Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Oncoimmunology ; 8(11): e1660121, 2019.
Article in English | MEDLINE | ID: mdl-31646109

ABSTRACT

Before the era of immune checkpoint blockade, a meta-analysis encompassing fifteen trials reported that adjuvant IFN-α significantly reduces the risk of relapse and improves survival of ulcerated melanoma (UM) with no benefit for higher doses compared to lower doses. IFNa2b affects many cell intrinsic features of tumor cells and modulates the host innate and cognate immune responses. To better understand the biological traits associated with ulceration that could explain the efficacy of prophylactic type 1 IFN, we performed immunohistochemical analysis of various molecules (major histocompatibility complex class I and class II, MX Dynamin Like GTPase 1 (MX1), inducible Nitric-Oxide Synthase (iNOS) or CD47) in two retrospective cohorts of melanoma patients, one diagnosed with a primary cutaneous melanoma (1995-2013, N = 172, among whom 49% were ulcerated melanoma (UM)) and a second one diagnosed with metastatic melanoma amenable to lymph node resection (EORTC 18952 and 18991 trials, N = 98, among whom 44% were UM). We found that primary and metastatic UM exhibit higher basal expression of MHC class I molecules, independently of Breslow thickness, histology and lymphocytic infiltration compared with NUM and that primary UM harbored higher constitutive levels of the antiviral protein Mx1 at the border of tumor beds than NUM. These findings suggest that UM expand in a tumor microenvironment where chronic exposure to type 1 IFN could favor a response to exogenous IFNs.

3.
Ann Pathol ; 37(1): 55-60, 2017 Feb.
Article in French | MEDLINE | ID: mdl-28111041

ABSTRACT

Prognosis and treatment of advanced melanoma have been transformed by the success of immunotherapies, in particular agents targeting PD-1. PD-L1 expression assessed by immunohistochemistry in not an effective predictive biomarker to select patients in this tumor type, since significant clinical benefit was observed in the group of patients with negative tumors. The predictive value of PD-L1 testing to select patients for combination of anti-PD-1 and anti-CTLA-4 agents is under evaluation. Other tissue biomarkers are emerging to identify sensitive tumors to anti-PD-1 agents. In particular, assessment of immune infiltrates in tumor tissue, mutational load and tumor neoantigens seem promising in melanoma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/analysis , Biomarkers/analysis , Immunotherapy , Melanoma/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Biomarkers, Tumor/analysis , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/immunology , Clinical Trials as Topic , Drug Monitoring , Drug Resistance, Neoplasm , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/chemistry , Melanoma/immunology , Melanoma/pathology , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , Nivolumab , Prognosis , Programmed Cell Death 1 Receptor/analysis , Programmed Cell Death 1 Receptor/immunology
4.
Cancer Res ; 77(2): 557-565, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27836854

ABSTRACT

Raman spectroscopy is a noninvasive and label-free optical technique that provides detailed information about the molecular composition of a sample. In this study, we evaluated the potential of Raman spectroscopy to predict skin toxicity due to tyrosine kinase inhibitors treatment. We acquired Raman spectra of skin of patients undergoing treatment with MEK, EGFR, or BRAF inhibitors, which are known to induce severe skin toxicity; for this pilot study, three patients were included for each inhibitor. Our algorithm, based on partial least squares-discriminant analysis (PLS-DA) and cross-validation by bootstrapping, discriminated to variable degrees spectra from patient suffering and not suffering cutaneous adverse events. For MEK and EGFR inhibitors, discriminative power was more than 90% in the viable epidermis skin layer; whereas for BRAF inhibitors, discriminative power was 71%. There was a 81.5% correlation between blood drug concentration and Raman signature of skin in the case of EGFR inhibitors and viable epidermis skin layer. Our results demonstrate the power of Raman spectroscopy to detect apparition of skin toxicity in patients treated with tyrosine kinase inhibitors at levels not detectable via dermatological inspection and histological evaluation. Cancer Res; 77(2); 557-65. ©2016 AACR.


Subject(s)
Algorithms , Antineoplastic Agents/adverse effects , Drug Eruptions/diagnosis , Skin/chemistry , Spectrum Analysis, Raman/methods , Aged , Area Under Curve , Biomarkers/analysis , Erlotinib Hydrochloride/adverse effects , Female , Humans , Imidazoles/adverse effects , Indoles/adverse effects , Male , Middle Aged , Oximes/adverse effects , Pilot Projects , Protein Kinase Inhibitors/adverse effects , Pyridones/adverse effects , Pyrimidinones/adverse effects , ROC Curve , Sensitivity and Specificity , Skin/pathology , Sulfonamides/adverse effects , Vemurafenib
5.
Cancer Res ; 76(6): 1476-84, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26825172

ABSTRACT

BRAF inhibitors (BRAFi) elicit therapeutic responses in metastatic melanoma, but alarmingly, also induce the formation of secondary benign and malignant skin tumors. Here, we report the emergence and molecular characterization of 73 skin and extracutaneous tumors in 31 patients who underwent BRAFi therapy. The majority of patients presented with classic epidermal tumors such as verrucous papillomas, keratoacanthomas, and squamous cell carcinomas (SCC). However, 15 patients exhibited new or rapidly progressing tumors distinct from these classic subtypes, such as lymph node metastasis, new melanomas, and genital and oral mucosal SCCs. Genotyping of the tumors revealed that oncogenic RAS mutations were found in 58% of the evaluable tumor samples (38/66) and 49% of the control tumors from patients not treated with BRAFi (30/62). Notably, proximity ligation assays demonstrated that BRAF-CRAF heterodimerization was increased in fixed tumor samples from BRAFi-treated patients compared with untreated patients. Our findings reveal that BRAF-CRAF complex formation is significantly associated with BRAFi treatment, and may therefore serve as a useful biomarker of BRAFi-induced cutaneous and extracutaneous tumor formation.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Cell Line, Tumor , Dimerization , Genotype , Humans , Mutation/drug effects , Proto-Oncogene Proteins B-raf/metabolism
6.
JAMA Dermatol ; 152(1): 45-51, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26501224

ABSTRACT

IMPORTANCE: Vitiligo is an autoimmune skin disorder that reacts against melanocytes. The association of vitiligo with tumor response in patients with melanoma who undergo immunotherapy has been reported but is still controversial. OBJECTIVE: To prospectively evaluate the appearance of vitiligo in patients receiving pembrolizumab, a monoclonal antibody directed against the programmed death cell receptor. DESIGN, SETTING, AND PARTICIPANTS: This prospective observational study was conducted from January 1, 2012, through September 24, 2013, in a single tertiary care hospital with a unit dedicated to patients with melanoma. Sixty-seven patients with metastatic melanoma who received pembrolizumab treatment in the context of a phase 1 study were included and screened for the emergence of vitiligo. Data were collected from January 1, 2012, to February 28, 2014, and analyzed from February through December 2014. MAIN OUTCOMES AND MEASURES: Objective tumor response with regard to the occurrence of vitiligo in patients receiving pembrolizumab therapy. Correlation between vitiligo occurrence and overall survival was also estimated using the Kaplan-Meier product-limit method and compared with a log-rank test. To prevent guarantee- or lead-time bias, a landmark analysis approach after 12, 16, and 20 weeks of treatment was retained. RESULTS: Of the 67 patients included in the study, 17 (25%) developed vitiligo during pembrolizumab treatment and 50 (75%) did not. An objective (complete or partial) response to treatment was associated with a higher occurrence of vitiligo (12 of 17 [71%] vs 14 of 50 [28%]; P = .002). The time to onset of vitiligo ranged from 52 to 453 (median, 126) days from the start of treatment. Of the 17 patients with vitiligo, 3 (18%) had a complete response, 9 (53%) had a partial response, 3 (18%) had stable disease, and 2 (12%) had progressive disease at the final follow-up. All the patients treated with pembrolizumab who developed vitiligo were alive at the time of analysis, with a median follow-up of 441 days. CONCLUSIONS AND RELEVANCE: Vitiligo, a clinically visible immune-related adverse event could be associated with clinical benefit in the context of pembrolizumab treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Vitiligo/etiology , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Female , Follow-Up Studies , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Prospective Studies , Skin Neoplasms/pathology , Survival Rate , Time Factors , Treatment Outcome , Young Adult
7.
Eur J Cancer ; 51(13): 1780-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26072362

ABSTRACT

INTRODUCTION: In patients with primary cutaneous melanoma, there is generally a delay between excisional biopsy of the primary tumour and sentinel-node biopsy. The objective of this study is to analyse the prognostic implications of this delay. PATIENTS AND METHOD: This was an observational, retrospective, cohort study in four tertiary referral hospitals. A total of 1963 patients were included. The factor of interest was the interval between the date of the excisional biopsy of the primary melanoma and the date of the sentinel-node biopsy (delay time) in the prognosis. The primary outcome was melanoma-specific survival and disease-free survival. RESULTS: A delay time of 40 days or less (hazard ratio (HR), 1.7; confidence interval (CI), 1.2-2.5) increased Breslow thickness (Breslow ⩾ 2 mm, HR, > 3.7; CI, 1.4-10.7), ulceration (HR, 1.6; CI, 1.1-2.3), sentinel-node metastasis (HR, 2.9; CI, 1.9-4.2), and primary melanoma localised in the head or neck were independently associated with worse melanoma-specific survival (all P < 0.03). The stratified analysis showed that the effect of delay time was at the expense of the patients with a negative sentinel-node biopsy and without regression. CONCLUSION: Early sentinel-node biopsy is associated with worse survival in patients with cutaneous melanoma.


Subject(s)
Lymph Nodes/pathology , Melanoma/secondary , Sentinel Lymph Node Biopsy , Skin Neoplasms/pathology , Waiting Lists , Adult , Aged , Disease-Free Survival , Female , France , History, Ancient , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Melanoma/mortality , Melanoma/therapy , Middle Aged , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Skin Neoplasms/mortality , Skin Neoplasms/therapy , Spain , Tertiary Care Centers , Time Factors , Treatment Outcome
8.
Nature ; 515(7528): 568-71, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25428505

ABSTRACT

Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8(+) T cells (termed adaptive immune resistance). Here we show that pre-existing CD8(+) T cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analysed samples from 46 patients with metastatic melanoma obtained before and during anti-PD-1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next-generation sequencing for T-cell antigen receptors (TCRs). In serially sampled tumours, patients responding to treatment showed proliferation of intratumoral CD8(+) T cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8-, PD-1- and PD-L1-expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression after therapeutic PD-1 blockade requires pre-existing CD8(+) T cells that are negatively regulated by PD-1/PD-L1-mediated adaptive immune resistance.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Melanoma/therapy , Models, Biological , Aged , Aged, 80 and over , Biomarkers , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/diagnosis , Melanoma/immunology , Melanoma/pathology , Middle Aged , Multivariate Analysis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
9.
Nature ; 513(7516): 105-9, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25079330

ABSTRACT

In BRAF(V600)-mutant tumours, most mechanisms of resistance to drugs that target the BRAF and/or MEK kinases rely on reactivation of the RAS-RAF-MEK-ERK mitogen-activated protein kinase (MAPK) signal transduction pathway, on activation of the alternative, PI(3)K-AKT-mTOR, pathway (which is ERK independent) or on modulation of the caspase-dependent apoptotic cascade. All three pathways converge to regulate the formation of the eIF4F eukaryotic translation initiation complex, which binds to the 7-methylguanylate cap (m(7)G) at the 5' end of messenger RNA, thereby modulating the translation of specific mRNAs. Here we show that the persistent formation of the eIF4F complex, comprising the eIF4E cap-binding protein, the eIF4G scaffolding protein and the eIF4A RNA helicase, is associated with resistance to anti-BRAF, anti-MEK and anti-BRAF plus anti-MEK drug combinations in BRAF(V600)-mutant melanoma, colon and thyroid cancer cell lines. Resistance to treatment and maintenance of eIF4F complex formation is associated with one of three mechanisms: reactivation of MAPK signalling, persistent ERK-independent phosphorylation of the inhibitory eIF4E-binding protein 4EBP1 or increased pro-apoptotic BCL-2-modifying factor (BMF)-dependent degradation of eIF4G. The development of an in situ method to detect the eIF4E-eIF4G interactions shows that eIF4F complex formation is decreased in tumours that respond to anti-BRAF therapy and increased in resistant metastases compared to tumours before treatment. Strikingly, inhibiting the eIF4F complex, either by blocking the eIF4E-eIF4G interaction or by targeting eIF4A, synergizes with inhibiting BRAF(V600) to kill the cancer cells. eIF4F not only appears to be an indicator of both innate and acquired resistance but also is a promising therapeutic target. Combinations of drugs targeting BRAF (and/or MEK) and eIF4F may overcome most of the resistance mechanisms arising in BRAF(V600)-mutant cancers.


Subject(s)
Drug Resistance, Neoplasm , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/metabolism , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Female , Humans , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Melanoma/pathology , Mice , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thyroid Neoplasms/pathology , Triterpenes/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
10.
Rev Prat ; 64(1): 75-80, 2014 Jan.
Article in French | MEDLINE | ID: mdl-24649552

ABSTRACT

Management of patients with metastatic melanoma has been revolutionized over the last few years with targeted anti-BRAF therapies for BRAF-mutant melanomas (in about 50% of the cases) and immunotherapy with anti-CTLA-4. Several new drugs are now authorized and available. Because of their new mechanisms of action, they also have new adverse events and guidelines concerning their safety are of critical importance. New innovative strategies using combination of targeted therapies and immunotherapies with anti-PD-1 are in accelerated development. The quality of patient-physician relationship is central to this promising but complex new paradigm of treatment.


Subject(s)
Melanoma/therapy , Skin Neoplasms/therapy , Brain Neoplasms/secondary , Humans , Lymphatic Metastasis , Melanoma/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Skin Neoplasms/pathology
11.
Cancer Res ; 74(8): 2238-45, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24523442

ABSTRACT

Treatment with RAF inhibitors such as vemurafenib causes the development of cutaneous squamous cell carcinomas (cSCC) or keratoacanthomas as a side effect in 18% to 30% of patients. It is known that RAF inhibitors activate the mitogen-activated protein kinase (MAPK) pathway and stimulate growth of RAS-mutated cells, possibly accounting for up to 60% of cSCC or keratoacanthoma lesions with RAS mutations, but other contributing events are obscure. To identify such events, we evaluated tumors from patients treated with vemurafenib for the presence of human papilloma virus (HPV) DNA and identified 13% to be positive. Using a transgenic murine model of HPV-driven cSCC (K14-HPV16 mice), we conducted a functional test to determine whether administration of RAF inhibitors could promote cSCC in HPV-infected tissues. Vemurafenib treatment elevated MAPK markers and increased cSCC incidence from 22% to 70% in this model. Furthermore, 55% of the cSCCs arising in vemurafenib-treated mice exhibited a wild-type Ras genotype, consistent with the frequency observed in human patients. Our results argue that HPV cooperates with vemurafenib to promote tumorigenesis, in either the presence or absence of RAS mutations.


Subject(s)
Carcinoma, Squamous Cell/etiology , Human papillomavirus 16/physiology , Indoles/adverse effects , Skin Neoplasms/etiology , Sulfonamides/adverse effects , Animals , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Early Detection of Cancer , Genotype , Human papillomavirus 16/genetics , Humans , Indoles/administration & dosage , Keratin-14/genetics , MAP Kinase Signaling System/drug effects , Mice , Mice, Transgenic , Promoter Regions, Genetic , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Skin Neoplasms/virology , Sulfonamides/administration & dosage , Vemurafenib
12.
Am J Dermatopathol ; 36(8): e146-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24335517

ABSTRACT

Trichoblastoma is a benign cutaneous adnexal tumor, composed mostly of follicular germinative cells. Its pigmented variant is colonized by numerous dendritic melanocytes. So far, only one case in the literature describes a combination of trichoblastoma and melanoma. We report the case of a 62-year-old man who had a slow-growing mass of the left flank present since childhood. This 8-cm mass was surgically removed when it became ulcerated and associated with axillary lymph nodes. Histologically, this tumor was strictly dermal and composed of 2 intermingled components. Large sheets of atypical, proliferating epithelioid cells predominated. Dispersed solid nests or cribriform epithelial islets encased in fibrous tissue were also seen. Some nests displayed a massive colonization by pigmented dendritic melanocytes. On immunohistochemical staining, the sheets of atypical cells expressed focally but strongly S100 protein, MelanA, HMB45, and MiTF. Epithelial structures diffusely expressed pancytokeratin AE1/AE3, KL1, and pleckstrin homology-like domain, family A, member 1. Based on these results, we diagnosed an intradermal melanoma, possibly developed from dendritic melanocytes colonizing a giant pigmented trichoblastoma. Direct sequencing of the melanoma revealed a rarely described NRAS mutation c.34G>T (G12C). Array comparative genomic hybridization displayed a complex profile somewhat divergent from standard melanoma profiles. The patient died of widespread metastatic disease 8 months after initial diagnosis.


Subject(s)
Biomarkers, Tumor/genetics , Comparative Genomic Hybridization , DNA Mutational Analysis , GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Neoplasms, Adnexal and Skin Appendage/genetics , Skin Neoplasms/genetics , Biomarkers, Tumor/analysis , Biopsy , Fatal Outcome , Humans , Immunohistochemistry , Male , Melanins/analysis , Melanocytes/chemistry , Melanocytes/pathology , Melanoma/chemistry , Melanoma/secondary , Middle Aged , Mutation , Neoplasms, Adnexal and Skin Appendage/chemistry , Neoplasms, Adnexal and Skin Appendage/pathology , Neoplasms, Adnexal and Skin Appendage/surgery , Skin Neoplasms/chemistry , Skin Neoplasms/pathology , Time Factors
13.
JAMA Dermatol ; 149(7): 855-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23699661

ABSTRACT

IMPORTANCE: The BRAF inhibitor, vemurafenib, was recently approved for the treatment of patients with BRAFV600 metastatic melanoma. Wider use of this drug and longer follow-up periods of treatment are resulting in the emergence of a growing number of reports detailing new adverse effects. Cutaneous adverse effects are preeminent with UV-A-dependent phototoxicity, hyperkeratotic folliculitis, hand-foot skin reaction, hair changes, verrucous papillomas, keratoacanthomas, and squamous cell carcinomas. OBSERVATIONS: We report 2 cases of dermatitis occurring on a previously irradiated skin area in patients treated with vemurafenib for a BRAFV600-mutated metastatic melanoma. The first case occurred 10 days after a low dose of radiation was delivered that usually does not induce any radiodermatitis, suggesting radiosensitization by vemurafenib. The second case occurred 30 days after radiotherapy and was diagnosed as radiation recall dermatitis. CONCLUSIONS AND RELEVANCE: Vemurafenib should be considered a potential cutaneous radiosensitizer and an inducer of radiation recall dermatitis. However, these adverse effects are easily managed with topical corticosteroids. Dose reduction or interruption of vemurafenib is not required. Further studies and reports will enlighten us as to whether this pharmacodynamic interaction between x-rays and vemurafenib is also seen with other BRAF or MEK inhibitors on the same mitogen-activated protein kinase pathway currently under development.


Subject(s)
Indoles/adverse effects , Melanoma/drug therapy , Protein Kinase Inhibitors/adverse effects , Radiation Tolerance/drug effects , Skin Neoplasms/drug therapy , Sulfonamides/adverse effects , Adult , Female , Humans , Indoles/therapeutic use , Male , Melanoma/radiotherapy , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/radiotherapy , Sulfonamides/therapeutic use , Vemurafenib
15.
Clin Cancer Res ; 18(1): 263-72, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22096025

ABSTRACT

PURPOSE: The emergence of skin tumors in patients treated with sorafenib or with more recent BRAF inhibitors is an intriguing and potentially serious event. We carried out a clinical, pathologic, and molecular study of skin lesions occurring in patients receiving sorafenib. EXPERIMENTAL DESIGN: Thirty-one skin lesions from patients receiving sorafenib were characterized clinically and pathologically. DNA extracted from the lesions was screened for mutation hot spots of HRAS, NRAS, KiRAS, TP53, EGFR, BRAF, AKT1, PI3KCA, TGFBR1, and PTEN. Biological effect of sorafenib was studied in vivo in normal skin specimen and in vitro on cultured keratinocytes. RESULTS: We observed a continuous spectrum of lesions: from benign to more inflammatory and proliferative lesions, all seemingly initiated in the hair follicles. Eight oncogenic HRAS, TGFBR1, and TP53 mutations were found in 2 benign lesions, 3 keratoacanthomas (KA) and 3 KA-like squamous cell carcinoma (SCC). Six of them correspond to the typical UV signature. Treatment with sorafenib led to an increased keratinocyte proliferation and a tendency toward increased mitogen-activated protein kinase (MAPK) pathway activation in normal skin. Sorafenib induced BRAF-CRAF dimerization in cultured keratinocytes and activated CRAF with a dose-dependent effect on MAP-kinase pathway activation and on keratinocyte proliferation. CONCLUSION: Sorafenib induces keratinocyte proliferation in vivo and a time- and dose-dependent activation of the MAP kinase pathway in vitro. It is associated with a spectrum of lesions ranging from benign follicular cystic lesions to KA-like SCC. Additional and potentially preexisting somatic genetic events, like UV-induced mutations, might influence the evolution of benign lesions to more proliferative and malignant tumors.


Subject(s)
Benzenesulfonates/adverse effects , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/adverse effects , Receptors, Transforming Growth Factor beta/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Cells, Cultured , Female , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/radiation effects , Male , Middle Aged , Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/diagnosis , Sorafenib , Ultraviolet Rays/adverse effects , raf Kinases/genetics , ras Proteins/genetics
16.
Int J Cancer ; 127(6): 1321-31, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20049837

ABSTRACT

KRAS mutations are a strong predictive marker of resistance to anti-epidermal growth factor receptor (EGFR) antibodies in advanced colorectal cancer (CRC) but only a subset of wild-type (WT) KRAS patients are responders, suggesting the existence of additional markers of resistance to this treatment. The activation of EGFR downstream signaling pathways may be one of these ones. In a series of 42 patients with advanced CRC treated with cetuximab/panitumumab, for whom KRAS status was previously determined, we retrospectively analyzed the intratumor expression of EGFR downstream signaling phosphoproteins of the RAS/MAPK and PI3K/AKT pathways (pERK1/2, pMEK1, pAKT, pP70S6K and pGSK3beta) using Bio-Plex phosphoprotein array. Association with tumor response, progression-free survival (PFS) and overall survival (OS) was assessed. The expression of all the phosphoproteins was higher in KRAS mutated tumors than in WT tumors. The expression of pP70S6K was lower in responders than in nonresponder patients. In univariate analysis, patients with high pMEK1 or pP70S6K expression had a shorter PFS than those with low expression. Patients with high pP70S6K expression also had a shorter OS. In multivariate analysis, PFS was shorter for patients with high pMEK1 or pP70S6K expression, independently of KRAS status, as OS for patients with high pP70S6K expression. Therefore, WT KRAS patients with high pP70S6K expression had a shorter survival than those with low expression. Our results suggest the importance of EGFR downstream signaling phosphoproteins expression in addition to KRAS status to define the subgroup of patients who will not benefit from anti-EGFR therapy.


Subject(s)
Antibodies/immunology , Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Genes, ras , Phosphoproteins/metabolism , Signal Transduction , Aged , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Disease-Free Survival , ErbB Receptors/immunology , Female , Humans , Male , Middle Aged , Mutation , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics
17.
Oncologist ; 14(12): 1182-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19939894

ABSTRACT

BACKGROUND: Chemokine receptor 4 (CXCR4) has been demonstrated to have a critical role in the early metastatic process. The aim of this study was to evaluate the prognostic value of CXCR4 expression in primary breast tumors and describe correlations with the occurrence of metastasis in organs expressing the CXCR4 ligand stromal cell-derived factor 1 (i.e., liver, lung, brain, and bone). PATIENTS AND METHODS: CXCR4 expression in primary breast tumors was evaluated by immunohistochemistry in 823 patients included in two prospective clinical trials. CXCR4 expression was considered positive when >1% of tumor cells were stained. The prognostic value of CXCR4 expression was assessed by a Cox regression model adjusted for clinical characteristics. We assessed the association of CXCR4 expression with the rate of distant metastasis to specific organ sites. RESULTS: CXCR4 was expressed in 92 of 794 primary tumors (12%). CXCR4 expression was not associated with clinical characteristics. CXCR4 was not prognostic for overall survival and showed a nonsignificant trend toward a higher risk for distant metastasis. CXCR4(+) tumors showed a significantly higher risk for bone metastasis. The 10-year incidences of bone metastases were 23% (13.6%-32.6%) and 12% (9.7%-15%) in CXCR4(+) and CXCR4(-) tumors, respectively. CONCLUSION: This study suggests that expression of CXCR4 in primary breast tumors is associated with a higher likelihood of developing bone metastases. This finding could open new avenues for the development of novel adjuvant strategies, including bone-targeting agents.


Subject(s)
Breast Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , Aged , Anthracyclines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Chemotherapy, Adjuvant , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Prospective Studies , Survival Rate
19.
J Clin Oncol ; 26(3): 374-9, 2008 Jan 20.
Article in English | MEDLINE | ID: mdl-18202412

ABSTRACT

PURPOSE: Cetuximab is efficient in advanced colorectal cancer (CRC). We previously showed that KRAS mutations were associated with resistance to cetuximab in 30 CRC patients. The aim of this study was to validate, in an independent larger series of 89 patients, the prognostic value of KRAS mutations on response to cetuximab and survival. PATIENTS AND METHODS: Eighty-nine metastatic CRC patients treated with cetuximab after treatment failure with irinotecan-based chemotherapy were analyzed for KRAS mutation by allelic discrimination on tumor DNA. The association between KRAS mutations and tumor response, skin toxicity, progression-free survival (PFS) and overall survival (OS) was analyzed. RESULTS: A KRAS mutation was present in 27% of the patients and was associated with resistance to cetuximab (0% v 40% of responders among the 24 mutated and 65 nonmutated patients, respectively; P < .001) and a poorer survival (median PFS: 10.1 v 31.4 weeks in patients without mutation; P = .0001; median OS: 10.1 v 14.3 months in patients without mutation; P = .026). When we pooled these 89 patients with patients from our previous study, the multivariate analysis showed that KRAS status was an independent prognostic factor associated with OS and PFS, whereas skin toxicity was only associated with OS. In a combined analysis, median OS times of patients with two, one, or no favorable prognostic factors (severe skin toxicity and no KRAS mutation) was of 15.6, 10.7, and 5.6 months, respectively. CONCLUSION: These results confirm the high prognostic value of KRAS mutations on response to cetuximab and survival in metastatic CRC patients treated with cetuximab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mutation/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Antibodies, Monoclonal, Humanized , Cetuximab , Colorectal Neoplasms/mortality , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins p21(ras) , Retrospective Studies , Survival Rate
20.
Ann Surg Oncol ; 14(11): 3188-94, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17705091

ABSTRACT

BACKGROUND: Dramatic responses to chemotherapy are occurring more and more frequently in patients with multiple colorectal liver metastases (LMs), leading to resection. In a few patients, some LMs vanish on imaging studies, remain undetected during hepatectomy, and are left in place, which defines the "missing LMs." The aim of our study was to assess the long-term outcome of such "missing LMs." PATIENTS: Between January 1999 and June 2004, among 228 patients treated for colorectal LMs, missing LMs were observed in 16 patients. All the patients were operated within 4 weeks of imaging. Hepatic arterial infusion (HAI) with oxaliplatin was administrated in 12 patients (75%): seven before hepatectomy and five after. RESULTS: Overall, 69 missing LMs were diagnosed and left in place. Among the persistent LMs resected, a complete pathological response was significantly more often observed in the group with preoperative HAI (6 of 7), than in the group without (2 of 9, P < .02). With a mean follow-up of 51 months (24-90), missing LMs did not reappear in 10 patients (62%). Adjuvant HAI was significantly correlated with the definitive eradication of missing LMs (P < .01), as it was not a complete pathological response. The overall 3-year survival rate of these highly selected 16 patients was 94%. CONCLUSION: Colorectal LMs under chemotherapy that vanish on high-quality imaging studies, remain undetected during hepatectomy, and are left in place, are definitively cured in 62% of cases. This excellent result seems to be due to the administration of adjuvant hepatic arterial infusion of chemotherapy and should stimulate new investigations.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/pathology , Hepatectomy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Organoplatinum Compounds/administration & dosage , Adult , Aged , Chemotherapy, Adjuvant , Colorectal Neoplasms/therapy , Female , Hepatic Artery , Humans , Infusions, Intra-Arterial , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/therapy , Oxaliplatin , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...