Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(16): 3476-3498.e35, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541199

ABSTRACT

To improve the understanding of chemo-refractory high-grade serous ovarian cancers (HGSOCs), we characterized the proteogenomic landscape of 242 (refractory and sensitive) HGSOCs, representing one discovery and two validation cohorts across two biospecimen types (formalin-fixed paraffin-embedded and frozen). We identified a 64-protein signature that predicts with high specificity a subset of HGSOCs refractory to initial platinum-based therapy and is validated in two independent patient cohorts. We detected significant association between lack of Ch17 loss of heterozygosity (LOH) and chemo-refractoriness. Based on pathway protein expression, we identified 5 clusters of HGSOC, which validated across two independent patient cohorts and patient-derived xenograft (PDX) models. These clusters may represent different mechanisms of refractoriness and implicate putative therapeutic vulnerabilities.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Proteogenomics , Female , Humans , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
3.
Semin Cancer Biol ; 86(Pt 3): 857-867, 2022 11.
Article in English | MEDLINE | ID: mdl-35122974

ABSTRACT

Ionizing radiation is a pillar of cancer therapy that is deployed in more than half of all malignancies. The therapeutic effect of radiation is attributed to induction of DNA damage that kills cancers cells, but radiation also affects signaling that alters the composition of the tumor microenvironment by activating transforming growth factor ß (TGFß). TGFß is a ubiquitously expressed cytokine that acts as biological lynchpin to orchestrate phenotypes, the stroma, and immunity in normal tissue; these activities are subverted in cancer to promote malignancy, a permissive tumor microenvironment and immune evasion. The radiobiology of TGFß unites targets at the forefront of oncology-the DNA damage response and immunotherapy. The cancer cell intrinsic and extrinsic network of TGFß responses in the irradiated tumor form a barrier to both genotoxic treatments and immunotherapy response. Here, we focus on the mechanisms by which radiation induces TGFß activation, how TGFß regulates DNA repair, and the dynamic regulation of the tumor immune microenvironment that together oppose effective cancer therapy. Strategies to inhibit TGFß exploit fundamental radiobiology that may be the missing link to deploying TGFß inhibitors for optimal patient benefit from cancer treatment.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Radiobiology , DNA Damage , Signal Transduction , Neoplasms/radiotherapy , Tumor Microenvironment
5.
Hum Mutat ; 42(11): 1488-1502, 2021 11.
Article in English | MEDLINE | ID: mdl-34420246

ABSTRACT

Germline pathogenic variants in BRCA1 confer a high risk of developing breast and ovarian cancer. The BRCA1 exon 11 (formally exon 10) is one of the largest exons and codes for the nuclear localization signals of the corresponding gene product. This exon can be partially or entirely skipped during pre-mRNA splicing, leading to three major in-frame isoforms that are detectable in most cell types and tissue, and in normal and cancer settings. However, it is unclear whether the splicing imbalance of this exon is associated with cancer risk. Here we identify a common genetic variant in intron 10, rs5820483 (NC_000017.11:g.43095106_43095108dup), which is associated with exon 11 isoform expression and alternative splicing, and with the risk of breast cancer, but not ovarian cancer, in BRCA1 pathogenic variant carriers. The identification of this genetic effect was confirmed by analogous observations in mouse cells and tissue in which a loxP sequence was inserted in the syntenic intronic region. The prediction that the rs5820483 minor allele variant would create a binding site for the splicing silencer hnRNP A1 was confirmed by pull-down assays. Our data suggest that perturbation of BRCA1 exon 11 splicing modifies the breast cancer risk conferred by pathogenic variants of this gene.


Subject(s)
Breast Neoplasms/genetics , Exons , Genes, BRCA1 , Genetic Carrier Screening , Genetic Predisposition to Disease , RNA Splicing , Female , Humans , Introns
6.
Adv Exp Med Biol ; 890: 75-110, 2016.
Article in English | MEDLINE | ID: mdl-26703800

ABSTRACT

The tumor microenvironment (TME) represents a milieu that enables tumor cells to acquire the hallmarks of cancer. The TME is heterogeneous in composition and consists of cellular components, growth factors, proteases, and extracellular matrix. Concerted interactions between genetically altered tumor cells and genetically stable intratumoral stromal cells result in an "activated/reprogramed" stroma that promotes carcinogenesis by contributing to inflammation, immune suppression, therapeutic resistance, and generating premetastatic niches that support the initiation and establishment of distant metastasis. The lungs present a unique milieu in which tumors progress in collusion with the TME, as evidenced by regions of aberrant angiogenesis, acidosis and hypoxia. Inflammation plays an important role in the pathogenesis of lung cancer, and pulmonary disorders in lung cancer patients such as chronic obstructive pulmonary disease (COPD) and emphysema, constitute comorbid conditions and are independent risk factors for lung cancer. The TME also contributes to immune suppression, induces epithelial-to-mesenchymal transition (EMT) and diminishes efficacy of chemotherapies. Thus, the TME has begun to emerge as the "Achilles heel" of the disease, and constitutes an attractive target for anti-cancer therapy. Drugs targeting the components of the TME are making their way into clinical trials. Here, we will focus on recent advances and emerging concepts regarding the intriguing role of the TME in lung cancer progression, and discuss future directions in the context of novel diagnostic and therapeutic opportunities.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Communication/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Lung Diseases, Obstructive/complications , Lung Diseases, Obstructive/drug therapy , Lung Diseases, Obstructive/genetics , Lung Diseases, Obstructive/metabolism , Lung Neoplasms/complications , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Pulmonary Emphysema/complications , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Tumor Microenvironment/genetics
7.
J Mammary Gland Biol Neoplasia ; 25(3): 179-180, 2020 09.
Article in English | MEDLINE | ID: mdl-32997280
8.
Carcinogenesis ; 36 Suppl 1: S160-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106136

ABSTRACT

Potentially carcinogenic compounds may cause cancer through direct DNA damage or through indirect cellular or physiological effects. To study possible carcinogens, the fields of endocrinology, genetics, epigenetics, medicine, environmental health, toxicology, pharmacology and oncology must be considered. Disruptive chemicals may also contribute to multiple stages of tumor development through effects on the tumor microenvironment. In turn, the tumor microenvironment consists of a complex interaction among blood vessels that feed the tumor, the extracellular matrix that provides structural and biochemical support, signaling molecules that send messages and soluble factors such as cytokines. The tumor microenvironment also consists of many host cellular effectors including multipotent stromal cells/mesenchymal stem cells, fibroblasts, endothelial cell precursors, antigen-presenting cells, lymphocytes and innate immune cells. Carcinogens can influence the tumor microenvironment through effects on epithelial cells, the most common origin of cancer, as well as on stromal cells, extracellular matrix components and immune cells. Here, we review how environmental exposures can perturb the tumor microenvironment. We suggest a role for disrupting chemicals such as nickel chloride, Bisphenol A, butyltins, methylmercury and paraquat as well as more traditional carcinogens, such as radiation, and pharmaceuticals, such as diabetes medications, in the disruption of the tumor microenvironment. Further studies interrogating the role of chemicals and their mixtures in dose-dependent effects on the tumor microenvironment could have important general mechanistic implications for the etiology and prevention of tumorigenesis.


Subject(s)
Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Tumor Microenvironment/drug effects , Animals , Carcinogenesis/chemically induced , Humans , Neoplasms/chemically induced
9.
Growth Factors ; 33(2): 79-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25586866

ABSTRACT

Changes in activity or levels of transforming growth factor-ß (TGF-ß) are associated with a variety of diseases; however, measurement of TGF-ß in biological fluids is highly variable. TGF-ß is biologically inert when associated with its latency-associated peptide (LAP). Most available immunoassays require exogenous activation by acid/heat to release TGF-ß from the latent complex. We developed a novel electrochemiluminescence-based multiplexed assay on the MesoScale Discovery® platform that eliminates artificial activation, simultaneously measures both active TGF-ß1 and LAP1 and includes an internal control for platelet-derived TGF-ß contamination in blood specimens. We optimized this assay to evaluate plasma levels as a function of activation type and clinical specimen preparation. We determined that breast cancer patients' plasma have higher levels of circulating latent TGF-ß (LTGF-ß) as measured by LAP1 than healthy volunteers (p < 0.0001). This assay provides a robust tool for correlative studies of LTGF-ß levels with disease, treatment outcomes and toxicity with a broad clinical applicability.


Subject(s)
Breast Neoplasms/blood , Luminescent Measurements/methods , Transforming Growth Factor beta1/chemistry , Animals , Cell Line, Tumor , Culture Media, Conditioned , Electrochemistry/methods , Epithelial Cells/cytology , Female , HSC70 Heat-Shock Proteins/chemistry , Healthy Volunteers , Humans , Immunoassay/methods , Limit of Detection , Luminescence , Lung/cytology , Mink , Reproducibility of Results
10.
Stem Cells ; 32(3): 649-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24038768

ABSTRACT

Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor ß (TGFß) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFß. Radiation-induced Notch concomitant with TGFß was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.


Subject(s)
Aging/pathology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/radiation effects , Mammary Neoplasms, Animal/pathology , Radiation, Ionizing , Receptors, Estrogen/metabolism , Stem Cells/pathology , Animals , Biomarkers/metabolism , Cell Line , Cell Lineage , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Computer Simulation , Dose-Response Relationship, Radiation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Female , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Morphogenesis/drug effects , Morphogenesis/radiation effects , Receptors, Notch/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/radiation effects , Transforming Growth Factor beta/pharmacology
11.
Nat Rev Cancer ; 5(11): 867-75, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16327765

ABSTRACT

Radiation rapidly and persistently alters the soluble and insoluble components of the tissue microenvironment. This affects the cell phenotype, tissue composition and the physical interactions and signalling between cells. These alterations in the microenvironment can contribute to carcinogenesis and alter the tissue response to anticancer therapy. Examples of these responses and their implications are discussed with a view to therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Cells/radiation effects , Neoplasms, Radiation-Induced , Neoplasms/etiology , Neoplasms/radiotherapy , Radiation, Ionizing , Animals , Humans
12.
J Mammary Gland Biol Neoplasia ; 18(1): 3-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325014

ABSTRACT

Radiation exposure is a well-documented risk factor for breast cancer in women. Compelling epidemiological evidence in different exposed populations around the world demonstrate that excess breast cancer increases with radiation doses above 10 cGy. Both frequency and type of breast cancer are affected by prior radiation exposure. Many epidemiological studies suggest that radiation risk is inversely related to age at exposure; exposure during puberty poses the greatest risk while exposures past the menopause appear to carry very low risk. These observations are supported by experimental studies in mice and rats, which together provide the basis for the pubertal 'window of susceptibility' hypothesis for carcinogenic exposure. One line of experimental investigation suggests that the pubertal epithelium is more sensitive because DNA damage responses are less efficient, an other suggests that radiation affects stem cells self-renewal. A recent line of investigation suggests that the irradiated microenvironment mediates cancer risk. Studying the biological basis for radiation effects provides potential routes for protection in vulnerable populations, which include survivors of childhood cancers, as well as insights into the biology for certain types of sporadic cancer.


Subject(s)
Breast Neoplasms/etiology , Mammary Glands, Human/radiation effects , Women's Health , Age Factors , Animals , Breast Neoplasms/epidemiology , Breast Neoplasms/metabolism , Cellular Microenvironment/radiation effects , Female , Genomic Instability/radiation effects , Humans , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/radiation effects , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Radiation Tolerance , Risk , Stem Cells/radiation effects
13.
bioRxiv ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38496519

ABSTRACT

Background: Transforming growth factor beta (TGFß) is well-recognized as an immunosuppressive player in the tumor microenvironment but also has a significant impact on cancer cell phenotypes. Loss of TGFß signaling impairs DNA repair competency, which is described by a transcriptomic score, ßAlt. Cancers with high ßAlt have more genomic damage and are more responsive to genotoxic therapy. The growing appreciation that cancer DNA repair deficits are important determinants of immune response prompted us to investigate the association of ßAlt with response to immune checkpoint blockade (ICB). We predicted that high ßAlt tumors would be infiltrated with lymphocytes because of DNA damage burden and hence responsive to ICB. Methods: We analyzed public transcriptomic data from clinical trials and preclinical models using transcriptomic signatures of TGFß targets, DNA repair genes, tumor educated immune cells and interferon. A high ßAlt, immune poor mammary tumor derived transplant model resistant to programmed death ligand 1 (PD-L1) antibodies was studied using multispectral flow cytometry to interrogate the immune system. Results: Metastatic bladder patients in IMvigor 210 who responded to ICB had significantly increased ßAlt scores and experienced significantly longer overall survival compared to those with low ßAlt scores (hazard ratio 0.62, P=0.011) . Unexpectedly, 75% of high ßAlt cancers were immune poor as defined by low expression of tumor educated immune cell and interferon signatures. The association of high ßAlt with immune poor cancer was also evident in TCGA and preclinical cancer models. We used a high ßAlt, immune poor cancer to test therapeutic strategies to overcome its inherent anti-PD-L1 resistance. Combination treatment with radiation and TGFß inhibition were necessary for lymphocytic infiltration and activated NK cells were required for ICB response. Bioinformatic analysis identified high ßAlt, immune poor B16 and CT26 preclinical models and paired biopsies of cancer patients that also demonstrated NK cell activation upon response to ICB. Conclusions: Our studies support ßAlt as a biomarker that predicts response to ICB albeit in immune poor cancers, which has implications for the development of therapeutic strategies to increase the number of cancer patients who will benefit from immunotherapy.

14.
Sci Rep ; 14(1): 14175, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898077

ABSTRACT

Central nervous system tumors have resisted effective chemotherapy because most therapeutics do not penetrate the blood-tumor-brain-barrier. Nanomedicines between ~ 10 and 100 nm accumulate in many solid tumors by the enhanced permeability and retention effect, but it is controversial whether the effect can be exploited for treatment of brain tumors. PLX038A is a long-acting prodrug of the topoisomerase 1 inhibitor SN-38. It is composed of a 15 nm 4-arm 40 kDa PEG tethered to four SN-38 moieties by linkers that slowly cleave to release the SN-38. The prodrug was remarkably effective at suppressing growth of intracranial breast cancer and glioblastoma (GBM), significantly increasing the life span of mice harboring them. We addressed the important issue of whether the prodrug releases SN-38 systemically and then penetrates the brain to exert anti-tumor effects, or whether it directly penetrates the blood-tumor-brain-barrier and releases the SN-38 cargo within the tumor. We argue that the amount of SN-38 formed systemically is insufficient to inhibit the tumors, and show by PET imaging that a close surrogate of the 40 kDa PEG carrier in PLX038A accumulates and is retained in the GBM. We conclude that the prodrug penetrates the blood-tumor-brain-barrier, accumulates in the tumor microenvironment and releases its SN-38 cargo from within. Based on our results, we pose the provocative question as to whether the 40 kDa nanomolecule PEG carrier might serve as a "Trojan horse" to carry other drugs past the blood-tumor-brain-barrier and release them into brain tumors.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Irinotecan , Prodrugs , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Irinotecan/pharmacokinetics , Blood-Brain Barrier/metabolism , Mice , Prodrugs/pharmacokinetics , Prodrugs/chemistry , Prodrugs/metabolism , Humans , Cell Line, Tumor , Female , Xenograft Model Antitumor Assays , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use
15.
Clin Cancer Res ; 29(11): 2025-2033, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36598437

ABSTRACT

Even though the number of agents that inhibit TGFß being tested in patients with cancer has grown substantially, clinical benefit from TGFß inhibition has not yet been achieved. The myriad mechanisms in which TGFß is protumorigenic may be a key obstacle to its effective deployment; cancer cells frequently employ TGFß-regulated programs that engender plasticity, enable a permissive tumor microenvironment, and profoundly suppress immune recognition, which is the target of most current early-phase trials of TGFß inhibitors. Here we discuss the implications of a less well-recognized aspect of TGFß biology regulating DNA repair that mediates responses to radiation and chemotherapy. In cancers that are TGFß signaling competent, TGFß promotes effective DNA repair and suppresses error-prone repair, thus conferring resistance to genotoxic therapies and limiting tumor control. Cancers in which TGFß signaling is intrinsically compromised are more responsive to standard genotoxic therapy. Recognition that TGFß is a key moderator of both DNA repair and immunosuppression might be used to synergize combinations of genotoxic therapy and immunotherapy to benefit patients with cancer.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction , DNA Repair , DNA Damage , Tumor Microenvironment/genetics
16.
Methods Cell Biol ; 180: 49-67, 2023.
Article in English | MEDLINE | ID: mdl-37890932

ABSTRACT

Transforming growth factor ß (TGFß) is exquisitely regulated under physiological conditions but its activity is highly dysregulated in cancer. All cells make TGFß and have receptors for the ligand, which is sequestered in the extracellular matrix in a latent form. Ionizing radiation elicits rapid release of TGFß from these stores, so-called activation, over a wide range of doses and exposures, including low dose (<1Gy) whole-body irradiation, creating an extraordinarily potent signal in the irradiated tissue or tumor. Hence, accurate evaluation of TGFß activity is complicated because of its ubiquitous distribution as a latent complex. Here we describe conditions for assays that reveal TGFß activity in situ using either tissue preparations or functional imaging.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Humans , Signal Transduction , Neoplasms/radiotherapy , Extracellular Matrix
17.
Oncoimmunology ; 12(1): 2222560, 2023.
Article in English | MEDLINE | ID: mdl-37363104

ABSTRACT

Focal radiation therapy (RT) has attracted considerable attention as a combinatorial partner for immunotherapy (IT), largely reflecting a well-defined, predictable safety profile and at least some potential for immunostimulation. However, only a few RT-IT combinations have been tested successfully in patients with cancer, highlighting the urgent need for an improved understanding of the interaction between RT and IT in both preclinical and clinical scenarios. Every year since 2016, ImmunoRad gathers experts working at the interface between RT and IT to provide a forum for education and discussion, with the ultimate goal of fostering progress in the field at both preclinical and clinical levels. Here, we summarize the key concepts and findings presented at the Sixth Annual ImmunoRad conference.


Subject(s)
Neoplasms , Humans , Combined Modality Therapy , Neoplasms/radiotherapy , Neoplasms/drug therapy , Immunotherapy
18.
J Mammary Gland Biol Neoplasia ; 16(2): 147-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21590374

ABSTRACT

Transforming growth factor ß1 (TGFß) affects stroma and epithelial composition and interactions that mediate mammary development and determine the course of cancer. The reduction of TGFß in Tgfß1 heterozygote mice, which are healthy and long-lived, provides an important model to dissect the contribution of TGFß in mammary gland biology and cancer. We used both intact mice and mammary chimeras in conjunction with Tgfß1 genetic depletion and TGFß neutralizing antibodies to evaluate how stromal or epithelial TGFß depletion affect mammary development and response to physiological stimuli. Our studies of radiation carcinogenesis have revealed new aspects of TGFß biology and suggest that the paradoxical TGFß switch from tumor suppressor to tumor promoter can be resolved by assessing distinct stromal versus epithelial actions.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial Cells/pathology , Female , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Stromal Cells/pathology , Transforming Growth Factor beta1/genetics
19.
Cancer Res ; 82(3): 365-376, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34903599

ABSTRACT

In breast cancer, the type and distribution of infiltrating immune cells are associated with clinical outcome. Moreover, cancers with abundant tumor-infiltrating lymphocytes (TIL) are more likely to respond to immunotherapy, whereas those in which CD8+ T cells are completely absent (deserts) or excluded are less likely to respond. Detailed understanding of this biology is limited by a lack of preclinical breast cancer models that recapitulate TIL distributions and their associated biology. Here we established mammary tumor-derived transplants (mTDT) from 12 Trp53-null mammary tumors in syngeneic BALB/cJ mice and examined the stability of their growth rate, TIL distribution, and transcriptomic profiles. All mTDTs were estrogen receptor negative. Half of the parental tumors were classified as infiltrated, and the rest were divided between excluded and desert phenotypes. After two orthotopic passages, most (70%) mTDT from infiltrated parents recapitulated this pattern, whereas the desert or excluded parental patterns were maintained in about half of daughter mTDT. Approximately 30% of mTDT gave rise to lung or liver metastases, although metastasis was not associated with a TIL phenotype. Unsupervised transcriptomic analysis clustered mTDT according to their TIL spatial patterns. Infiltrated mTDT transplanted subcutaneously or orthotopically were resistant to anti-PD-L1. Profiling implicated prolonged antigen stimulation and loss of effector function of lymphocytes rather than T-cell exhaustion in the lack of response of infiltrated mTDT to checkpoint blockade. In summary, the molecular diversity and immune complexity of mTDT should facilitate the dissection of mechanisms of breast cancer response to immunotherapies. SIGNIFICANCE: A set of diverse preclinical models of breast cancer is characterized to enable mechanistic dissection of tumor-immune interactions and to improve the efficacy of immunotherapies.


Subject(s)
Breast Neoplasms/physiopathology , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Animal/physiopathology , Animals , Disease Models, Animal , Female , Mice , Mice, Nude , Tumor Microenvironment
20.
Mol Cancer Ther ; 21(1): 16-24, 2022 01.
Article in English | MEDLINE | ID: mdl-34670783

ABSTRACT

TGFß is a pleiotropic cytokine that plays critical roles to define cancer cell phenotypes, construct the tumor microenvironment, and suppress antitumor immune responses. As such, TGFß is a lynchpin for integrating cancer cell intrinsic pathways and communication among host cells in the tumor and beyond that together affect responses to genotoxic, targeted, and immune therapy. Despite decades of preclinical and clinical studies, evidence of clinical benefit from targeting TGFß in cancer remains elusive. Here, we review the mechanisms by which TGFß acts to oppose successful cancer therapy, the reported prognostic and predictive value of TGFß biomarkers, and the potential impact of inhibiting TGFß in precision oncology. Paradoxically, the diverse mechanisms by which TGFß impedes therapeutic response are a principal barrier to implementing TGFß inhibitors because it is unclear which TGFß mechanism is functional in which patient. Companion diagnostic tools and specific biomarkers of TGFß targeted biology will be the key to exploiting TGFß biology for patient benefit.


Subject(s)
Neoplasms/drug therapy , Transforming Growth Factor beta/therapeutic use , Humans , Prognosis , Signal Transduction , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL