Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
5.
J Eur Acad Dermatol Venereol ; 35(1): 105-115, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32455474

ABSTRACT

BACKGROUND: The survival in metastatic melanoma has dramatically improved after the introduction of immune checkpoint- (ICIs) and MAPKinase inhibitors (MAPKis). OBJECTIVE: Our aim was to describe therapy response and survival in a real-world population as well as to assess the associations between clinical variables and therapy outcome for patients with metastatic melanoma receiving first-line ICIs or MAPKis. METHODS: A total of 252 patients with metastatic (stage IV) melanoma were prospectively followed between 1 January 2010 and 3 December 2017 with follow-up until 31 March 2019, at the Karolinska University Hospital, Sweden. Hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS) were analysed with Cox regression, and logistic regression was used to estimate odds ratios (ORs) for therapy response. RESULTS: Patients receiving ICIs (n = 138) experienced longer PFS compared to patients that received MAPKis (n = 114; median PFS for ICIs was 6.8 months, and median PFS for MAPKis was 5.3 months). In the multivariable analyses of clinical markers, increasing M-stage (OR 0.65; 95% CI 0.45-0.94; P = 0.022) and male sex (OR 0.41; 95% CI 0.19-0.90; P = 0.027) were significantly associated with lower response to ICIs. Lower baseline albumin levels (OR 0.90; 95% CI 0.83-0.98; P = 0.019) and male sex (OR 0.33; 95% CI 0.12-0.93; P = 0.036) were related with lower response to MAPKis. For ICIs, increasing M-stage (HR 1.34; 95% CI 1.07-1.68; P = 0.010), increasing LDH (HR 1.73; 95% CI 1.19-2.50; P = 0.004) and decreasing albumin (HR 1.06; 95% CI 1.01-1.10; P = 0.011) were significantly associated lower PFS in the adjusted model. The corresponding markers for MAPKis were increasing LDH (HR 1.44; 95% CI 1.08-1.92; P = 0.013) and decreasing albumin (HR 1.05; 95% CI 1.02-1.09; P = 0.005) for PFS. CONCLUSION: ICIs and MAPKis were effective in this real-world population, and we could confirm the importance of previously reported clinical prognostic markers. Albumin values may be associated with therapy outcome but need further validation.


Subject(s)
Melanoma , Biomarkers , Humans , Male , Melanoma/drug therapy , Prognosis , Sweden , Treatment Outcome
6.
Oncogene ; 36(41): 5734-5744, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28581517

ABSTRACT

With very similar 3D structures, the widely expressed ß-arrestin isoforms 1 and 2 play at times identical, distinct or even opposing roles in regulating various aspects of G protein-coupled receptors (GPCR) expression and signalling. Recent evidence recognizes the ß-arrestin system as a key regulator of not only GPCRs, but also receptor tyrosine kinases, including the highly cancer relevant insulin-like growth factor type 1 receptor (IGF-1R). Binding of ß-arrestin1 to IGF-1R leads to ligand-dependent degradation of the receptor and generates additional MAPK/ERK signalling, protecting cancer cells against anti-IGF-1R therapy. Because the interplay between ß-arrestin isoforms governs the biological effects for most GPCRs, as yet unexplored for the IGF-1R, we sought to investigate specifically the regulatory roles of the ß-arrestin2 isoform on expression and function of the IGF-1R. Results from controlled expression of either ß-arrestin isoform demonstrate that ß-arrestin2 acts in an opposite manner to ß-arrestin1 by promoting degradation of an unstimulated IGF-1R, but protecting the receptor against agonist-induced degradation. Although both isoforms co-immunoprecipitate with IGF-1R, the ligand-occupied receptor has greater affinity for ß-arrestin1; this association lasts longer, sustains MAPK/ERK signalling and mitigates p53 activation. Conversely, ß-arrestin2 has greater affinity for the ligand-unoccupied receptor; this interaction is transient, triggers receptor ubiquitination and degradation without signalling activation, and leads to a lack of responsiveness to IGF-1, cell cycle arrest and decreased viability of cancer cells. This study reveals contrasting abilities of IGF-1R to interact with each ß-arrestin isoform, depending on the presence of the ligand and demonstrates the antagonism between the two ß-arrestin isoforms in controlling IGF-1R expression and function, which could be developed into a practical anti-IGF-1R strategy for cancer therapy.


Subject(s)
Neoplasms/genetics , Receptor, IGF Type 1/genetics , beta-Arrestins/genetics , Animals , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Receptor, IGF Type 1/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , beta-Arrestins/metabolism
7.
Oncogene ; 36(23): 3274-3286, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28092675

ABSTRACT

Melanoma tumors usually retain wild-type p53; however, its tumor-suppressor activity is functionally disabled, most commonly through an inactivating interaction with mouse double-minute 2 homolog (Mdm2), indicating p53 release from this complex as a potential therapeutic approach. P53 and the tumor-promoter insulin-like growth factor type 1 receptor (IGF-1R) compete as substrates for the E3 ubiquitin ligase Mdm2, making their relative abundance intricately linked. Hence we investigated the effects of pharmacological Mdm2 release from the Mdm2/p53 complex on the expression and function of the IGF-1R. Nutlin-3 treatment increased IGF-1R/Mdm2 association with enhanced IGF-1R ubiquitination and a dual functional outcome: receptor downregulation and selective downstream signaling activation confined to the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. This Nutlin-3 functional selectivity translated into IGF-1-mediated bioactivities with biphasic effects on the proliferative and metastatic phenotype: an early increase and late decrease in the number of proliferative and migratory cells, while the invasiveness was completely inhibited following Nutlin-3 treatment through an impaired IGF-1-mediated matrix metalloproteinases type 2 activation mechanism. Taken together, these experiments reveal the biased agonistic properties of Nutlin-3 for the mitogen-activated protein kinase pathway, mediated by Mdm2 through IGF-1R ubiquitination and provide fundamental insights into destabilizing p53/Mdm2/IGF-1R circuitry that could be developed for therapeutic gain.


Subject(s)
Biomarkers, Tumor/metabolism , Insulin-Like Growth Factor I/metabolism , Melanoma/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Receptor, IGF Type 1/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Proliferation , Humans , Melanoma/genetics , Melanoma/metabolism , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Proteins c-mdm2/genetics , Receptor, IGF Type 1/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
8.
Prog Mol Biol Transl Sci ; 141: 277-311, 2016.
Article in English | MEDLINE | ID: mdl-27378760

ABSTRACT

Cell-surface receptors govern the critical information passage from outside to inside the cell and hence control important cellular decisions such as survival, growth, and differentiation. These receptors, structurally grouped into different families, utilize common intracellular signaling-proteins and pathways, yet promote divergent biological consequences. In rapid processing of extracellular signals to biological outcomes, posttranslational modifications offer a repertoire of protein processing options. Protein ubiquitination was originally identified as a signal for protein degradation through the proteasome system. It is now becoming increasingly recognized that both ubiquitin and ubiquitin-like proteins, all evolved from a common ubiquitin structural superfold, are used extensively by the cell and encompass signal tags for many different cellular fates. In this chapter we examine the current understanding of the ubiquitin regulation surrounding the insulin-like growth factor and insulin signaling systems, major members of the larger family of receptor tyrosine kinases (RTKs) and key regulators of fundamental physiological and pathological states.


Subject(s)
Receptor, IGF Type 1/metabolism , Signal Transduction , Ubiquitination , Animals , Humans , Models, Biological , Phosphorylation , Receptor, Insulin/metabolism
9.
Transplant Proc ; 47(7): 2254-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26361693

ABSTRACT

BACKGROUND: Highly sensitized patients, who produce antibodies against multiple anti-human leukocyte antigens, have significantly reduced chances for renal transplantation. Traditionally, desensitization protocols to reduce the levels of antibodies have relied on the use of intravenous immunoglobulin and plasmapheresis. RESULTS: Here we report the case of a patient with a calculated panel-reactive antibody level of 100% who was desensitized using multiple courses of bortezomib, a proteasome inhibitor, in an intravenous immunoglobulin-free regimen. The patient underwent a successful transplantation with an allograft from a living donor and has continued to do well post-transplantation. CONCLUSIONS: The expression of anti-human leukocyte antigen antibodies decreases the likelihood of transplantation for patients by restricting the available donor pool. New protocols that reduce antibody expression in these patients and allow for renal transplantation are needed. Bortezomib, as used in the patient reported here, represents a promising new medication for successful desensitization and transplantation.


