNI limits this study. CONCLUSIONS: The association of incidental PNI with clinicopathological indicators of poor prognosis suggests that incidental PNI may serve as a marker to improve the precision in the prognostic assessment of patients with CSCC.


Subject(s)
Carcinoma, Squamous Cell/surgery , Neoplasm Recurrence, Local/pathology , Peripheral Nervous System Neoplasms/secondary , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Aged , Aged, 80 and over , Biopsy, Needle , Carcinoma, Squamous Cell/secondary , Female , Follow-Up Studies , Humans , Immunohistochemistry , Incidental Findings , Male , Middle Aged , Mohs Surgery/methods , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Peripheral Nervous System Neoplasms/pathology , Peripheral Nervous System Neoplasms/surgery , Prospective Studies , Risk Assessment , Treatment Outcome
4.
J Am Acad Dermatol ; 66(6): 911-6, 916.e1-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22445792

ABSTRACT

BACKGROUND: Although criteria are established for the histologic diagnosis of atypical nevi (AN), consensus about the criteria in the diagnosis of and in the definition of AN is limited. Moreover, intraobserver and interobserver differences in the application of these criteria for the diagnosis of AN have been observed. OBJECTIVE: We sought to determine the usefulness of HLA antigen expression as a biomarker of AN. METHODS: The immunoperoxidase reaction was used to mark common nevi and AN with HLA class I heavy chain-, ß2microglobulin (ß2m)-, and HLA class II ß chain-specific monoclonal antibodies. RESULTS: HLA class I heavy chain, ß2m, and HLA class II ß chain were expressed in 5 (8.6%) of the 58 common nevi and in 46 (∼72%) of the 64 atypical melanocytic lesions. Among common lesions, only halo nevi expressed HLA class I heavy chain, ß2m, and HLA class II ß chain. The level of HLA class I heavy chain ß2m and of HLA class II ß chain expression correlated with the degree of cytologic atypia and architectural disorder. LIMITATIONS: The number of lesions tested and the subjective nature of the analysis of immunohistochemical staining of tissue sections are both limitations. CONCLUSIONS: The data presented suggest that HLA antigen expression is an objective biomarker that correlates well with the degree of cytologic atypia in AN and may: (1) be useful to distinguish common nevi from AN, and (2) represent a more objective measure to determine which AN should be excised.


Subject(s)
HLA Antigens/metabolism , Melanocytes/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal , Child , Dysplastic Nevus Syndrome/metabolism , Female , Histocompatibility Antigens Class I/metabolism , Humans , Immunoenzyme Techniques , Immunohistochemistry , Male , Middle Aged , Nevus/metabolism , Nevus, Halo/metabolism , Young Adult
5.
Semin Immunopathol ; 33(4): 321-34, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21523560

ABSTRACT

Malignant transformation of cells is often associated with changes in classical and non-classical HLA class I antigen, HLA class II antigen as well as NK cell activating ligand (NKCAL) expression. These changes are believed to play a role in the clinical course of the disease since these molecules are critical to the interactions between tumor cells and components of both innate and adaptive immune system. For some time, it has been assumed that alterations in the expression profile of HLA antigens and NKCAL on malignant cells represented loss of classical HLA class I antigen and induction of HLA class II antigen, non-classical HLA class I antigen and/or NKCAL expression. In contrast to these assumptions, experimental evidence suggests that in some cases dysplastic and malignant cells can acquire classical HLA class I antigen expression and/or lose the ability to express HLA class II antigens. In light of the latter findings as well as of the revival of the cancer immune surveillance theory, a reevaluation of the interpretation of changes in HLA antigen and NKCAL expression in malignant lesions is warranted. In this article, we first briefly describe the conventional types of changes in HLA antigen and NKCAL expression that have been identified in malignant cells to date. Second, we discuss the evidence indicating that, in at least some cell types, classical HLA class I antigen expression can be acquired and/or the ability to express HLA class II antigens is lost. Third, we review the available evidence for the role of immune selective pressure in the generation of malignant lesions with changes in HLA antigen expression. This information contributes to our understanding of the role of the immune system in the control of tumor development and to the optimization of the design of immunotherapeutic strategies for the treatment of cancer.


