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










Publication year range
1.
Clin Cancer Res ; 27(4): 1037-1047, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33272982

ABSTRACT

PURPOSE: TNF blockers can be used to manage gastrointestinal inflammatory side effects following nivolumab and/or ipilimumab treatment in patients with advanced melanoma. Our preclinical data showed that anti-TNF could promote the efficacy of immune checkpoint inhibitors. PATIENTS AND METHODS: TICIMEL (NTC03293784) is an open-label, two-arm phase Ib clinical trial. Fourteen patients with advanced and/or metastatic melanoma (stage IIIc/IV) were enrolled. Patients were treated with nivolumab (1 mg/kg) and ipilimumab (3 mg/kg) combined to infliximab (5 mg/kg, N = 6) or certolizumab (400/200 mg, N = 8). The primary endpoint was safety and the secondary endpoint was antitumor activity. Adverse events (AEs) were graded according to the NCI Common Terminology Criteria for Adverse Events and response was assessed following RECIST 1.1. RESULTS: Only one dose-limiting toxicity was observed in the infliximab cohort. The two different combinations were found to be safe. We observed lower treatment-related AEs with infliximab as compared with certolizumab. In the certolizumab cohort, one patient was not evaluable for response. In this cohort, four of eight patients exhibited hepatobiliary disorders and seven of seven evaluable patients achieved objective response including four complete responses (CRs) and three partial responses (PRs). In the infliximab cohort, we observed one CR, two PRs, and three progressive diseases. Signs of activation and maturation of systemic T-cell responses were seen in patients from both cohorts. CONCLUSIONS: Our results show that both combinations are safe in human and provide clinical and biological activities. The high response rate in the certolizumab-treated patient cohort deserves further investigations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Certolizumab Pegol/administration & dosage , Certolizumab Pegol/adverse effects , Female , Follow-Up Studies , Humans , Infliximab/administration & dosage , Infliximab/adverse effects , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Male , Melanoma/diagnosis , Melanoma/mortality , Melanoma/secondary , Middle Aged , Nivolumab/administration & dosage , Nivolumab/adverse effects , Progression-Free Survival , Skin Neoplasms/diagnosis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
2.
Mol Cancer Ther ; 18(2): 289-300, 2019 02.
Article in English | MEDLINE | ID: mdl-30482853

ABSTRACT

BRAF inhibitors (BRAFi) are used to treat patients with melanoma harboring the V600E mutation. However, resistance to BRAFi is inevitable. Here, we identified sphingosine 1-phosphate (S1P) receptors as regulators of BRAFV600E-mutant melanoma cell-autonomous resistance to BRAFi. Moreover, our results reveal a distinct sphingolipid profile, that is, a tendency for increased very long-chain ceramide species, in the plasma of patients with melanoma who achieve a response to BRAFi therapy as compared with patients with progressive disease. Treatment with BRAFi resulted in a strong decrease in S1PR1/3 expression in sensitive but not in resistant cells. Genetic and pharmacologic interventions, that increase ceramide/S1P ratio, downregulated S1PR expression and blocked BRAFi-resistant melanoma cell growth. This effect was associated with a decreased expression of MITF and Bcl-2. Moreover, the BH3 mimetic ABT-737 improved the antitumor activity of approaches targeting S1P-metabolizing enzymes in BRAFi-resistant melanoma cells. Collectively, our findings indicate that targeting the S1P/S1PR axis could provide effective therapeutic options for patients with melanoma who relapse after BRAFi therapy.


Subject(s)
Biphenyl Compounds/administration & dosage , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/administration & dosage , Melanoma/drug therapy , Nitrophenols/administration & dosage , Receptors, Lysosphingolipid/metabolism , Sphingolipids/blood , Sulfonamides/administration & dosage , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Humans , Lysophospholipids/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Nitrophenols/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Piperazines/administration & dosage , Piperazines/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Sulfonamides/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
3.
Acta Derm Venereol ; 99(2): 206-210, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30393817

ABSTRACT

Antibodies targeting immune checkpoints were recently approved for metastatic melanoma. However, not all patients will respond to the treatment and some will experience grade III-IV immune-related adverse events. Therefore, early identification of non-responder patients would greatly aid clinical practice. Detection of circulating tumour DNA (ctDNA) is a non-invasive approach to monitor tumour response. Digital droplet PCR was used to quantify BRAF and NRAS mutations in the plasma of patients with metastatic melanoma treated with immunotherapy. In 16 patients, ctDNA variations mirrored tumour response (p = 0.034) and ctDNA augmentation during follow-up detected tumour progression with 100% specificity. In 13 patients, early ctDNA variation was associated with clinician decision at first evaluation (p = 0.0046), and early ctDNA increase with shorter progression-free survival (median 21 vs. 145 days; p = 0.001). Monitoring ctDNA variations early during immunotherapy may help clinicians rapidly to discriminate non-responder patients, allow early adaptation of therapeutic strategies, and reduce exposure to ineffective, expensive treatment.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Immunotherapy/methods , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Disease Progression , Female , Humans , Male , Melanoma/blood , Melanoma/genetics , Melanoma/immunology , Middle Aged , Progression-Free Survival , Proof of Concept Study , Retrospective Studies , Skin Neoplasms/blood , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Time Factors
4.
Cell Cycle ; 16(21): 2108-2118, 2017.
Article in English | MEDLINE | ID: mdl-28933587

ABSTRACT

It has become more and more evident that the BCL-2 family proteins mediate a wide range of non-apoptotic functions. The pro-apoptotic BAX protein has been reported in interphasic nuclei. Whether the nuclear form of BAX could be involved in non-apoptotic function is still unknown. Our study showed for the first time that BAX was associated with chromatin in vitro. Next, we used gain and loss of function approaches to decipher the potential role of nuclear BAX in non-apoptotic cells. In vitro, nuclear BAX promoted cell proliferation in lung epithelial cells and primary human lung fibroblasts by modulating CDKN1A expression. Interestingly, BAX occupancy of CDKN1A promoter was specifically enriched close to the transcription-starting site. Nuclear BAX also modulated the basal myofibroblastic differentiation and migration of primary human lung fibroblasts. Finally, BAX nuclear localization was associated in vivo with the remodelling of lung parenchyma during development, tumorigenesis as well as fibrosis compared to control adult human lungs. Hence, our study established for the first time, a strong link between the nuclear localization of the pro-apoptotic BAX protein and key basic cellular functions in the non-apoptotic setting.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Interphase , bcl-2-Associated X Protein/metabolism , Apoptosis/physiology , Cell Nucleus/metabolism , Fibroblasts/metabolism , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L615-29, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26773067

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by an accumulation of extracellular matrix proteins and fibroblasts in the distal airways. Key developmental lung signaling pathways are reactivated in IPF. For instance, fibroblast growth factor 9 (FGF9) and FGF18, involved in epithelial-mesenchymal interactions, are critical for lung development. We evaluated the expression of FGF9, FGF18, and FGF receptors (FGFRs) in lung tissue from controls and IPF patients and assessed their effect on proliferation, survival, migration, and differentiation of control and IPF human lung fibroblasts (HLFs). FGF9, FGF18, and all FGFRs were present in the remodeled alveolar epithelium close to the fibroblast foci in IPF lungs. FGFR3 was generally detected in fibroblast foci by immunohistochemistry. In vitro, HLFs mainly expressed mesenchyme-associated FGFR isoforms (FGFR1c and FGFR3c) and FGFR4. FGF9 did not affect fibroblast proliferation, whereas FGF18 inhibited cell growth in control fibroblasts. FGF9 and FGF18 decreased Fas-ligand-induced apoptosis in control but not in IPF fibroblasts. FGF9 prevented transforming growth factor ß1-induced myofibroblast differentiation. FGF9 and FGF18 increased the migratory capacities of HLF, and FGF9 actively modulated matrix metalloproteinase activity. In addition, FGFR3 inhibition by small interfering RNA impacted p-ERK activation by FGF9 and FGF18 and their effects on differentiation and migration. These results identify FGF9 as an antiapoptotic and promigratory growth factor on HLF, maintaining fibroblasts in an undifferentiated state. The biological effects of FGF9 and FGF18 were partially driven by FGFR3. FGF18 was a less potent molecule. Both growth factors likely contribute to the fibrotic process in vivo.


Subject(s)
Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/physiology , Myofibroblasts/physiology , Receptors, Fibroblast Growth Factor/metabolism , Aged , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis , Lung/metabolism , Lung/pathology , Middle Aged
6.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L838-47, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25260753

ABSTRACT

Aberrant expression of master phenotype regulators or alterations in their downstream pathways in lung fibroblasts may play a central role in idiopathic pulmonary fibrosis (IPF). Interrogating IPF fibroblast transcriptome datasets, we identified Forkhead Box F1 (FOXF1), a DNA-binding protein required for lung development, as a candidate actor in IPF. Thus we determined FOXF1 expression levels in fibroblasts cultured from normal or IPF lungs in vitro, and explored FOXF1 functions in these cells using transient and stable loss-of-function and gain-of-function models. FOXF1 mRNA and protein were expressed at higher levels in IPF fibroblasts compared with normal fibroblasts (mRNA: +44%, protein: +77%). Immunohistochemistry showed FOXF1 expression in nuclei of bronchial smooth muscle cells, endothelial cells, and lung fibroblasts including fibroblastic foci of IPF lungs. In normal lung fibroblasts, FOXF1 repressed cell growth and expression of collagen-1 (COL1) and actin-related protein 2/3 complex, subunit 2 (ARPC2). ARPC2 knockdown inhibited cell growth and COL1 expression, consistent with FOXF1 acting in part through ARPC2 repression. In IPF fibroblasts, COL1 and ARPC2 repression by FOXF1 was blunted, and FOXF1 did not repress growth. FOXF1 expression was induced by the antifibrotic mediator prostaglandin E2 and repressed by the profibrotic cytokine transforming growth factor-ß1 in both normal and IPF lung fibroblasts. Ex vivo, FOXF1 knockdown conferred CCL-210 lung fibroblasts the ability to implant in uninjured mouse lungs. In conclusion, FOXF1 functions and regulation were consistent with participation in antifibrotic pathways. Alterations of pathways downstream of FOXF1 may participate to fibrogenesis in IPF fibroblasts.


Subject(s)
Actin-Related Protein 2-3 Complex/biosynthesis , Collagen Type I/biosynthesis , Forkhead Transcription Factors/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Actin-Related Protein 2-3 Complex/genetics , Animals , Apoptosis , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Dinoprostone/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/transplantation , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Humans , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
7.
Am J Respir Cell Mol Biol ; 51(1): 11-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24450438

ABSTRACT

Idiopathic pulmonary fibrosis has been associated with the reactivation of developmental pathways, notably the Hedgehog-Glioma-associated oncogene homolog (GLI) pathway. In this study, we determined whether the Hedgehog pathway was activated in bleomycin-induced lung injury in mice, and whether targeting the Hedgehog-Gli pathway could decrease bleomycin-induced lung fibrosis. After intratracheal injection of bleomycin on Day 0, C57Bl6 mice received GDC-0449 (an inhibitor of Smoothened, the transducer of the pathway), or 2,2'-[[Dihydro-2-(4-pyridinyl)-1,3(2H,4H)-pyrimidinediyl]bis(methylene)]bis[N,N dimethylbenzenamine (GANT61; an inhibitor of GLI transcription factors in the nucleus), from Day 7 to Day 13. At Day 14, whole-lung homogenates were obtained for morphological analysis, assessment of cell apoptosis and proliferation, collagen quantification, and evaluation of profibrotic (transforming growth factor-ß, connective tissue growth factor, plasminogen activator inhibitor 1, vascular endothelial growth factor-A) and proinflammatory mediators (IL-1ß) expression. We showed that the Hedgehog pathway was activated in bleomycin-induced lung fibrosis on Day 14 after injury, with an increased lung expression of the ligand, Sonic Hedgehog, and with increased messenger RNA expression and nuclear localization of GLI1 and GLI2. Inhibition of Smoothened with GDC-0449 did not influence the development of bleomycin-induced lung fibrosis. By contrast, the inhibition of GLI activity with GANT61 decreased lung fibrosis and lung collagen accumulation, and promoted an antifibrotic and anti-inflammatory environment. Our results identify the hedgehog-Gli pathway as a profibrotic pathway in experimental fibrosis. Inhibition of the Hedgehog-Gli pathway at the level of GLI transcriptional activity could be a therapeutic option in fibrotic lung diseases.


Subject(s)
Anilides/pharmacology , Bleomycin/toxicity , Glioma/drug therapy , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Collagen/genetics , Collagen/metabolism , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/pathology , Immunoenzyme Techniques , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smoothened Receptor , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1
8.
Antioxid Redox Signal ; 18(1): 66-79, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22703534

ABSTRACT

AIMS: Oxidants have been implicated in the pathophysiology of idiopathic pulmonary fibrosis (IPF), especially in myofibroblastic differentiation. We aimed at testing the hypothesis that nuclear factor erythroid 2-related factor 2 (Nrf2), the main regulator of endogenous antioxidant enzymes, is involved in fibrogenesis via myofibroblastic differentiation. Fibroblasts were cultured from the lungs of eight controls and eight IPF patients. Oxidants-antioxidants balance, nuclear Nrf2 expression, and fibroblast phenotype (α-smooth muscle actin and collagen I expression, proliferation, migration, and contraction) were studied under basal conditions and after Nrf2 knockdown or activation by Nrf2 or Keap1 siRNA transfection. The effects of sulforaphane (SFN), an Nrf2 activator, on the fibroblast phenotype were tested under basal and pro-fibrosis conditions (transforming growth factor ß [TGF-ß]). RESULTS: Decreased Nrf2 expression was associated with a myofibroblast phenotype in IPF compared with control fibroblasts. Nrf2 knockdown induced oxidative stress and myofibroblastic differentiation in control fibroblasts. Conversely, Nrf2 activation increased antioxidant defences and myofibroblastic dedifferentation in IPF fibroblasts. SFN treatment decreased oxidants, and induced Nrf2 expression, antioxidants, and myofibroblastic dedifferentiation in IPF fibroblasts. SFN inhibited TGF-ß profibrotic deleterious effects in IPF and control fibroblasts and restored antioxidant defences. Nrf2 knockdown abolished SFN antifibrosis effects, suggesting that they were Nrf2 mediated. INNOVATION AND CONCLUSION: Our findings confirm that decreased nuclear Nrf2 plays a role in myofibroblastic differentiation and that SFN induces human pulmonary fibroblast dedifferentiation in vitro via Nrf2 activation. Thus, Nrf2 could be a novel therapeutic target in IPF.


Subject(s)
Cell Dedifferentiation , Cell Nucleus/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Aldehydes/metabolism , Animals , Becaplermin , Cells, Cultured , Collagen Type I/metabolism , Epoxide Hydrolases/metabolism , Gene Knockdown Techniques , Heme Oxygenase-1/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Isothiocyanates , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/physiology , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/physiology , Oxidative Stress , Phenotype , Proto-Oncogene Proteins c-sis/physiology , RNA, Small Interfering/genetics , Sulfoxides , Thiocyanates/pharmacology , Transforming Growth Factor beta/physiology
9.
Am J Pathol ; 181(6): 2126-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23031257

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown cause. Key signaling developmental pathways are aberrantly expressed in IPF. The hedgehog pathway plays a key role during fetal lung development and may be involved in lung fibrogenesis. We determined the expression pattern of several Sonic hedgehog (SHH) pathway members in normal and IPF human lung biopsies and primary fibroblasts. The effect of hedgehog pathway inhibition was assayed by lung fibroblast proliferation and differentiation with and without transforming growth factor (TGF)-ß1. We showed that the hedgehog pathway was reactivated in the IPF lung. Importantly, we deciphered the cross talk between the hedgehog and TGF-ß pathway in human lung fibroblasts. TGF-ß1 modulated the expression of key components of the hedgehog pathway independent of Smoothened, the obligatory signal transducer of the pathway. Smoothened was required for TGF-ß1-induced myofibroblastic differentiation of control fibroblasts, but differentiation of IPF fibroblasts was partially resistant to Smoothened inhibition. Furthermore, functional hedgehog pathway machinery from the primary cilium, as well as GLI-dependent transcription in the nucleus, was required for the TGF-ß1 effects on normal and IPF fibroblasts during myofibroblastic differentiation. These data identify the GLI transcription factors as potential therapeutic targets in lung fibrosis.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cilia/drug effects , Cilia/pathology , Female , Gene Expression Regulation/drug effects , Hedgehog Proteins/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunohistochemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Middle Aged , Models, Biological , Myofibroblasts/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Veratrum Alkaloids/pharmacology
10.
J Exp Med ; 208(5): 973-86, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21482699

ABSTRACT

Angiogenesis plays an essential role in several diseases of the eye and in the growth of solid tumors, but existing antiangiogenic therapies have limited benefits in several cases. We report the antiangiogenic effects of a monoclonal antibody, CL1-R2, in several animal models of neovascularization. CL1-R2 recognizes human CD160, a membrane receptor which is conserved in various mammal species. We show that CD160 is expressed on the endothelial cells of newly formed blood vessels in human colon carcinoma and mouse B16 melanoma but not in vessels of healthy tissues. CL1-R2 reduced fibroblast growth factor 2-induced neovascularization in the rabbit cornea, in a mouse model of oxygen-induced retinopathy, and in a mouse Matrigel plug assay. Treatment of B16 melanoma-bearing mice with CL1-R2 combined with cyclophosphamide chemotherapy caused regression of the tumor vasculature and normalization of the remaining vessels as shown by Doppler ultrasonography, intravital microscopy, and histology. These studies validate CD160 as a potential new target in cases of human pathological ocular and tumor neoangiogenesis that do not respond or become resistant to existing antiangiogenic drugs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD/metabolism , Neovascularization, Pathologic/drug therapy , Receptors, Immunologic/metabolism , Animals , Antigens, CD/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Corneal Neovascularization/drug therapy , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Cyclophosphamide/pharmacology , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Melanoma/blood supply , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rabbits , Receptors, Immunologic/genetics , Retinal Diseases/drug therapy , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...