Subject(s)
Bortezomib/therapeutic use , Desensitization, Immunologic/methods , Graft Rejection/therapy , HLA Antigens/immunology , Isoantibodies/immunology , Kidney Transplantation , Living Donors , Antineoplastic Agents/therapeutic use , Blood Grouping and Crossmatching , Graft Rejection/immunology , Humans , Immunoglobulins, Intravenous , Male , Middle Aged , Plasmapheresis
10.
Am J Transplant ; 15(1): 101-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25534446

ABSTRACT

A prospective iterative trial of proteasome inhibitor (PI)-based therapy for reducing HLA antibody (Ab) levels was conducted in five phases differing in bortezomib dosing density and plasmapheresis timing. Phases included 1 or 2 bortezomib cycles (1.3 mg/m(2) × 6-8 doses), one rituximab dose and plasmapheresis. HLA Abs were measured by solid phase and flow cytometry (FCM) assays. Immunodominant Ab (iAb) was defined as highest HLA Ab level. Forty-four patients received 52 desensitization courses (7 patients enrolled in multiple phases): Phase 1 (n = 20), Phase 2 (n = 12), Phase 3 (n = 10), Phase 4 (n = 5), Phase 5 (n = 5). iAb reductions were observed in 38 of 44 (86%) patients and persisted up to 10 months. In Phase 1, a 51.5% iAb reduction was observed at 28 days with bortezomib alone. iAb reductions increased with higher bortezomib dosing densities and included class I, II, and public antigens (HLA DRß3, HLA DRß4 and HLA DRß5). FCM median channel shifts decreased in 11/11 (100%) patients by a mean of 103 ± 54 mean channel shifts (log scale). Nineteen out of 44 patients (43.2%) were transplanted with low acute rejection rates (18.8%) and de novo DSA formation (12.5%). In conclusion, PI-based desensitization consistently and durably reduces HLA Ab levels providing an alternative to intravenous immune globulin-based desensitization.


Subject(s)
Boronic Acids/therapeutic use , Desensitization, Immunologic , Graft Rejection/immunology , Graft Survival/immunology , HLA Antigens/immunology , Kidney Diseases/immunology , Proteasome Inhibitors/therapeutic use , Pyrazines/therapeutic use , Adolescent , Adult , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Bortezomib , Drug Therapy, Combination , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/drug therapy , Graft Survival/drug effects , Histocompatibility Testing , Humans , Immunoglobulins, Intravenous/administration & dosage , Kidney Diseases/surgery , Kidney Function Tests , Kidney Transplantation , Male , Middle Aged , Plasmapheresis , Prognosis , Prospective Studies , Risk Factors , Rituximab , Young Adult
11.
Am J Transplant ; 13(12): 3142-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24266968

ABSTRACT

Optimal induction regimens for patients at high risk for antibody and/or cell-mediated rejection have not been established. This pilot, prospective, randomized study evaluated addition of B cell/plasma cell-targeting agents to T cell-based induction with rabbit antithymocyte globulin (rATG) in high immunologic risk renal transplant recipients. Patients were randomized to induction with rATG, rATG + rituximab, rATG + bortezomib or rATG + rituximab + bortezomib. Inclusion criteria were: (1) current cytotoxic panel reactive antibody (PRA) ≥20% or peak cytotoxic PRA ≥50% or (2) T or B cell positive flow crossmatch with donor-specific antibody (DSA) or (3) historical positive serologic or cytotoxic crossmatch or DSA to donor or (4) prior allograft loss with more than one acute rejection. Median overall follow-up was 496 days: 1-year and overall acute rejection were 25% and 27.5%, and 25% of patients developed de novo DSA within 1 year. One-year and overall patient survival were 97.5% and 92.5%, and 1-year and overall death-censored allograft survival were 97.5% and 95%. Renal allograft function posttransplant was similar among all arms. Eight of nine cases of peripheral neuropathy were mild, whereas one case was moderate and required a narcotic prescription. In conclusion, addition of rituximab and/or bortezomib to rATG induction has an acceptable safety/toxicity profile in a high immunologic risk renal transplant population.


Subject(s)
Antilymphocyte Serum/administration & dosage , B-Lymphocytes/cytology , HLA Antigens/chemistry , Kidney Transplantation , Renal Insufficiency/immunology , Adult , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , Female , Follow-Up Studies , Graft Rejection , Humans , Immunosuppressive Agents/administration & dosage , Male , Middle Aged , Pilot Projects , Postoperative Complications , Prospective Studies , Pyrazines/administration & dosage , Rabbits , Renal Insufficiency/therapy , Rituximab , Treatment Outcome
12.
Oncogene ; 31(3): 352-65, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-21685939

ABSTRACT

The human cathelicidin antimicrobial protein-18 and its C terminal peptide, LL-37, displays broad antimicrobial activity that is mediated through direct contact with the microbial cell membrane. In addition, recent studies reveal that LL-37 is involved in diverse biological processes such as immunomodulation, apoptosis, angiogenesis and wound healing. An intriguing role for LL-37 in carcinogenesis is also beginning to emerge and the aim of this paper was to explore if and how LL-37 contributes to the signaling involved in tumor development. To this end, we investigated the putative interaction between LL-37 and growth factor receptors known to be involved in tumor growth and progression. Among several receptors tested, LL-37 bound with the highest affinity to insulin-like growth factor 1 receptor (IGF-1R), a receptor that is strongly linked to malignant cellular transformation. Furthermore, this interaction resulted in a dose-dependent phosphorylation and ubiquitination of IGF-1R, with downstream signaling confined to the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)-pathway but not affecting phosphatidylinositol 3 kinase/Akt signaling. We found that signaling induced by LL-37 was dependent on the recruitment of ß-arrestin to the fully functional IGF-1R and by using mutant receptors we demonstrated that LL-37 signaling is dependent on ß-arrestin-1 binding to the C-terminus of IGF-1R. When analyzing the biological consequences of increased ERK activation induced by LL-37, we found that it resulted in enhanced migration and invasion of malignant cells in an IGF-1R/ß-arrestin manner, but did not affect cell proliferation. These results indicate that LL-37 may act as a partial agonist for IGF-1R, with subsequent intra-cellular signaling activation driven by the binding of ß-arrestin-1 to the IGF-1R. Functional experiments show that LL-37-dependent activation of the IGF-1R signaling resulted in increased migratory and invasive potential of malignant cells.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Cell Transformation, Neoplastic/metabolism , Receptor, IGF Type 1/agonists , Antimicrobial Cationic Peptides/chemistry , Arrestins/metabolism , Cell Line , Cell Movement , Cell Proliferation , Humans , MAP Kinase Signaling System , Neoplasm Invasiveness/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Ubiquitination , beta-Arrestin 1 , beta-Arrestins , Cathelicidins
13.
Pediatr Transplant ; 15(6): 548-56, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21884344

ABSTRACT

AMR is being recognized with increasing efficiency, but continues to present a significant threat to renal allograft survival. Traditional therapies for AMR (IVIG, plasmapheresis, rituximab, and antilymphocyte preparations) in general have provided inconsistent results and do not deplete the source of antibody production, viz., the mature plasma cell. Recently, the first plasma cell-targeted therapy in humans has been developed using bortezomib (a first in class PI) for AMR treatment in kidney transplant recipients. Initial experience with bortezomib involved treatment of refractory AMR. Subsequently, the efficacy of bortezomib in primary therapy for AMR was demonstrated. In a multicenter collaborative effort, the initial results with bortezomib in AMR have been confirmed and expanded to pediatric and adult heart transplant recipients. More recently, results from a prospective, staged desensitization trial has shown that bortezomib alone can substantially reduce anti-HLA antibody levels. These results demonstrate the significant potential of proteasome inhibition in addressing humoral barriers. However, the major advantage of proteasome inhibition lies in the numerous potential strategies for achieving synergy.


Subject(s)
Antibodies/chemistry , Graft Rejection/drug therapy , Graft Rejection/immunology , Kidney Transplantation/methods , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Boronic Acids/therapeutic use , Bortezomib , Humans , Plasma Cells/cytology , Prospective Studies , Proteasome Endopeptidase Complex/chemistry , Pyrazines/therapeutic use , Time Factors , Treatment Outcome
14.
Oncogene ; 27(11): 1629-38, 2008 Mar 06.
Article in English | MEDLINE | ID: mdl-17828296

ABSTRACT

The insulin-like growth factor 1 receptor (IGF-1R) is crucial for growth and survival of malignant cells. Experience in targeting IGF-1R in cancer models has shown that strategies promoting downregulation of the receptor are much more efficient in inducing apoptosis than those inhibiting the IGF-1R activity. Recently, we found that the cyclolignan picropodophyllin (PPP) inhibits phosphorylation of IGF-1R and activation of downstream signaling without interfering with the highly homologous insulin receptor (IR). Furthermore, PPP treatment caused strong regression of tumor grafts and prolonged survival of animals with systemic tumor disease. Here we demonstrate that PPP also downregulates the IGF-1R, whereas the IR and several other receptors were not affected. PPP-induced IGF-1R downregulation required expression of the MDM2 E3 ligase, which recently was found to ubiquitinate and cause degradation of the IGF-1R. In addition knockdown of beta-arrestin1, the adaptor molecule known to bridges MDM2 and IGF-1R, prevented downregulation of the receptor and significantly decreased PPP-induced cell death. All together these data suggest that PPP downregulates IGF-1R by interfering with the action of beta-arrestin1/MDM2 as well as the achieved receptor downregulation contributes to the apoptotic effect of PPP.


Subject(s)
Arrestins/metabolism , Down-Regulation , Neoplasms/metabolism , Podophyllotoxin/analogs & derivatives , Proto-Oncogene Proteins c-mdm2/metabolism , Receptor, IGF Type 1/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apoptosis , Arrestins/genetics , Blotting, Western , Cell Proliferation , Cells, Cultured , Flow Cytometry , Humans , Immunoprecipitation , Insulin-Like Growth Factor I/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Podophyllotoxin/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , Receptor, Insulin/metabolism , Swine , Ubiquitin/metabolism , beta-Arrestins
15.
Br J Cancer ; 96 Suppl: R2-6, 2007.
Article in English | MEDLINE | ID: mdl-17393577

ABSTRACT

The insulin-like growth factor (IGF-1) signalling is highly implicated in cancer. In this signalling the IGF-1 receptor (IGF-1R) is unquestionable, the predominating single factor. IGF-1R is crucial for tumour transformation and survival of malignant cell, but is only partially involved in normal cell growth. This is in part due to the interactions with oncogenes. Recent findings suggest a close interplay with the p53/MDM2 pathway. Disturbances in components in the p53/MDM2/IGF-1R network may cause IGF-1R upregulation and growth advantage for the cancer cell. Targeting of IGF-1R is more and more seen as a promising option for future cancer therapy. Single chain antibodies and small molecules with selective effects on IGF-1R dependent malignant growth are of particular interest. Forthcoming clinical trials are welcome and will indeed be the only way to evaluate the impact of IGF-1R targeting in human cancer.

16.
Oncogene ; 25(22): 3186-95, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16407828

ABSTRACT

The cyclolignan PPP was recently demonstrated to inhibit the activity of insulin-like growth factor-1 receptor (IGF-1R), without affecting the highly homologous insulin receptor. In addition, PPP caused complete regression of xenografts derived from various types of cancer. These data highlight the use of this compound in cancer treatment. However, a general concern with antitumor agents is development of resistance. In light of this problem, we aimed to investigate whether malignant cells may develop serious resistance to PPP. After trying to select 10 malignant cell lines, with documented IGF-1R expression and apoptotic responsiveness to PPP treatment (IC50s less than 0.1 microM), only two survived an 80-week selection but could only tolerate maximal PPP doses of 0.2 and 0.5 microM, respectively. Any further increase in the PPP dose resulted in massive cell death. These two cell lines were demonstrated not to acquire any essential alteration in responsiveness to PPP regarding IGF-1-induced IGF-1R phosphorylation. Neither did they exhibit any increase in expression of the multidrug resistance proteins MDR1 or MRP1. Consistently, they did not exhibit decreased sensitivity to conventional cytostatic drugs. Rather, the sensitivity was increased. During the first half of the selection period, both cell lines responded with a temporary and moderate increase in IGF-1R expression, which appeared to be because of an increased transcription of the IGF-1R gene. This increase in IGF-1R might be necessary to make cells competent for further selection but only up to a PPP concentration of 0.2 and 0.5 microM. In conclusion, malignant cells develop no or remarkably weak resistance to the IGF-1R inhibitor PPP.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Podophyllotoxin/analogs & derivatives , Receptor, IGF Type 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis/drug effects , Dose-Response Relationship, Drug , Humans , Multidrug Resistance-Associated Proteins/metabolism , Neoplasms/metabolism , Phosphorylation , Podophyllotoxin/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
Transpl Immunol ; 15(1): 63-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16223674

ABSTRACT

UNLABELLED: The complement activation demonstrated by vascular C4d deposition is used to diagnose antibody-mediated rejection (AMR) in renal allografts, but remains controversial in lung transplantation (LTX). METHODS: C4d deposition was assessed by immunohistochemistry in 192 lung transplant biopsies from 32 patients. ELISA analysis was performed on 415 serum samples in those 32 temporally and rejection-grade matched LTX patients; 16 patients developed HLA-Ab, while the other 16 patients remained negative. The specificity of C4d staining was further compared in 18 additional LTX patients without HLA-Ab or acute cellular rejection (ACR), but in the presence of CMV-pneumonitis or reperfusion injury. RESULTS: Specific subendothelial C4d deposition was seen in 5 of 16 (31%) patients with HLA-Ab and was absent in 16 patients without HLA-Ab (p<0.05). All patients with specific C4d deposition exhibited donor-specific HLA-Ab. There were 13 patients with bronchiolitis obliterans syndrome in the group of 16 HLA-Ab positive patients, versus 2/16 in ELISA-negative patients (p<0.005). One of 7 patients with CMV pneumonitis and 2 of 11 patients with reperfusion injury also showed C4d positivity (not statistically significant). CONCLUSIONS: In this study, specific subendothelial C4d deposition was a marker for the involvement of HLA-Ab in lung allograft rejection. The patchy nature, low sensitivity, and specificity of C4d staining might limit clinical use in protocol biopsies. However, in patients with decreasing pulmonary function, refractory ACR and/or HLA-Ab, specific C4d deposition may serve as a marker of coexistent AMR.


Subject(s)
Complement C4b/analysis , Graft Rejection/diagnosis , HLA Antigens/immunology , Isoantibodies/blood , Lung Transplantation/immunology , Lung/immunology , Peptide Fragments/analysis , Acute Disease , Graft Rejection/immunology , Humans
18.
Br J Cancer ; 92(12): 2097-101, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15956962

ABSTRACT

The insulin-like growth factor (IGF-1) signalling is highly implicated in cancer. In this signalling the IGF-1 receptor (IGF-1R) is unquestionable, the predominating single factor. IGF-1R is crucial for tumour transformation and survival of malignant cell, but is only partially involved in normal cell growth. This is in part due to the interactions with oncogenes. Recent findings suggest a close interplay with the p53/MDM2 pathway. Disturbances in components in the p53/MDM2/IGF-1R network may cause IGF-1R upregulation and growth advantage for the cancer cell. Targeting of IGF-1R is more and more seen as a promising option for future cancer therapy. Single chain antibodies and small molecules with selective effects on IGF-1R dependent malignant growth are of particular interest. Forthcoming clinical trials are welcome and will indeed be the only way to evaluate the impact of IGF-1R targeting in human cancer.