Subject(s)
Antigens, CD/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Neoplasms/immunology , Receptors, Immunologic/immunology , Animals , Antigens, CD/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Neoplasms/metabolism , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family , Tumor Escape
6.
Adv Cancer Res ; 109: 73-121, 2010.
Article in English | MEDLINE | ID: mdl-21070915

ABSTRACT

The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the development of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma. For many years, T cell-based immunotherapy has been emphasized in part because of the disappointing results of the monoclonal antibody (mAb)-based clinical trials conducted in the early 1980s and in part because of the postulated major role played by T cells in tumor growth control. More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases. As a result, there has been increased interest in identifying and characterizing antibody-defined melanoma antigens. Among them, the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA) or melanoma chondroitin sulfate proteoglycan (MCSP), has attracted much attention in recent years because of the growing experimental evidence that it fulfills two requirements for immunotherapy to be therapeutically effective: (1) targeting of cancer stem cells (CSC) and (2) development of combinatorial therapies to counteract the escape mechanisms driven by the genetic instability of tumor cells. With this in mind, in this chapter, we have reviewed recent information related to the distribution of CSPG4 on various types of tumors, including CSC, its expression on pericytes in the tumor microenvironment, its recognition by T cells, its role in cell biology as well as the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.


Subject(s)
Antigens/physiology , Neoplasms/genetics , Neoplasms/therapy , Proteoglycans/physiology , Animals , Antigens/chemistry , Antigens/genetics , Antigens/metabolism , Cell Movement/genetics , Cell Movement/physiology , Evolution, Molecular , Humans , Immunotherapy/methods , Molecular Targeted Therapy/methods , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Phylogeny , Proteoglycans/chemistry , Proteoglycans/genetics , Proteoglycans/metabolism , Tissue Distribution
7.
Clin Cancer Res ; 16(1): 11-20, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028761

ABSTRACT

A few tumor antigen (TA)-specific monoclonal antibodies (mAb) have been approved by the Food and Drug Administration for the treatment of several major malignant diseases and are commercially available. Once in the clinic, mAbs have an average success rate of approximately 30% and are well tolerated. These results have changed the face of cancer therapy, bringing us closer to more specific and more effective biological therapy of cancer. The challenge facing tumor immunologists at present is represented by the identification of the mechanism(s) underlying the patients' differential clinical response to mAb-based immunotherapy. This information is expected to lead to the development of criteria to select patients to be treated with mAb-based immunotherapy. In the past, in vitro and in vivo evidence has shown that TA-specific mAbs can mediate their therapeutic effect by inducing tumor cell apoptosis, inhibiting the targeted antigen function, blocking tumor cell signaling, and/or mediating complement- or cell-dependent lysis of tumor cells. More recent evidence suggests that TA-specific mAb can induce TA-specific cytotoxic T-cell responses by enhancing TA uptake by dendritic cells and cross-priming of T cells. In this review, we briefly summarize the TA-specific mAbs that have received Food and Drug Administration approval. Next, we review the potential mechanisms underlying the therapeutic efficacy of TA-specific mAbs with emphasis on the induction of TA-specific cellular immune responses and their potential to contribute to the clinical efficacy of TA-specific mAb-based immunotherapy. Lastly, we discuss the potential negative effect of immune escape mechanisms on the clinical efficacy of TA-specific mAb-based immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Neoplasms/drug therapy , Drug Resistance, Neoplasm , Humans , T-Lymphocytes, Helper-Inducer/immunology
8.
Clin Cancer Res ; 11(23): 8304-11, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322289

ABSTRACT

PURPOSE: To determine the frequency of abnormalities in human leukocyte antigen (HLA) and antigen processing machinery (APM) component expression in malignant brain tumors. This information may contribute to our understanding of the immune escape mechanisms used by malignant brain tumors because HLA antigens mediate interactions of tumor cells with the host's immune system. EXPERIMENTAL DESIGN: Eighty-eight surgically removed malignant astrocytic tumors, classified according to the WHO criteria, were stained in immunoperoxidase reactions with monoclonal antibody recognizing monomorphic, locus-specific, and allospecific determinants of HLA class I antigens, beta2-microglobulin, APM components (LMP2, LMP7, TAP1, TAP2, calnexin, calreticulin, and tapasin), and HLA class II antigens. RESULTS: HLA class I antigens were lost in approximately 50% of the 47 glioblastoma multiforme (GBM) lesions and in approximately 20% of the 18 grade 2 astrocytoma lesions stained. Selective HLA-A2 antigen loss was observed in approximately 80% of the 24 GBM lesions and in approximately 50% of the 12 grade 2 astrocytoma lesions stained. HLA class I antigen loss was significantly (P < 0.025) correlated with tumor grade. Among the APM components investigated, tapasin expression was down-regulated in approximately 20% of the GBM lesions analyzed; it was associated, although not significantly, with HLA class I antigen down-regulation and tumor grade. HLA class II antigen expression was detected in approximately 30% of the 44 lesions analyzed. CONCLUSION: The presence of HLA antigen defects in malignant brain tumors may provide an explanation for the relatively poor clinical response rates observed in the majority of the T cell-based immunotherapy clinical trials conducted to date in patients with malignant brain tumors.


Subject(s)
Antiporters/metabolism , Astrocytoma/metabolism , Brain Neoplasms/metabolism , HLA-A2 Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoglobulins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/metabolism , Antibodies, Monoclonal , Antigen Presentation , Astrocytoma/pathology , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Calnexin/metabolism , Calreticulin/metabolism , Cysteine Endopeptidases/metabolism , Down-Regulation , Humans , Membrane Transport Proteins , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , beta 2-Microglobulin/metabolism
9.
Cancer Treat Res ; 123: 61-88, 2005.
Article in English | MEDLINE | ID: mdl-16211866

ABSTRACT

The results from in vitro immunological experiments, murine tumor models and patients with cancer clearly demonstrate that tumors have multiple mechanisms to evade the immune response. During the early stages of tumor development malignant cells can be poor stimulators, present poor targets or become resistant to the innate immune response, while at later stages, progressively growing tumors impair the adaptive immune response by blocking the maturation and function of antigen presenting cells and causing alterations in T cell signal transduction and function. Preliminary results also suggest a correlation between some of these changes and an increased metastatic potential of the tumor cells, a diminished response to immunotherapy, and poor prognosis. Carefully coordinated basic research studies and clinical immunotherapy trials will be required to fully determine the impact on the outcome of the disease and the response to treatment. However, understanding the mechanisms used by tumor cells to evade the immune system could result in new therapeutic approaches for preventing and/or reversing these immune alterations and have the potential of improving the current results of immunotherapy trials.


Subject(s)
Neoplasms/immunology , Tumor Escape , Animals , Humans
10.
J Immunol Methods ; 299(1-2): 139-51, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15896802

ABSTRACT

Abnormalities in HLA class I antigen expression are frequently found in malignant tumors. Their potential role in the clinical course of the disease and in the outcome of T cell-based immunotherapy has stimulated interest in the characterization of the molecular mechanisms underlying HLA class I antigen abnormalities in malignant cells. Multiple mechanisms have been identified. Among them are abnormalities in antigen processing machinery (APM) component expression. In spite of this information, APM component expression in malignant lesions has been investigated only to a limited extent because of the lack of availability, for most APM components, of monoclonal antibodies (mAb) which stain formalin-fixed, paraffin-embedded tissues. The latter are the substrate of choice in immunohistochemical (IHC) reactions. To overcome this limitation, we have developed a simple and reproducible method to generate APM component-specific mAb which stain formalin-fixed, paraffin-embedded tissue sections. This method involves five steps: (i) immunogenic amino acid sequences, which display low homology with their mouse counterparts when possible, are identified in APM components and utilized to synthesize peptides; (ii) BALB/c mice are immunized with keyhole limpet hemocyanin (KLH)-conjugated synthetic peptides and with sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE)-purified recombinant APM component proteins; (iii) immunized mice, which develop high titer APM component-specific antibodies, are utilized to generate hybridomas which are screened for APM component-specific antibody production by Western blotting assays, with lymphoid cell lysates; (iv) identified APM component-specific mAb are characterized in their specificity and in their reactivity with permeabilized cells in ELISA and/or flow cytometry; and (v) mAb, with the appropriate reactivity pattern, are tested in IHC reactions with formalin-fixed, paraffin-embedded tissue sections. The use of the methodology we have developed resulted in the generation of a panel of APM component-specific mAb capable of staining formalin-fixed, paraffin-embedded tissue sections in IHC reactions. These reagents will facilitate the analysis of APM component expression in tissues under physiological and pathological conditions. In addition, the methodology we have described is likely to be applicable to other antigenic systems to develop mAb capable of detecting protein components of interest in formalin-fixed, paraffin-embedded tissue sections.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/analysis , Immunohistochemistry/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/isolation & purification , Antibody Specificity , Antigens, Neoplasm/immunology , Fixatives , Formaldehyde/chemistry , Humans , Hybridomas/immunology , Mice , Molecular Sequence Data , Paraffin Embedding
11.
Adv Cancer Res ; 93: 189-234, 2005.
Article in English | MEDLINE | ID: mdl-15797448

ABSTRACT

Changes in classical and nonclassical HLA class I antigen and NK cell-activating ligand expression have been identified in malignant lesions. These changes, which are described in this chapter, are believed to play a major role in the clinical course of the disease since both HLA class I antigens and NK cell-activating ligands are critical to the interaction between tumor cells and components of both innate and adaptive immune systems. Nevertheless, there is still debate in the literature about the biologic and functional significance of HLA class I antigen and NK cell-activating ligand abnormalities in malignant lesions. The reasons for this debate are reviewed. They include (i) the incomplete association between classical HLA class I antigen changes and the clinical course of the disease; (ii) the relatively limited number of malignant lesions that have been analyzed for nonclassical HLA class I antigen and NK cell-activating ligand expression; and (iii) the conflicting data regarding the role of immunoselection in the generation of malignant cells with HLA antigen and NK cell-activating ligand abnormalities. The technical limitations associated with the assessment of HLA antigen and NK cell-activating ligand expression in malignant lesions as well as the immunological and nonimmunological variables that may confound the impact of HLA antigen and NK cell-activating ligand changes on the clinical course of the disease are also discussed. Future studies aimed at overcoming these limitations and characterizing these variables are expected to provide a solution to the current debate regarding the significance of HLA class I antigen and NK cell-activating ligand abnormalities in malignant lesions.


Subject(s)
Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms/immunology , Antigens, Neoplasm/immunology , Ligands
12.
J Immunol ; 174(3): 1462-71, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661905

ABSTRACT

Scanty information is available about the mechanisms underlying HLA class I Ag abnormalities in malignant cells exposed to strong T cell-mediated selective pressure. In this study, we have characterized the molecular defects underlying HLA class I Ag loss in five melanoma cell lines derived from recurrent metastases following initial clinical responses to T cell-based immunotherapy. Point mutations in the translation initiation codon (ATG-->ATA) and in codon 31 (TCA-->TGA) of the beta(2)-microglobulin (beta(2)m) gene were identified in the melanoma cell lines 1074MEL and 1174MEL, respectively. A hot-spot CT dinucleotide deletion within codon 13-15 was found in the melanoma cell lines 1106MEL, 1180MEL, and 1259MEL. Reconstitution of beta(2)m expression restored HLA class I Ag expression in the five melanoma cell lines; however, the HLA-A and HLA-B,-C gene products were differentially expressed by 1074MEL, 1106MEL, and 1259MEL cells. In addition, in 1259MEL cells, the Ag-processing machinery components calnexin, calreticulin, and low m.w. polypeptide 10 are down-regulated, and HLA-A2 Ags are selectively lost because of a single cytosine deletion in the HLA-A2 gene exon 4. Our results in conjunction with those in the literature suggest the emergence of a preferential beta(2)m gene mutation in melanoma cells following strong T cell-mediated immune selection. Furthermore, the presence of multiple HLA class I Ag defects within a tumor cell population may reflect the accumulation of multiple escape mechanisms developed by melanoma cells to avoid distinct sequential T cell-mediated selective events.


