Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552609

ABSTRACT

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Subject(s)
Neoplasms , Humans , Carcinogenesis , Microbiota , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Obesity/complications , Quality of Life
2.
Cell ; 181(2): 219-222, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302564

ABSTRACT

Mounting evidence indicates that the nervous system plays a central role in cancer pathogenesis. In turn, cancers and cancer therapies can alter nervous system form and function. This Commentary seeks to describe the burgeoning field of "cancer neuroscience" and encourage multidisciplinary collaboration for the study of cancer-nervous system interactions.


Subject(s)
Neoplasms/metabolism , Nervous System/metabolism , Humans , Neurosciences
3.
Cell ; 172(4): 643-644, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29425485

ABSTRACT

While functional heterogeneity of fibroblastic cells populating the tumor microenvironment is increasingly recognized, lack of definitive markers complicates elucidation of roles among ostensibly distinctive fibroblastic states. In this issue of Cell, Su et al. characterize a new pro-tumorigenic cancer-associated fibroblast subset mediating chemoresistance defined and driven by a novel signaling pathway.


Subject(s)
Cancer-Associated Fibroblasts , Tumor Microenvironment , Carcinogenesis , Fibroblasts , Humans , Signal Transduction
4.
Immunity ; 56(1): 162-179.e6, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36630914

ABSTRACT

Immunotherapies have shown remarkable, albeit tumor-selective, therapeutic benefits in the clinic. Most patients respond transiently at best, highlighting the importance of understanding mechanisms underlying resistance. Herein, we evaluated the effects of the engineered immunocytokine PD1-IL2v in a mouse model of de novo pancreatic neuroendocrine cancer that is resistant to checkpoint and other immunotherapies. PD1-IL2v utilizes anti-PD-1 as a targeting moiety fused to an immuno-stimulatory IL-2 cytokine variant (IL2v) to precisely deliver IL2v to PD-1+ T cells in the tumor microenvironment. PD1-IL2v elicited substantial infiltration by stem-like CD8+ T cells, resulting in tumor regression and enhanced survival in mice. Combining anti-PD-L1 with PD1-IL2v sustained the response phase, improving therapeutic efficacy both by reprogramming immunosuppressive tumor-associated macrophages and enhancing T cell receptor (TCR) immune repertoire diversity. These data provide a rationale for clinical trials to evaluate the combination therapy of PD1-IL2v and anti-PD-L1, particularly in immunotherapy-resistant tumors infiltrated with PD-1+ stem-like T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Macrophages , Neoplasms , Animals , Mice , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Immunotherapy/methods , Macrophages/immunology , Macrophages/metabolism , Neoplasms/therapy , Tumor Microenvironment , Antibodies, Bispecific/immunology , Interleukin-2 , Programmed Cell Death 1 Receptor/immunology
5.
Cell ; 153(1): 86-100, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540692

ABSTRACT

Glutamate and its receptor N-methyl-D-aspartate receptor (NMDAR) have been associated with cancer, although their functions are not fully understood. Herein, we implicate glutamate-driven NMDAR signaling in a mouse model of pancreatic neuroendocrine tumorigenesis (PNET) and in selected human cancers. NMDAR was upregulated at the periphery of PNET tumors, particularly invasive fronts. Moreover, elevated coexpression of NMDAR and glutamate exporters correlated with poor prognosis in cancer patients. Treatment of a tumor-derived cell line with NMDAR antagonists impaired cancer cell proliferation and invasion. Flow conditions mimicking interstitial fluid pressure induced autologous glutamate secretion, activating NMDAR and its downstream MEK-MAPK and CaMK effectors, thereby promoting invasiveness. Congruently, pharmacological inhibition of NMDAR in mice with PNET reduced tumor growth and invasiveness. Therefore, beyond its traditional role in neurons, NMDAR may be activated in human tumors by fluid flow consequent to higher interstitial pressure, inducing an autocrine glutamate signaling circuit with resultant stimulation of malignancy.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Glutamic Acid/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment
6.
Nature ; 610(7930): 161-172, 2022 10.
Article in English | MEDLINE | ID: mdl-36171284

ABSTRACT

Expansion and differentiation of antigen-experienced PD-1+TCF-1+ stem-like CD8+ T cells into effector cells is critical for the success of immunotherapies based on PD-1 blockade1-4. Hashimoto et al. have shown that, in chronic infections, administration of the cytokine interleukin (IL)-2 triggers an alternative differentiation path of stem-like T cells towards a distinct population of 'better effector' CD8+ T cells similar to those generated in an acute infection5. IL-2 binding to the IL-2 receptor α-chain (CD25) was essential in triggering this alternative differentiation path and expanding better effectors with distinct transcriptional and epigenetic profiles. However, constitutive expression of CD25 on regulatory T cells and some endothelial cells also contributes to unwanted systemic effects from IL-2 therapy. Therefore, engineered IL-2 receptor ß- and γ-chain (IL-2Rßγ)-biased agonists are currently being developed6-10. Here we show that IL-2Rßγ-biased agonists are unable to preferentially expand better effector T cells in cancer models and describe PD1-IL2v, a new immunocytokine that overcomes the need for CD25 binding by docking in cis to PD-1. Cis binding of PD1-IL2v to PD-1 and IL-2Rßγ on the same cell recovers the ability to differentiate stem-like CD8+ T cells into better effectors in the absence of CD25 binding in both chronic infection and cancer models and provides superior efficacy. By contrast, PD-1- or PD-L1-blocking antibodies alone, or their combination with clinically relevant doses of non-PD-1-targeted IL2v, cannot expand this unique subset of better effector T cells and instead lead to the accumulation of terminally differentiated, exhausted T cells. These findings provide the basis for the development of a new generation of PD-1 cis-targeted IL-2R agonists with enhanced therapeutic potential for the treatment of cancer and chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Receptors, Interleukin-2 , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Infections/drug therapy , Infections/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/agonists , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Interleukin-2/agonists
7.
Cell ; 144(5): 646-74, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376230

ABSTRACT

The hallmarks of cancer comprise six biological capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metabolism and evading immune destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment." Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer.


Subject(s)
Neoplasms/pathology , Neoplasms/physiopathology , Animals , Genomic Instability , Humans , Neoplasm Invasiveness , Neoplasms/metabolism , Signal Transduction , Stromal Cells/pathology
8.
Nature ; 573(7775): 526-531, 2019 09.
Article in English | MEDLINE | ID: mdl-31534217

ABSTRACT

Metastasis-the disseminated growth of tumours in distant organs-underlies cancer mortality. Breast-to-brain metastasis (B2BM) is a common and disruptive form of cancer and is prevalent in the aggressive basal-like subtype, but is also found at varying frequencies in all cancer subtypes. Previous studies revealed parameters of breast cancer metastasis to the brain, but its preference for this site remains an enigma. Here we show that B2BM cells co-opt a neuronal signalling pathway that was recently implicated in invasive tumour growth, involving activation by glutamate ligands of N-methyl-D-aspartate receptors (NMDARs), which is key in model systems for metastatic colonization of the brain and is associated with poor prognosis. Whereas NMDAR activation is autocrine in some primary tumour types, human and mouse B2BM cells express receptors but secrete insufficient glutamate to induce signalling, which is instead achieved by the formation of pseudo-tripartite synapses between cancer cells and glutamatergic neurons, presenting a rationale for brain metastasis.


Subject(s)
Brain Neoplasms/physiopathology , Brain Neoplasms/secondary , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Brain Neoplasms/ultrastructure , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Neoplasm Metastasis , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/ultrastructure , Synaptic Transmission
9.
Proc Natl Acad Sci U S A ; 116(48): 24184-24195, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31704767

ABSTRACT

MicroRNA-mediated gene regulation has been implicated in various diseases, including cancer. This study examined the role of microRNAs (miRNAs) during tumorigenesis and malignant progression of pancreatic neuroendocrine tumors (PanNETs) in a genetically engineered mouse model. Previously, a set of miRNAs was observed to be specifically up-regulated in a highly invasive and metastatic subtype of mouse and human PanNET. Using functional assays, we now implicate different miRNAs in distinct phenotypes: miR-137 stimulates tumor growth and local invasion, whereas the miR-23b cluster enables metastasis. An algorithm, Bio-miRTa, has been developed to facilitate the identification of biologically relevant miRNA target genes and applied to these miRNAs. We show that a top-ranked miR-137 candidate gene, Sorl1, has a tumor suppressor function in primary PanNETs. Among the top targets for the miR-23b cluster, Acvr1c/ALK7 has recently been described to be a metastasis suppressor, and we establish herein that it is down-regulated by the miR-23b cluster, which is crucial for its prometastatic activity. Two other miR-23b targets, Robo2 and P2ry1, also have demonstrable antimetastatic effects. Finally, we have used the Bio-miRTa algorithm in reverse to identify candidate miRNAs that might regulate activin B, the principal ligand for ALK7, identifying thereby a third family of miRNAs-miRNA-130/301-that is congruently up-regulated concomitant with down-regulation of activin B during tumorigenesis, suggestive of functional involvement in evasion of the proapoptotic barrier. Thus, dynamic up-regulation of miRNAs during multistep tumorigenesis and malignant progression serves to down-regulate distinctive suppressor mechanisms of tumor growth, invasion, and metastasis.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Activin Receptors, Type I/genetics , Activins/genetics , Algorithms , Animals , Cell Line, Tumor , Computational Biology/methods , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , LDL-Receptor Related Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Membrane Transport Proteins/genetics , Mice , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/mortality , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Prognosis , Receptors, LDL/genetics , Xenograft Model Antitumor Assays
10.
Genes Dev ; 25(9): 907-16, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21478273

ABSTRACT

The ubiquitous deregulation of Myc in human cancers makes it an intriguing therapeutic target, a notion supported by recent studies in Ras-driven lung tumors showing that inhibiting endogenous Myc triggers ubiquitous tumor regression. However, neither the therapeutic mechanism nor the applicability of Myc inhibition to other tumor types driven by other oncogenic mechanisms is established. Here, we show that inhibition of endogenous Myc also triggers ubiquitous regression of tumors in a simian virus 40 (SV40)-driven pancreatic islet tumor model. Such regression is presaged by collapse of the tumor microenvironment and involution of tumor vasculature. Hence, in addition to its diverse intracellular roles, endogenous Myc serves an essential and nonredundant role in coupling diverse intracellular oncogenic pathways to the tumor microenvironment, further bolstering its credentials as a pharmacological target.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Tumor Microenvironment/physiology , Adenoma, Islet Cell , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Doxycycline/pharmacology , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Mice , Neuroendocrine Tumors/blood supply , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/physiopathology , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/metabolism , Simian virus 40/physiology
11.
Genes Dev ; 23(1): 24-36, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19136624

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Survival , Cells, Cultured , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1
12.
Genes Dev ; 23(18): 2152-65, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19759263

ABSTRACT

While altered expression of microRNAs (miRs) in tumors has been well documented, it remains unclear how the miR transcriptome intersects neoplastic progression. By profiling the miR transcriptome we identified miR expression signatures associated with steps in tumorigenesis and the acquisition of hallmark capabilities in a prototypical mouse model of cancer. Metastases and a rare subset of primary tumors shared a distinct miR signature, implicating a discrete lineage for metastatic tumors. The miR-200 family is strongly down-regulated in metastases and met-like primary tumors, thereby relieving repression of the mesenchymal transcription factor Zeb1, which in turn suppresses E-cadherin. Treatment with a clinically approved angiogenesis inhibitor normalized angiogenic signature miRs in primary tumors, while altering expression of metastatic signature miRs similarly to liver metastases, suggesting their involvement in adaptive resistance to anti-angiogenic therapy via enhanced metastasis. Many of the miR changes associated with specific stages and hallmark capabilities in the mouse model are similarly altered in human tumors, including cognate pancreatic neuroendocrine tumors, implying a generality.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neuroendocrine Tumors/physiopathology , Pancreatic Neoplasms/physiopathology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neovascularization, Pathologic/physiopathology , Pyrroles/pharmacology , Sunitinib , Tumor Cells, Cultured
13.
Biol Chem ; 397(9): 871-81, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27149201

ABSTRACT

The cathepsin family of lysosomal proteases is increasingly being recognized for their altered expression in cancer and role in facilitating tumor progression. The aspartyl protease cathepsin E is overexpressed in several cancers and has been investigated as a biomarker for pancreatic ductal adenocarcinoma (PDAC). Here we show that cathepsin E expression in mouse PDAC tumors is increased by more than 400-fold when compared to healthy pancreatic tissue. Cathepsin E accumulates over the course of disease progression and accounts for more than 3% of the tumor protein in mice with end-stage disease. Through immunoblot analysis we determined that only procathepsin E exists in mouse PDAC tumors and cell lines derived from these tumors. By decreasing the pH, this procathepsion E is converted to the mature form, resulting in an increase in proteolytic activity. Although active site inhibitors can bind procathepsin E, treatment of PDAC mice with the aspartyl protease inhibitor ritonavir did not decrease tumor burden. Lastly, we used multiplex substrate profiling by mass spectrometry to identify two synthetic peptides that are hydrolyzed by procathepsin E near neutral pH. This work represents a comprehensive analysis of procathepsin E in PDAC and could facilitate the development of improved biomarkers for disease detection.


Subject(s)
Cathepsin E/metabolism , Enzyme Precursors/metabolism , Pancreatic Neoplasms/pathology , Amino Acid Sequence , Animals , Cathepsin E/antagonists & inhibitors , Cathepsin E/chemistry , Cell Line, Tumor , Disease Progression , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/chemistry , Gene Expression Regulation, Neoplastic , Hydrogen-Ion Concentration , Mice , Protease Inhibitors/pharmacology , Pancreatic Neoplasms
14.
Proc Natl Acad Sci U S A ; 110(48): 19507-12, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218578

ABSTRACT

Copper is an essential trace element, the imbalances of which are associated with various pathological conditions, including cancer, albeit via largely undefined molecular and cellular mechanisms. Here we provide evidence that levels of bioavailable copper modulate tumor growth. Chronic exposure to elevated levels of copper in drinking water, corresponding to the maximum allowed in public water supplies, stimulated proliferation of cancer cells and de novo pancreatic tumor growth in mice. Conversely, reducing systemic copper levels with a chelating drug, clinically used to treat copper disorders, impaired both. Under such copper limitation, tumors displayed decreased activity of the copper-binding mitochondrial enzyme cytochrome c oxidase and reduced ATP levels, despite enhanced glycolysis, which was not accompanied by increased invasiveness of tumors. The antiproliferative effect of copper chelation was enhanced when combined with inhibitors of glycolysis. Interestingly, larger tumors contained less copper than smaller tumors and exhibited comparatively lower activity of cytochrome c oxidase and increased glucose uptake. These results establish copper as a tumor promoter and reveal that varying levels of copper serves to regulate oxidative phosphorylation in rapidly proliferating cancer cells inside solid tumors. Thus, activation of glycolysis in tumors may in part reflect insufficient copper bioavailability in the tumor microenvironment.


Subject(s)
Copper/toxicity , Drinking Water/analysis , Neoplasms/physiopathology , Oxidative Phosphorylation/drug effects , Tumor Microenvironment/physiology , Water Pollutants, Chemical/toxicity , Adenosine Triphosphate/metabolism , Animals , Biological Availability , Blotting, Western , DNA Primers/genetics , Energy Metabolism/drug effects , Glycolysis/physiology , Membrane Potential, Mitochondrial/drug effects , Mice , Positron-Emission Tomography , Real-Time Polymerase Chain Reaction , Respiration
15.
Proc Natl Acad Sci U S A ; 110(16): E1480-9, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23542378

ABSTRACT

Based on clinical presentation, glioblastoma (GBM) is stratified into primary and secondary types. The protein 53 (p53) pathway is functionally incapacitated in most GBMs by distinctive type-specific mechanisms. To model human gliomagenesis, we used a GFAP-HRas(V12) mouse model crossed into the p53ER(TAM) background, such that either one or both copies of endogenous p53 is replaced by a conditional p53ER(TAM) allele. The p53ER(TAM) protein can be toggled reversibly in vivo between wild-type and inactive conformations by administration or withdrawal of 4-hydroxytamoxifen (4-OHT), respectively. Surprisingly, gliomas that develop in GFAP-HRas(V12);p53(+/KI) mice abrogate the p53 pathway by mutating p19(ARF)/MDM2 while retaining wild-type p53 allele. Consequently, such tumors are unaffected by restoration of their p53ER(TAM) allele. By contrast, gliomas arising in GFAP-HRas(V12);p53(KI/KI) mice develop in the absence of functional p53. Such tumors retain a functional p19(ARF)/MDM2-signaling pathway, and restoration of p53ER(TAM) allele triggers p53-tumor-suppressor activity. Congruently, growth inhibition upon normalization of mutant p53 by a small molecule, Prima-1, in human GBM cultures also requires p14(ARF)/MDM2 functionality. Notably, the antitumoral efficacy of p53 restoration in tumor-bearing GFAP-HRas(V12);p53(KI/KI) animals depends on the duration and frequency of p53 restoration. Thus, intermittent exposure to p53ER(TAM) activity mitigated the selective pressure to inactivate the p19(ARF)/MDM2/p53 pathway as a means of resistance, extending progression-free survival. Our results suggest that intermittent dosing regimes of drugs that restore wild-type tumor-suppressor function onto mutant, inactive p53 proteins will prove to be more efficacious than traditional chronic dosing by similarly reducing adaptive resistance.


Subject(s)
Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Signal Transduction/physiology , Tamoxifen/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Primers/genetics , Fluorescent Antibody Technique , Glioblastoma/metabolism , Histological Techniques , Humans , Immunoblotting , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
16.
Lancet ; 383(9916): 558-63, 2014 Feb 08.
Article in English | MEDLINE | ID: mdl-24351321

ABSTRACT

Some 40 years ago a metaphor was posed that cancer was such an insidious adversary that a declaration of war on the disease was justified. Although this statement was a useful inspiration for enlistment of resources, despite extraordinary progress in our understanding of disease pathogenesis, in most cases and for most forms of cancer this war has not been won. A second metaphor was about magic bullets--targeted therapies based on knowledge of mechanisms that were envisaged to strike with devastating consequences for the disease. The reality, however, is that targeted therapies are generally not curative or even enduringly effective, because of the adaptive and evasive resistance strategies developed by cancers under attack. In this Series paper, I suggest that, much like in modern warfare, the war on cancer needs to have a battlespace vision.


Subject(s)
Neoplasms/prevention & control , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Health Planning , Humans , Metaphor , Molecular Targeted Therapy
17.
Proc Natl Acad Sci U S A ; 108(49): E1275-84, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22084065

ABSTRACT

Preclinical trials in mice represent a critical step in the evaluation of experimental therapeutics. Genetically engineered mouse models (GEMMs) represent a promising platform for the evaluation of drugs, particularly those targeting the tumor microenvironment. We evaluated sunitinib, an angiogenesis inhibitor that targets VEGF and PDGF receptor signaling, in two GEMMs of pancreatic cancer. Sunitinib did not reduce tumor burden in pancreatic ductal adenocarcinoma (PDAC), whereas tumor burden was reduced in the pancreatic neuroendocrine tumor (PNET) model, the latter results confirming and extending previous studies. To explore the basis for the lack of pathologic response in PDAC, we used noninvasive microbubble contrast-enhanced ultrasound imaging, which revealed that sunitinib reduced blood flow both in PDAC and in PNET, concomitant with a reduction in vessel density; nevertheless, PDAC tumors continued to grow, whereas PNET were growth impaired. These results parallel the response in humans, where sunitinib recently garnered FDA and European approval in PNET, whereas two antiangiogenic drugs failed to demonstrate efficacy in PDAC clinical trials. The demonstration of on-target activity but with discordant benefit in the PDAC and PNET GEMMs illustrates the potential value of linked preclinical and clinical trials.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Indoles/therapeutic use , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrroles/therapeutic use , Animals , Antigens, CD34/metabolism , Blood Flow Velocity/drug effects , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/genetics , Clinical Trials as Topic , Contrast Media , Drug Evaluation, Preclinical , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microbubbles , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/genetics , Pancreas/blood supply , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prognosis , Receptors, Platelet-Derived Growth Factor/metabolism , Sunitinib , Treatment Outcome , Tumor Burden/drug effects , Ultrasonography
18.
Proc Natl Acad Sci U S A ; 108(24): 9939-44, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21622854

ABSTRACT

Whereas the roles of proangiogenic factors in carcinogenesis are well established, those of endogenous angiogenesis inhibitors (EAIs) remain to be fully elaborated. We investigated the roles of three EAIs during de novo tumorigenesis to further test the angiogenic balance hypothesis, which suggests that blood vessel development in the tumor microenvironment can be governed by a net loss of negative regulators of angiogenesis in addition to the well-established principle of up-regulated angiogenesis inducers. In a mouse model of pancreatic neuroendocrine cancer, administration of endostatin, thrombospondin-1, and tumstatin peptides, as well as deletion of their genes, reveal neoplastic stage-specific effects on angiogenesis, tumor progression, and survival, correlating with endothelial expression of their receptors. Deletion of tumstatin and thrombospondin-1 in mice lacking the p53 tumor suppressor gene leads to increased incidence and reduced latency of angiogenic lymphomas associated with diminished overall survival. The results demonstrate that EAIs are part of a balance mechanism regulating tumor angiogenesis, serving as intrinsic microenvironmental barriers to tumorigenesis.


Subject(s)
Autoantigens/metabolism , Collagen Type IV/metabolism , Endostatins/metabolism , Neoplasms/metabolism , Thrombospondin 1/metabolism , Amino Acid Sequence , Animals , Autoantigens/chemistry , Autoantigens/genetics , Cell Line , Collagen Type IV/chemistry , Collagen Type IV/genetics , Disease Progression , Endostatins/chemistry , Endostatins/genetics , Female , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Neoplasm Staging , Neoplasms/genetics , Neoplasms/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/prevention & control , Peptides/pharmacology , Propionates/pharmacology , Survival Analysis , Thrombospondin 1/chemistry , Thrombospondin 1/genetics , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
19.
Nat Cancer ; 5(6): 827-843, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38918437

ABSTRACT

Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Humans , Neovascularization, Pathologic/history , Neoplasms/blood supply , Neoplasms/history , Neoplasms/pathology , Angiogenesis Inhibitors/therapeutic use , Animals , History, 20th Century , Molecular Targeted Therapy/methods
20.
Cancer Discov ; 14(1): 36-48, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38047596

ABSTRACT

Cancer cells adapt and survive through the acquisition and selection of molecular modifications. This process defines cancer evolution. Building on a theoretical framework based on heritable genetic changes has provided insights into the mechanisms supporting cancer evolution. However, cancer hallmarks also emerge via heritable nongenetic mechanisms, including epigenetic and chromatin topological changes, and interactions between tumor cells and the tumor microenvironment. Recent findings on tumor evolutionary mechanisms draw a multifaceted picture where heterogeneous forces interact and influence each other while shaping tumor progression. A comprehensive characterization of the cancer evolutionary toolkit is required to improve personalized medicine and biomarker discovery. SIGNIFICANCE: Tumor evolution is fueled by multiple enabling mechanisms. Importantly, genetic instability, epigenetic reprogramming, and interactions with the tumor microenvironment are neither alternative nor independent evolutionary mechanisms. As demonstrated by findings highlighted in this perspective, experimental and theoretical approaches must account for multiple evolutionary mechanisms and their interactions to ultimately understand, predict, and steer tumor evolution.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/pathology , Epigenomics , Precision Medicine , Tumor Microenvironment/genetics
SELECTION OF CITATIONS
SEARCH DETAIL