Subject(s)
Neoplasms/physiopathology , Receptor, IGF Type 1/physiology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor/drug effects , Genes, Tumor Suppressor/physiology , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogenes/drug effects , Proto-Oncogenes/physiology , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/physiology
19.
Cancer Res ; 60(18): 5278-83, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016658

ABSTRACT

We investigated the functional impact of p53 on insulin-like growth factor I receptor (IGF-IR) expression in malignant cells. Using the BL-41tsp53-2 cell line, a transfectant carrying temperature-sensitive (ts) p53 and endogenous mutant p53 (codon 248), we demonstrated a drastic down-regulation of plasma membrane-bound IGF-IRs on induction of wild-type p53. However, a similar response was obtained by treatment of BL-41tsp53-2 cells expressing mutant ts p53 with a p53 antisense oligonucleotide. Thus, even if the negative effect of wild-type p53 predominates under a competitive condition, these data indicate that mutant p53 may be important for up-regulation of IGF-IR. To further elucidate this issue, three melanoma cell lines (BE, SK-MEL-5, and SK-MEL-28) that overexpressed p53 were investigated. The BE cell line has a "hot spot" mutation (codon 248) and expresses only codon 248-mutant p53. SK-MEL-28 has a point mutation at codon 145. SK-MEL-5 cells did not exhibit any p53 mutations, but the absence of p21Waf1 expression suggested functionally aberrant p53. Our data suggest that interaction with Mdm-2 may underlie p53 inactivation in these cells. Using p53 antisense oligonucleotides, we demonstrated a substantial down-regulation of cell surface expression of IGF-IR proteins in all melanoma cell lines after 24 h. This was paralleled by decreased tyrosine phosphorylation of IGF-IR and growth arrest, and, subsequently, massive cell death was observed (this was also seen in BL-41tsp53-2 cells with mutant conformation of ts p53). Taken together, our results suggest that up-regulation of IGF-IR as a result of expression of aberrant p53 may be important for the growth and survival of malignant cells.


Subject(s)
Acetylcysteine/analogs & derivatives , Receptor, IGF Type 1/biosynthesis , Tumor Suppressor Protein p53/physiology , Acetylcysteine/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Humans , Melanoma/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
20.
Oncogene ; 19(37): 4298-301, 2000 Aug 31.
Article in English | MEDLINE | ID: mdl-10980604

ABSTRACT

The EWS/FLI-1 fusion gene is characteristic of most cases of Ewing's sarcoma and has been shown to be crucial for tumor transformation and cell growth. In this study we demonstrate a drastic down-regulation of the EWS/FLI-1 protein, and a growth arrest, following serum depletion of Ewing's sarcoma cells. This indicates that growth factor circuits may be involved in regulation of the fusion gene product. Of four different growth factors tested, basic fibroblast growth factor (bFGF) was found to be of particular significance. In fact, upon treatment of serum-depleted cells with bFGF, expression of the EWS/FLI-1 protein and growth of the Ewing's sarcoma cells were restored. In addition, a bFGF-neutralizing antibody, which was confirmed to inhibit FGF receptor (FGFR) phosphorylation, caused down-regulation of EWS/FLI-1. Experiments using specific cell cycle blockers (thymidine and colcemide) suggest that EWS/FLI-1 is directly linked to bFGF stimulation, and not indirectly to cell proliferation. We also demonstrated expression of FGFRs in several tumor samples of Ewing's sarcoma. Taken together, our data suggest that expression of FGFR is a common feature of Ewing's sarcoma and, in particular, that the bFGF pathway may be important for the maintenance of a malignant phenotype of Ewing's sarcoma cells through up-regulating the EWS/FLI-1 protein. Oncogene (2000) 19, 4298 - 4301


Subject(s)
Bone Neoplasms/pathology , Fibroblast Growth Factor 2/physiology , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/biosynthesis , Sarcoma, Ewing/pathology , Transcription Factors/biosynthesis , Adenocarcinoma/pathology , Antibodies, Monoclonal/pharmacology , Bone Neoplasms/metabolism , Cell Cycle/drug effects , Cell Division/drug effects , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 22/ultrastructure , Culture Media, Serum-Free , Demecolcine/pharmacology , Drug Synergism , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/immunology , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Platelet-Derived Growth Factor/pharmacology , Prostatic Neoplasms/pathology , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/physiology , Sarcoma, Ewing/metabolism , Thymidine/pharmacology , Transcription Factors/genetics , Translocation, Genetic , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL