Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Sci Transl Med ; 12(565)2020 10 14.
Article En | MEDLINE | ID: mdl-33055240

Defects in tumor-intrinsic interferon (IFN) signaling result in failure of immune checkpoint blockade (ICB) against cancer, but these tumors may still maintain sensitivity to T cell-based adoptive cell therapy (ACT). We generated models of IFN signaling defects in B16 murine melanoma observed in patients with acquired resistance to ICB. Tumors lacking Jak1 or Jak2 did not respond to ICB, whereas ACT was effective against Jak2 KO tumors, but not Jak1 KO tumors, where both type I and II tumor IFN signaling were defective. This was a direct result of low baseline class I major histocompatibility complex (MHC I) expression in B16 and the dependency of MHC I expression on either type I or type II IFN signaling. We used genetic and pharmacologic approaches to uncouple this dependency and restore MHC I expression. Through independent mechanisms, overexpression of NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) and intratumoral delivery of BO-112, a potent nanoplexed version of polyinosinic:polycytidylic acid (poly I:C), each restored the efficacy of ACT against B16-Jak1 KO tumors. BO-112 activated double-stranded RNA (dsRNA) sensing (via protein kinase R and Toll-like receptor 3) and induced MHC I expression via nuclear factor κB, independent of both IFN signaling and NLRC5. In summary, we demonstrated that in the absence of tumor IFN signaling, MHC I expression is essential and sufficient for the efficacy of ACT. For tumors lacking MHC I expression due to deficient IFN signaling, activation of dsRNA sensors by BO-112 affords an alternative approach to restore the efficacy of ACT.


Antigen Presentation , Interferon-gamma , Animals , Humans , Immunotherapy , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Mice , NF-kappa B , Signal Transduction
2.
Curr Drug Targets ; 17(15): 1714-1727, 2016.
Article En | MEDLINE | ID: mdl-26953245

Antiangiogenic resistance is a major problem in cancer therapeutics. Preclinical research has identified several compensatory proangiogenic pathways that arise upon vascular endothelial growth factor inhibition, several of which have led to the development of novel drugs. However, the combination of two or more targeted agents in the angiogenesis system is hampered by toxicity, as the system is involved in normal physiology. We propose a different approach for improving the efficacy of this drug class, which takes advantage of aberrant cancer metabolism. Several features distinguish cancer metabolism from that of normal cells, including increased glycolysis, glutaminolysis, and pentose-phosphate shunt, as well as an anaplerotic shift of the Krebs cycle. In addition, these aberrations are driven by most of the common mutations that can be targeted by drugs. Antiangiogenics may hamper the ability of cancer to sustain aberrant metabolism due to their impacts on nutrient and oxygen supplies, and thus they may induce some metabolic pathways to become essential for tumor survival (induced essentiality or contextual lethality, a type of synthetic lethality). Thus, some metabolic and signaling pathways that are otherwise nonessential may induce synthetic lethality when inhibited in combination with antiangiogenics. The key problems, however, are interpatient and intratumor heterogeneity, as not all patients with the same tumor type show the same metabolic traits and the same metabolic reprogramming in response to antiangiogenics. With each cancer there are heterogeneous hypoxic areas. Integrating dynamic tracking of metabolism may allow us to tailor our choices of companion drugs with antiangiogenics, taking advantage of window-of-opportunity designs.


Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm , Neoplasms/metabolism , Synthetic Lethal Mutations , Animals , Cell Proliferation/drug effects , Cell Survival , Gene Regulatory Networks/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine , Signal Transduction/drug effects
3.
Cancer Res ; 75(15): 3032-42, 2015 Aug 01.
Article En | MEDLINE | ID: mdl-26069249

Immune escape is a hallmark of cancer, but whether it relies upon extrinsic immune-selective pressure or is inherently orchestrated by oncogenic pathways is unresolved. To address this question, we took advantage of an in vitro model of sequentially transformed human mesenchymal stem cells (hMSC). Neoplastic transformation in this model increased the natural immune-evasive properties of hMSC, both by reducing their immunogenicity and by increasing their capacity to inhibit mitogen-driven T-cell proliferation. We also found that IFNγ signaling was globally affected in transformed hMSC. As a consequence, the natural inhibitory effect of hMSC on T-cell proliferation switched from an inducible mechanism depending on IFNγ signaling and mediated by indoleamine 2,3-dioxygenase to a constitutive mechanism that relied upon IL1ß involving both secreted and membrane-expressed molecules. After transformation, increased IL1ß expression both sustained the immunosuppressive properties of hMSC and increased their tumorigenicity. Thus, in this model system, IL1ß acted as intrinsic inflammatory mediator that exerted an autocrine influence on tumor growth by coordinately linking immune escape and tumorigenicity. Collectively, our findings show how oncogenes directly orchestrate inflammation and immune escape to drive the multistep process of cancer progression, independently of any need for immunoediting in the tumor microenvironment.


Inflammation/pathology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Humans , Immune Evasion/physiology , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Mice, Nude , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
4.
Cell Immunol ; 289(1-2): 174-84, 2014.
Article En | MEDLINE | ID: mdl-24841856

Stem cells from mesenchymal origin (MSC) exert a plethora of immunomodulatory effects. We created a neoplastic model based on in vitro step-wise transformation to assess whether oncogenic pathways have the capacity to mould the cross-talk of MSC and lymphocytes. Neoplastic MSC exhibit an increased inhibitory effect on T cell proliferation, either directly or mediated by myeloid derived suppressor cells. Additionally, transformation of MSC enhances T cell apoptosis without reducing either the percentage of CD25 expressing cells or the level of this protein expression. Malignant transformation drives MSC to lose dependency on nitric oxide for immunosuppression whilst increasing the constitutive production of PGE2. Our results indicate that oncogenesis tunes the interplay between MSC and immune cells, favoring cancer immune evasion.


Cell Communication/immunology , Cell Transformation, Neoplastic/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Dinoprostone/biosynthesis , Female , Immunosuppression Therapy , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Tumor Escape/immunology , Tumor Suppressor Protein p53/genetics
5.
Mol Cancer ; 13: 20, 2014 Feb 03.
Article En | MEDLINE | ID: mdl-24491031

BACKGROUND: The transcription factor Nrf2 is a key regulator of the cellular antioxidant response, and its activation by chemoprotective agents has been proposed as a potential strategy to prevent cancer. However, activating mutations in the Nrf2 pathway have been found to promote tumorigenesis in certain models. Therefore, the role of Nrf2 in cancer remains contentious. METHODS: We employed a well-characterized model of stepwise human mesenchymal stem cell (MSC) transformation and breast cancer cell lines to investigate oxidative stress and the role of Nrf2 during tumorigenesis. The Nrf2 pathway was studied by microarray analyses, qRT-PCR, and western-blotting. To assess the contribution of Nrf2 to transformation, we established tumor xenografts with transformed MSC expressing Nrf2 (n = 6 mice per group). Expression and survival data for Nrf2 in different cancers were obtained from GEO and TCGA databases. All statistical tests were two-sided. RESULTS: We found an accumulation of reactive oxygen species during MSC transformation that correlated with the transcriptional down-regulation of antioxidants and Nrf2-downstream genes. Nrf2 was repressed in transformed MSC and in breast cancer cells via oncogene-induced activation of the RAS/RAF/ERK pathway. Furthermore, restoration of Nrf2 function in transformed cells decreased reactive oxygen species and impaired in vivo tumor growth (P = 0.001) by mechanisms that included sensitization to apoptosis, and a decreased hypoxic/angiogenic response through HIF-1α destabilization and VEGFA repression. Microarray analyses showed down-regulation of Nrf2 in a panel of human tumors and, strikingly, low Nrf2 expression correlated with poorer survival in patients with melanoma (P = 0.0341), kidney (P = 0.0203) and prostate (P = 0.00279) cancers. CONCLUSIONS: Our data indicate that oncogene-induced Nrf2 repression is an adaptive response for certain cancers to acquire a pro-oxidant state that favors cell survival and in vivo tumor growth.


Cell Transformation, Neoplastic/metabolism , Mesenchymal Stem Cells/metabolism , NF-E2-Related Factor 2/biosynthesis , Neoplasms/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Down-Regulation , Heterografts , Humans , Mesenchymal Stem Cells/pathology , Mice , Neoplasms/genetics , Neoplasms/mortality , Oligonucleotide Array Sequence Analysis , Oxidative Stress/physiology , Proportional Hazards Models , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Survival Analysis
6.
Stem Cells ; 31(10): 2061-72, 2013 Oct.
Article En | MEDLINE | ID: mdl-23836491

Increasing evidence supports that mesenchymal stromal/stem cells (MSCs) may represent the target cell for sarcoma development. Although different sarcomas have been modeled in mice upon expression of fusion oncogenes in MSCs, sarcomagenesis has not been successfully modeled in human MSCs (hMSCs). We report that FUS-CHOP, a hallmark fusion gene in mixoid liposarcoma (MLS), has an instructive role in lineage commitment, and its expression in hMSC sequentially immortalized/transformed with up to five oncogenic hits (p53 and Rb deficiency, hTERT over-expression, c-myc stabilization, and H-RAS(v12) mutation) drives the formation of serially transplantable MLS. This is the first model of sarcoma based on the expression of a sarcoma-associated fusion protein in hMSC, and allowed us to unravel the differentiation processes and signaling pathways altered in the MLS-initiating cells. This study will contribute to test novel therapeutic approaches and constitutes a proof-of-concept to use hMSCs as target cell for modeling other fusion gene-associated human sarcomas.


Liposarcoma, Myxoid/metabolism , Mesenchymal Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , RNA-Binding Protein FUS/metabolism , Transcription Factor CHOP/metabolism , Adipogenesis , Animals , Carcinogenesis/metabolism , Cell Line, Transformed , Gene Expression , Humans , Liposarcoma, Myxoid/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein FUS/genetics , Signal Transduction , Transcription Factor CHOP/genetics , Transcriptome
7.
Cell Cycle ; 10(22): 3912-9, 2011 Nov 15.
Article En | MEDLINE | ID: mdl-22067611

INTRODUCTION: Residing within human dental pulp are cells of an ectomesenchymal origin which have the potential to differentiate into odontoblast-like cells. These cells have a limited growth potential owing to the effects of cell senescence. This study examines the effects of immortalizing odontoblast-like cells on cell proliferation and mineralization by comparing transformed dental pulp stem cells (tDPSCs) and non-transformed dental pulp stem cells (nDPSCs). RESULTS: With the exogenous expression of hTERT, tDPSCs maintained a continued expression of odontogenic markers for cell proliferation and mineralization (ALP, COL-1, DMP-1, DSPP, OCN amd OPN)as did nDPScs. Oncoprotein expression was seen in both groups except for a noted absence of p16 in the tDPSCs. nDPSCs also showed lower levels of total ALP and DNA activity in comparison to tDPSCs when assayed as well as low telomerase activity readings. METHODS: Using a retroviral vector, exogenous human telomerase reverse transcriptase (hTERT) was expressed in tDPSCs. Both cell groups were cultured and their telomerase activities is determined using a telomerase quantification assay. Also examined were the expression of genes involved in proliferation and mineralization such as human alkaline phosphatase (ALP), ß-actin, collagen 1 (col-1), core binding factor (cbfa-1), dentin matrix protein (DMP-1), dentin sialophosphoprotein (DSPP), GAPDH, hTERT, osteocalcin (OCN), osteopontin (OPN) as well as oncoproteins involved in senescence (p16, p21 and p53) using RT-PCR. DNA and alkaline phosphatase activity was assayed in both cell groups. CONCLUSIONS: These results indicate maintainance of odontoblast-like differentiation characteristics after retroviral transformation with hTERT and suggest a possible link with a reduced p16 expression.


Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dental Pulp/metabolism , Telomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Colorimetry , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cytoskeleton/ultrastructure , Dental Pulp/cytology , Dental Pulp/ultrastructure , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/pharmacology , Transduction, Genetic , Tumor Suppressor Protein p53/analysis , beta-Galactosidase/analysis
8.
Clin Cancer Res ; 17(6): 1535-45, 2011 Mar 15.
Article En | MEDLINE | ID: mdl-21278247

PURPOSE: The importance of genetic and epigenetic alterations maybe in their aggregate role in altering core pathways in tumorigenesis. EXPERIMENTAL DESIGN: Merging genome-wide genomic and epigenomic alterations, we identify key genes and pathways altered in colorectal cancers (CRC). DNA methylation analysis was tested for predicting survival in CRC patients using Cox proportional hazard model. RESULTS: We identified 29 low frequency-mutated genes that are also inactivated by epigenetic mechanisms in CRC. Pathway analysis showed the extracellular matrix (ECM) remodeling pathway is silenced in CRC. Six ECM pathway genes were tested for their prognostic potential in large CRC cohorts (n = 777). DNA methylation of IGFBP3 and EVL predicted for poor survival (IGFBP3: HR = 2.58, 95% CI: 1.37-4.87, P = 0.004; EVL: HR = 2.48, 95% CI: 1.07-5.74, P = 0.034) and simultaneous methylation of multiple genes predicted significantly worse survival (HR = 8.61, 95% CI: 2.16-34.36, P < 0.001 for methylation of IGFBP3, EVL, CD109, and FLNC). DNA methylation of IGFBP3 and EVL was validated as a prognostic marker in an independent contemporary-matched cohort (IGFBP3 HR = 2.06, 95% CI: 1.04-4.09, P = 0.038; EVL HR = 2.23, 95% CI: 1.00-5.0, P = 0.05) and EVL DNA methylation remained significant in a secondary historical validation cohort (HR = 1.41, 95% CI: 1.05-1.89, P = 0.022). Moreover, DNA methylation of selected ECM genes helps to stratify the high-risk stage 2 colon cancers patients who would benefit from adjuvant chemotherapy (HR: 5.85, 95% CI: 2.03-16.83, P = 0.001 for simultaneous methylation of IGFBP3, EVL, and CD109). CONCLUSIONS: CRC that have silenced genes in ECM pathway components show worse survival suggesting that our finding provides novel prognostic biomarkers for CRC and reflects the high importance of integrative analyses linking genetic and epigenetic abnormalities with pathway disruption in cancer.


Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Epigenomics , Genomics , Aged , Biomarkers, Tumor , Cell Line, Tumor , Chemotherapy, Adjuvant/methods , DNA Methylation , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models , Treatment Outcome
9.
Carcinogenesis ; 31(10): 1854-62, 2010 Oct.
Article En | MEDLINE | ID: mdl-20466760

Genome-wide DNA hypomethylation was one of the first epigenetic alterations described in cancer cells. However, the cause of this hypomethylation is still poorly understood. We have previously developed a line of primary mesenchymal stem cells (MSC, the putative origin of various types of sarcoma) in which five oncogenic steps toward a fully transformed state are sequentially introduced including: human telomerase, inactivation of p53 and pRb tumor suppressor genes and activation of the oncogenes c-Myc and H-Ras. We hypothesized that DNA hypomethyation would occur during stepwise transformation of MSC and could be a model to investigate the mechanism of global hypomethylation in cancer. Here we show, firstly, that satellite-2 and long interspersed nuclear element 1 repetitive elements became hypomethylated (54 and 30% reduction, respectively) on the introduction of oncogenic H-Ras after the final step of transformation. Secondly, we observed hypomethylation only after 4 weeks in culture following the introduction of H-Ras, suggesting a gradual loss of methylation. Finally, using an inducible estrogen receptor-Ras fusion construct, we were able to transform MSC's in the absence of detectable hypomethylation, suggesting that it was not a requirement for transformation. These studies show that DNA hypomethylation can occur late during stepwise transformation, although in vitro transformation could also take place in the absence of hypomethylation. These data support the hypothesis that DNA hypomethylation occurs via a gradual mechanism and is not a requirement for transformation.


Cell Transformation, Neoplastic , DNA Methylation , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Acetyltransferases/genetics , Animals , Cells, Cultured , Genes, ras , Humans , Mice
10.
Mol Cancer Ther ; 8(1): 249-60, 2009 Jan.
Article En | MEDLINE | ID: mdl-19139135

Since the sequencing of the human genome, recent efforts in cancer drug target discovery have focused more on the identification of novel functions of known genes and the development of more appropriate tumor models. In the present study, we investigated in vitro transformed human adult mesenchymal stem cells (MSC) to identify novel candidate cancer drug targets by analyzing the transcriptional profile of known enzymes compared with non-transformed MSC. The identified enzymes were compared with published cancer gene expression data sets. Surprisingly, the majority of up-regulated enzymes are already known cancer drug targets or act within known druggable pathways. Only three enzymes (RNASEH2A, ADARB1, and PPAP2C) are potentially novel targets that are up-regulated in transformed MSC and expressed in numerous carcinomas and sarcomas. We confirmed the overexpression of RNASEH2A, PPAP2C, and ADARB1 in transformed MSC, transformed fibroblasts, and cancer cell lines MCF7, SK-LMS1, MG63, and U2OS. In functional assays, we show that small interfering RNA knockdown of RNASEH2A inhibits anchorage-independent growth but does not alter in vitro proliferation of cancer cell lines, normal MSC, or normal fibroblasts. Knockdown of PPAP2C impaired anchorage-dependent in vitro growth of cancer cell lines and impaired the in vitro growth of primary MSC but not differentiated human fibroblasts. We show that the knockdown of PPAP2C decreases cell proliferation by delaying entry into S phase of the cell cycle and is transcriptionally regulated by p53. These in vitro data validate PPAP2C and RNASEH2A as putative cancer targets and endorse this in silico approach for identifying novel candidates.


Adenosine Deaminase/genetics , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic , Neoplasms/genetics , Phosphatidate Phosphatase/genetics , Ribonuclease H/genetics , Stem Cells/metabolism , Adenosine Deaminase/metabolism , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Genomics , Humans , Neoplasms/metabolism , Phosphatidate Phosphatase/metabolism , RNA-Binding Proteins , Ribonuclease H/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
11.
Proc Natl Acad Sci U S A ; 104(15): 6223-8, 2007 Apr 10.
Article En | MEDLINE | ID: mdl-17384149

An increased dependency on glycolysis for ATP production is considered to be a hallmark of tumor cells. Whether this increase in glycolytic activity is due mainly to inherent metabolic alterations or to the hypoxic microenvironment remains controversial. Here we have transformed human adult mesenchymal stem cells (MSC) using genetic alterations as described for differentiated cells. Our data suggest that MSC require disruption of the same pathways as have been shown for differentiated cells to confer a fully transformed phenotype. Furthermore, we found that MSC are more glycolytic than primary human fibroblasts and, in contrast to differentiated cells, do not depend on increased aerobic glycolysis for ATP production during transformation. These data indicate that aerobic glycolysis (the Warburg effect) is not an intrinsic component of the transformation of adult stem cells, and that oncogenic adaptation to bioenergetic requirements, in some circumstances, may also rely on increases in oxidative phosphorylation. We did find, however, a reversible increase in the transcription of glycolytic enzymes in tumors generated by transformed MSC, indicating this is a secondary phenomenon resulting from adaptation of the tumor to its microenvironment.


Adenosine Triphosphate/biosynthesis , Cell Transformation, Neoplastic/metabolism , Energy Metabolism/physiology , Mesenchymal Stem Cells/physiology , Oxidative Phosphorylation , Adult , Blotting, Western , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Glucose/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glycolysis/physiology , Humans , Immunophenotyping , Lactic Acid/metabolism , Male , Mesenchymal Stem Cells/metabolism , NADP/metabolism
12.
EMBO J ; 24(17): 3104-16, 2005 Sep 07.
Article En | MEDLINE | ID: mdl-16096642

D cyclins (D1, D2 and D3) and their catalytic subunits (cyclin-dependent kinases cdk4 and cdk6) have a facilitating, but nonessential, role in cell cycle entry. Tissue-specific functions for D-type cyclins and cdks have been reported; however, the biochemical properties of these kinases are indistinguishable. We report that an F box protein, Fbxo7, interacted with cellular and viral D cyclins and distinguished among the cdks that bind D-type cyclins, specifically binding cdk6, in vitro and in vivo. Fbxo7 specifically regulated D cyclin/cdk6 complexes: Fbxo7 knockdown decreased cdk6 association with cyclin and its overexpression increased D cyclin/cdk6 activity and E2F activity. Fbxo7 interacted with p27, but its enhancement of cyclin D/cdk6 activity was p21/p27 independent. Fbxo7 overexpression transformed murine fibroblasts, rendering them tumorigenic in athymic nude mice. Transformed phenotypes were dependent on cdk6, as knockdown of cdk6 reversed them. Fbxo7 was highly expressed in epithelial tumors, but not in normal tissues, suggesting that it may have a proto-oncogenic role in human cancers.


Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , F-Box Proteins/metabolism , Active Transport, Cell Nucleus , Adenocarcinoma/metabolism , Animals , Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Cyclin D , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cytoplasm/metabolism , F-Box Proteins/genetics , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Mice , Protein Binding , RNA, Small Interfering/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Santa Fe; Castellvi; 1978. 405 p. ilus, map, tab.
Monography Es | BINACIS | ID: biblio-1188126
14.
Santa Fe; Castellvi; 1978. 405 p. ilus, mapas, tab. (59892).
Monography Es | BINACIS | ID: bin-59892
...