Subject(s)
Antigen Presentation/genetics , Down-Regulation/genetics , HLA-A2 Antigen/genetics , Immunotherapy , Melanoma/genetics , Melanoma/immunology , Point Mutation , beta 2-Microglobulin/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Transformed , Cell Line, Tumor , DNA, Complementary/genetics , Down-Regulation/immunology , Epitopes/biosynthesis , Epitopes/genetics , Gene Expression Regulation, Neoplastic , HLA-A Antigens/biosynthesis , HLA-A Antigens/genetics , HLA-A2 Antigen/biosynthesis , HLA-B Antigens/biosynthesis , HLA-B Antigens/genetics , HLA-C Antigens/biosynthesis , HLA-C Antigens/genetics , Humans , Loss of Heterozygosity , Melanoma/secondary , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Protein Subunits/biosynthesis , Protein Subunits/genetics , Transfection , Tumor Escape/genetics , Tumor Escape/immunology , beta 2-Microglobulin/isolation & purification
13.
Crit Rev Immunol ; 24(4): 267-96, 2004.
Article in English | MEDLINE | ID: mdl-15588226

ABSTRACT

The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the application of novel strategies for the treatment of patients with malignant melanoma. Because of its expression in a large percentage of melanoma lesions and its restricted distribution in normal tissues, the high molecular weight-melanoma-associated antigen (HMW-MAA), also known as the melanoma chondroitin sulfate proteoglycan (MCSP), has been used to implement immunotherapy of melanoma. The potential clinical relevance of HMW-MAA/MCSP has stimulated investigations to characterize its structural properties and biological function in melanoma cells. Over the last 10 years, the field of HMW-MAA/MCSP research has seen tremendous growth. Specifically, a significant amount of information has been accumulated regarding (1) the structural characteristics of the HMW-MAA/MCSP, (2) its role in the biology of melanoma cells, and (3) the potential molecular mechanisms underlying the association between HMW-MAA/MCSP-specific immunity and survival prolongation in melanoma patients immunized with HMW-MAA/MCSP mimics. In this review, we summarize the characteristics of the HMW-MAA/MCSP in terms of its structure, antigenic profile, tissue distribution, and similarities with its counterparts in other animal species. Additionally, we discuss the role the HMW-MAA/MCSP plays in melanoma cell biology with emphasis on the recently identified signal transduction pathways triggered by the HMW-MAA/MCSP. Finally, we discuss the potential molecular mechanisms underlying the beneficial effect of anti-HMW-MAA/MCSP antibodies on the clinical course of the disease in patients with melanoma.


Subject(s)
Antigens, Neoplasm/physiology , Chondroitin Sulfate Proteoglycans/physiology , Animals , Antigens, Neoplasm/biosynthesis , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Chondroitin Sulfate Proteoglycans/biosynthesis , Humans , Melanoma/metabolism , Melanoma/pathology , Melanoma/therapy , Models, Biological , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Signal Transduction/physiology
14.
J Immunol Methods ; 294(1-2): 23-35, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15604013

ABSTRACT

The phage display Ab library technology has been found to be a useful method to isolate antigen-specific Ab fragments, since the repertoire of antibody specificities is broad and since it bypasses the need of immunization. However, when screening clones isolated from a phage display Ab library, the yield of isolating antigen-specific Ab fragments is low and the rate of false negative results is high. This limitation reflects the low affinity/avidity of Ab fragments and/or the low density of the target antigen. To facilitate the isolation of Ab fragments with a broad range of affinities to antigens of interest from phage display Ab libraries, we have developed a simple method to increase the sensitivity of binding assays to detect the reactivity of single-chain fragments of antibody variable regions (scFv) with target antigens. This method involves the mixing of scFv fragments, expressing a c-myc epitope tag, with anti-tag mAb 9E10 prior to their use in binding assays in order to form stable dimeric Ab fragment-anti-tag mAb complexes. The increase in the reactivity of scFv fragments with the corresponding antigen is observed over a broad range of scFv fragment (6-800 microg/ml) and mAb 9E10 (0.5-30 microg/ml) concentrations, thereby facilitating the testing of scFv fragment preparations with unknown scFv fragment concentrations. Use of this method in binding assays resulted in a twofold increase in the reactivity of low-affinity purified scFv fragments with the corresponding antigen. Moreover, application of this method to screen clones isolated from phage display scFv libraries resulted in a reproducible increase in both the yield of antigen-specific scFv clones and the titer of scFv fragment preparations by a factor of 5 and 2- to 32-fold, respectively. Lastly, this method can be applied in both ELISA and flow cytometry and is independent of the characteristics of the antigen (i.e. whole cells, carbohydrates and purified protein) and/or of the library (synthetic scFv Library (#1), a large semi-synthetic phage display scFv library and the human synthetic VH+VL scFv library (Griffin.1 library)) used. Therefore, the method we have described represents a sensitive, simple and reproducible technique that will facilitate the isolation and use of scFv fragments.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Affinity/genetics , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibody Affinity/immunology , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Cloning, Molecular/methods , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/genetics , Epitopes/immunology , Gene Library , Humans , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/metabolism , Mice , Protein Binding/genetics , Protein Binding/immunology , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Reproducibility of Results , Sensitivity and Specificity
15.
Curr Opin Immunol ; 16(5): 644-50, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15342012

ABSTRACT

HLA class I antigen defects are frequently found in malignant cells. They appear to play a role in the clinical course of the disease, probably because they provide tumor cells with a mechanism to escape cytotoxic T lymphocyte (CTL) recognition and destruction. Expression of HLA class I antigens, however, is not always associated with the susceptibility of tumor cells to CTL lysis. Many mechanisms may underlie this finding, including the lack of tumor antigen (TA)-derived peptide presentation by a given HLA class I allospecificity, and/or the expression of immunosuppressive molecules such as HLA-G. These findings emphasize the need to develop probes to measure HLA class I allospecificity-TA peptide complex expression in malignant cells. Furthermore, the evaluation of the role of HLA class I antigens in the interaction of malignant cells with host immune cells should take into account the potential interference of tumor-derived immunomodulators.


Subject(s)
Cell Communication/immunology , Histocompatibility Antigens Class I/biosynthesis , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Histocompatibility Antigens Class I/immunology , Humans
16.
Expert Rev Vaccines ; 3(2): 171-87, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15056043

ABSTRACT

The lack of effective treatment for advanced stage melanoma by conventional therapies, such as radiation and chemotherapy, has highlighted the need to develop alternative therapeutic strategies. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma and the availability of well-characterized melanoma antigens to target melanoma lesions with immunological effector mechanisms. In recent years, T-cell-based immunotherapy has been emphasized, in part because of the disappointing results of the antibody-based trials conducted in the early 1980s, and in part because of the postulated major role played by T-cells in tumor growth control. In this review, the characteristics of antibody and T-cell-defined melanoma antigens will first be described, with emphasis on those used in clinical trials. Following a review of the current immunization and immunomonitoring strategies, the results from the T-cell-based immunotherapy clinical trials conducted to date will be reviewed.


Subject(s)
Immunotherapy , Melanoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Humans , Melanoma/immunology , Randomized Controlled Trials as Topic , Vaccines, Subunit/therapeutic use
17.
Ann N Y Acad Sci ; 1028: 340-50, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15650259

ABSTRACT

Induction of humoral anti-human high molecular weight melanoma-associated antigen (anti-HMW-MAA) immunity following active specific immunotherapy is associated with a statistically significant prolongation of survival in patients with melanoma. This association does not appear to be mediated by immunological mechanisms because anti-HMW-MAA antibodies are poor mediators of complement- and cell-mediated cytotoxicity of melanoma cells. Therefore, we have been investigating nonimmunological mechanisms by which anti-HMW-MAA antibodies (Abs) affect the biology of melanoma cells. We have demonstrated that anti-HMW-MAA mAbs interfere with the interaction of HMW-MAA with extracellular matrix (ECM) components, a process known to be crucial in the early phase of melanoma metastasis. Furthermore, anti-HMW-MAA mAbs appear to block the series of signal transduction events triggered by the interaction of HMW-MAA with ECM. They include the activation of the family of Rho GTPases, p130cas, and focal adhesion kinase (FAK). These findings parallel the inhibition of the rat homologue of HMW-MAA NG2 function by anti-NG2 antibodies. Taken together, all these results provide a mechanistic explanation not only for the therapeutic effect of anti-HMW-MAA antibodies in the treatment of melanoma, but also for the function of HMW-MAA in the biology of melanoma cells. This information is expected to serve as a useful background to design effective HMW-MAA-targeted immunotherapy in patients with melanoma.


Subject(s)
Antigens, Neoplasm/chemistry , Immunotherapy/methods , Melanoma/immunology , Melanoma/therapy , Animals , Cell Movement , Crk-Associated Substrate Protein , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Melanoma/metabolism , Models, Biological , Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Retinoblastoma-Like Protein p130 , Signal Transduction , Time Factors
19.
Keio J Med ; 52(4): 220-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14748474

ABSTRACT

Depending on the tumor types, HLA class I antigen downregulation or loss has been found in 16% to 50% of malignant lesions in many malignancies with a clinical association with histopathological markers of poor prognosis of the disease and with reduced free interval and survival. These findings may reflect the escape of tumor cells with HLA class I abnormalities from recognition and destruction by HLA class I-restricted, tumor-associated antigen-specific cytotoxic T lymphocytes. This possibility has stimulated investigations on the mechanisms underlying HLA class I antigen abnormalities in malignant cells. Distinct molecular defects underlying an abnormal HLA class I phenotype have been identified and characterized. These defects range from structural alterations of the genes which encode HLA class I antigen subunits to deregulation of antigen processing machinery components responsible for a functional HLA class I antigen expression. These findings, in conjunction with those of clinical recurrence of lesions with HLA class I antigen loss following T cell-based immunotherapy in patients, suggests that immunoselection may play a role in the generation of malignant lesions with HLA class I antigen abnormalities. This possibility has stressed the need to effectively monitor functional HLA class I antigen expression in malignant lesions in the application of T cell-based immunotherapy as well as to develop strategies to circumvent the negative impact of immunoselection.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class I/immunology , Neoplasms/genetics , Neoplasms/immunology , Humans
20.
Vaccine ; 20 Suppl 4: A40-5, 2002 Dec 19.
Article in English | MEDLINE | ID: mdl-12477427

ABSTRACT

Poor clinical response rates have been observed in the majority of the T cell-based immunotherapy clinical trials conducted to date. One reason might be the presence of abnormalities in HLA class I antigen presentation in malignant lesions. An increased frequency of HLA class I abnormalities has been observed in malignant lesions from patients treated with T cell-based immunotherapy and in lesions which have recurred in patients who had experienced clinical responses following T cell-based immunotherapy. These observations are compatible with the possibility that the outgrowth of a patient's tumor reflects immune selection of tumor cells which have acquired escape mechanisms from immune recognition.


Subject(s)
Histocompatibility Antigens Class I/immunology , Neoplasms/immunology , Histocompatibility Antigens Class I/genetics , Humans , Immunotherapy , Mutation , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL