Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Nat Commun ; 15(1): 5352, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914547

ABSTRACT

Immune checkpoint blockade (ICB) approaches have changed the therapeutic landscape for many tumor types. However, half of cutaneous squamous cell carcinoma (cSCC) patients remain unresponsive or develop resistance. Here, we show that, during cSCC progression in male mice, cancer cells acquire epithelial/mesenchymal plasticity and change their immune checkpoint (IC) ligand profile according to their features, dictating the IC pathways involved in immune evasion. Epithelial cancer cells, through the PD-1/PD-L1 pathway, and mesenchymal cancer cells, through the CTLA-4/CD80 and TIGIT/CD155 pathways, differentially block antitumor immune responses and determine the response to ICB therapies. Accordingly, the anti-PD-L1/TIGIT combination is the most effective strategy for blocking the growth of cSCCs that contain both epithelial and mesenchymal cancer cells. The expression of E-cadherin/Vimentin/CD80/CD155 proteins in cSCC, HNSCC and melanoma patient samples predicts response to anti-PD-1/PD-L1 therapy. Collectively, our findings indicate that the selection of ICB therapies should take into account the epithelial/mesenchymal features of cancer cells.


Subject(s)
B7-H1 Antigen , Carcinoma, Squamous Cell , Cell Plasticity , Epithelial-Mesenchymal Transition , Immune Checkpoint Inhibitors , Immunotherapy , Skin Neoplasms , Animals , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Mice , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Male , Immunotherapy/methods , Epithelial-Mesenchymal Transition/immunology , Cell Plasticity/drug effects , Cell Line, Tumor , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , CTLA-4 Antigen/immunology , Receptors, Virus/metabolism , Receptors, Virus/genetics , B7-1 Antigen/metabolism , Receptors, Immunologic/metabolism
2.
Br J Dermatol ; 191(2): 187-199, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38367194

ABSTRACT

BACKGROUND: Chronic spontaneous urticaria (CSU) is a relatively common skin disease associated with hives and angio-oedema. Eosinophils play a role in CSU pathogenesis. Benralizumab, an anti-interleukin-5 receptor-α monoclonal antibody, has been shown to induce nearly complete depletion of eosinophils. OBJECTIVES: To determine the clinical efficacy and safety of benralizumab in patients with CSU who were symptomatic despite H1 antihistamine treatment. METHODS: The 24-week, randomized, double-blind, placebo-controlled, phase IIb portion of the ARROYO trial enrolled adult patients with CSU who were currently on H1 antihistamine treatment. Patients were randomized to one of five treatment groups according to benralizumab dose and regimen for a 24-week treatment period. The primary endpoint was change from baseline in Itch Severity Score (ISS)7 at week 12. The key secondary endpoint was change from baseline in Urticaria Activity Score (UAS)7 at week 12. Additional secondary endpoints included other metrics to assess CSU at week 24, blood eosinophil levels, and pharmacokinetics and immunogenicity assessments. Exploratory subgroup analyses were conducted to explore responses according to demographics, clinical features and biomarkers. Safety was assessed in all treatment groups. RESULTS: Of 155 patients, 59 were randomized to benralizumab 30 mg, 56 to benralizumab 60 mg and 40 to placebo. Baseline and disease characteristics were consistent with what was expected for patients with CSU. There were no significant differences in change from baseline in ISS7 score at week 12 between benralizumab and placebo [benralizumab 30 mg vs. placebo, least-squares mean difference -1.01, 95% confidence interval (CI) -3.28 to 1.26; benralizumab 60 mg vs. placebo, least-squares mean difference -1.79, 95% CI -4.09 to 0.50] nor in change from baseline in UAS7 score at week 12 between benralizumab and placebo (benralizumab 30 mg vs. placebo, P = 0.407; benralizumab 60 mg vs. placebo, P = 0.082). Depletion of blood eosinophil levels was observed at week 24 in patients treated with benralizumab. All other secondary endpoints and exploratory/subgroup analyses indicated no significant differences between benralizumab and placebo. Safety results were consistent with the known profile of benralizumab. CONCLUSIONS: Although benralizumab resulted in near-complete depletion of blood eosinophils, there was no clinical benefit over placebo.


Chronic spontaneous urticaria (CSU) is a common disease characterized by hives, itching and inflammation (swelling) of the skin. CSU is mainly driven by what we call 'mast cells'. 'Eosinophils' are a type of white blood cell that protect the body from infections and allergens. These cells are abundant in skin biopsy samples of people with CSU, especially in the hives that contribute to swelling. Therefore, we thought that reducing eosinophils would be beneficial for treating CSU. Benralizumab is a drug that has been shown to reduce eosinophils in other diseases. This study, called 'ARROYO', was a 24-week clinical trial that compared benralizumab treatment with a placebo (inactive medicine) in adults with CSU who were taking antihistamines. We aimed to determine whether benralizumab would improve symptoms of CSU over time. Several assessments were used to measure changes in CSU symptoms, including hives, severity of itchiness, swelling of the skin, and other aspects related to overall psychological and physical wellbeing. The characteristics of the 155 people who took part in this study were consistent with what was expected for patients with CSU. We found that while benralizumab reduced eosinophil levels in people with CSU, there were no differences in symptoms in people receiving benralizumab compared with those receiving placebo. There were no new safety concerns related to benralizumab and no deaths. Overall, although benralizumab is effective at reducing the number of eosinophils, it is not effective at treating the symptoms of CSU. More studies are needed to uncover potential treatment targets in CSU.


Subject(s)
Antibodies, Monoclonal, Humanized , Chronic Urticaria , Humans , Double-Blind Method , Male , Female , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Chronic Urticaria/drug therapy , Middle Aged , Adult , Treatment Outcome , Eosinophils/immunology , Aged , Histamine H1 Antagonists/administration & dosage , Histamine H1 Antagonists/adverse effects , Young Adult
3.
Nat Commun ; 14(1): 6213, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37813842

ABSTRACT

Rank signaling pathway regulates mammary gland homeostasis and epithelial cell differentiation. Although Rank receptor is expressed by basal cells and luminal progenitors, its role in each individual cell lineage remains unclear. By combining temporal/lineage specific Rank genetic deletion with lineage tracing techniques, we found that loss of luminal Rank reduces the luminal progenitor pool and leads to aberrant alveolar-like differentiation with high protein translation capacity in virgin mammary glands. These Rank-deleted luminal cells are unable to expand during the first pregnancy, leading to lactation failure and impairment of protein synthesis potential in the parous stage. The unfit parous Rank-deleted luminal cells in the alveoli are progressively replaced by Rank-proficient cells early during the second pregnancy, thereby restoring lactation. Transcriptomic analysis and functional assays point to the awakening of basal bipotency after pregnancy by the induction of Rank/NF-κB signaling in basal parous cell to restore lactation and tissue homeostasis.


Subject(s)
Epithelial Cells , Stem Cells , Pregnancy , Female , Animals , Epithelial Cells/metabolism , Stem Cells/metabolism , Cell Differentiation , Cell Lineage , Signal Transduction , Mammary Glands, Animal/metabolism
4.
Cell Rep Med ; 4(8): 101120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37451269

ABSTRACT

The combination of endocrine therapy (ET) and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (CDK4/6i) was a hallmark in metastatic luminal breast cancer (BC). However, intrinsic and acquired resistance affects long-term efficacy. Here, we study the role of the receptor activator of nuclear factor-κB (RANK) pathway in CDK4/6i resistance. We find that RANK overexpression in luminal BC is associated with intrinsic resistance to CDK4/6i, both in vitro and in mouse xenografts, and decreased proliferation rate and chronic interferon (IFN) γ response are highlighted as resistance drivers. Gene expression data from the NeoPalAna CDK4/6i clinical trial, and studies with palbociclib-resistant cell lines, show that RANK is upregulated after treatment with CDK4/6i, supporting a role in acquired resistance. Our study shows that RANK ligand (RANKL) inhibitors can restore sensitivity to CDK4/6i and prevent acquired resistance. On the basis of these findings, we conclude that pharmacological inhibition of the RANK pathway through RANKL blocking could represent an add-on to ET + CDK4/6i, warranting further clinical studies.


Subject(s)
Breast Neoplasms , Signal Transduction , Humans , Protein Kinase Inhibitors/pharmacology , Animals , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Interferons/metabolism
5.
J Mammary Gland Biol Neoplasia ; 28(1): 17, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37450065

ABSTRACT

On 8 December 2022 the organizing committee of the European Network for Breast Development and Cancer labs (ENBDC) held its fifth annual Think Tank meeting in Amsterdam, the Netherlands. Here, we embraced the opportunity to look back to identify the most prominent breakthroughs of the past ten years and to reflect on the main challenges that lie ahead for our field in the years to come. The outcomes of these discussions are presented in this position paper, in the hope that it will serve as a summary of the current state of affairs in mammary gland biology and breast cancer research for early career researchers and other newcomers in the field, and as inspiration for scientists and clinicians to move the field forward.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , Humans , Female , Breast , Biology
6.
Clin Chim Acta ; 543: 117327, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-37015298

ABSTRACT

BACKGROUND: Therapeutic drug monitoring is a key tool for optimizing tacrolimus therapy in transplant recipients. The modified ACMIA assay from Siemens Healthcare Diagnostics is an immunoassay commonly used for tacrolimus monitoring, but it is not known whether this assay is resistant to interference from endogenous substances in real-world use. OBJECTIVE: To describe a case of unexpected interference in tacrolimus monitoring using the modified ACMIA method in a kidney transplant recipient, and to highlight the importance of careful interpretation of laboratory results and effective communication with clinicians in optimizing patient care. CASE DESCRIPTION: This case report describes a significant interference in the monitoring of tacrolimus in a kidney transplant recipient using the new ACMIA method. In this case, when aberrant results for tacrolimus were found using the new ACMIA method, they were re-analyzed using the CMIA method from Abbott. The presence of positive ANCA-MPO autoantibodies was found to be the most likely cause of the interference after an extensive workup. CONCLUSIONS: This is the first report of major interference with the modified ACMIA tacrolimus method and emphasizes the importance of proper interpretation of laboratory results and effective communication with clinicians in optimizing patient care.


Subject(s)
Kidney Transplantation , Tacrolimus , Humans , Tacrolimus/therapeutic use , Immunosuppressive Agents/therapeutic use , Immunoassay/methods , Drug Monitoring/methods
7.
EMBO Mol Med ; 15(4): e16715, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36880458

ABSTRACT

Despite strong preclinical data, the therapeutic benefit of the RANKL inhibitor, denosumab, in breast cancer patients, beyond the bone, is unclear. Aiming to select patients who may benefit from denosumab, we hereby analyzed RANK and RANKL protein expression in more than 2,000 breast tumors (777 estrogen receptor-negative, ER- ) from four independent cohorts. RANK protein expression was more frequent in ER- tumors, where it associated with poor outcome and poor response to chemotherapy. In ER- breast cancer patient-derived orthoxenografts (PDXs), RANKL inhibition reduced tumor cell proliferation and stemness, regulated tumor immunity and metabolism, and improved response to chemotherapy. Intriguingly, tumor RANK protein expression associated with poor prognosis in postmenopausal breast cancer patients, activation of NFKB signaling, and modulation of immune and metabolic pathways, suggesting that RANK signaling increases after menopause. Our results demonstrate that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and ER- breast cancer patients and support the therapeutic benefit of RANK pathway inhibitors, such as denosumab, in breast cancer patients with RANK+ ER- tumors after menopause.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/pathology , Denosumab/pharmacology , Denosumab/therapeutic use , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor Activator of Nuclear Factor-kappa B/therapeutic use , Postmenopause , RANK Ligand , Signal Transduction
9.
J Mammary Gland Biol Neoplasia ; 28(1): 2, 2023 02 18.
Article in English | MEDLINE | ID: mdl-36808257

ABSTRACT

Determination of the mammary epithelial cell that serves as the cell of origin for breast cancer is key to understand tumor heterogeneity and clinical management. In this study, we aimed to decipher whether Rank expression in the presence of PyMT and Neu oncogenes might affect the cell of origin of mammary gland tumors. We observed that Rank expression in PyMT+/- and Neu+/- mammary glands alters the basal and luminal mammary cell populations already in preneoplasic tissue, which may interfere with the tumor cell of origin restricting their tumorigenesis ability upon transplantation assays. In spite of this, Rank expression eventually promotes tumor aggressiveness once tumorigenesis is established.


Subject(s)
Ectopic Gene Expression , Mammary Neoplasms, Experimental , Animals , Humans , Mice , Mammary Neoplasms, Experimental/pathology , Carcinogenesis/pathology , Epithelial Cells/metabolism , Oncogenes , Mice, Transgenic
10.
J Equine Vet Sci ; 120: 104162, 2023 01.
Article in English | MEDLINE | ID: mdl-36343832

ABSTRACT

This study sought to compare the Schirmer Tear Test (STT)-1 results at 30 (STT30) versus 60 (STT60) seconds in healthy horses. This study included a total of 56 healthy horses. STT-1 was performed in both eyes, right eye first, and the wetting lengths were measured in STT30 and STT60. To evaluate the reduction of the initial reflex phase, the wetting length velocity was measured during the first 30 seconds. The effects of eye, age, weight, sex, and ambient temperature and humidity on STT values were evaluated. Mean (standard deviation) STT30 and STT60 were 19.06 (3.88) and 24.26 (4.50) mm. There was a linear correlation between the STT 30 and STT60, expressed according to the following equation: STT60 = 2.20 + 1.18 × STT30 (P = .001). STT30 or STT60 values did not vary between the sexes or correlate with age, weight, ambient temperature, or humidity. In conclusion, STT30 allows for an accurate, reliable, and applicable diagnosis of tear production compared with the standard STT60 value. The proposed method is shorter and may be a suitable alternative to the 1-min test.


Subject(s)
Diagnostic Techniques, Ophthalmological , Tears , Animals , Horses , Diagnostic Techniques, Ophthalmological/veterinary , Reference Values
11.
J Gastrointest Surg ; 26(12): 2579-2584, 2022 12.
Article in English | MEDLINE | ID: mdl-36224480

ABSTRACT

INTRODUCTION: Total neoadjuvant therapy (TNT) is a new therapeutic strategy in patients with rectal cancer. We examined the role of TNT, in addition to other pre-operative factors, as a predictor for pathologic complete response (pCR). METHODS: A retrospective analysis of all rectal cancer patients who underwent surgery between 2016 and 2021 was conducted. Patients were classified into two groups-pCR group and residual tumor group. Patient data were reviewed and entered into univariate and multivariate analyses to determine predictors of pCR. RESULTS: A total of 172 patients were treated with neoadjuvant therapy and underwent surgery during the study period. Sixty patients (34.9%) were treated with TNT while 112 (65.1%) were treated with traditional neoadjuvant chemoradiation. The overall pCR rate was 25.6% (44 patients), with 31.6% (19 patients) in patients who received TNT compared to 22.3% (25 patients) in patients who received neoadjuvant chemoradiation (NCRT). Univariate analysis of clinical and radiological factors correlated with pCR demonstrated no significant differences between the two groups in cT stage (p = 0.46), cN stage (p = 0.52), positive circumferential resection margin (CRM) (p = 0.72), tumor location (p = 0.35), symptomatic presentation (p = 0.09), and anal sphincter involvement (p = 0.68). Multivariate logistic analysis demonstrated that only pre-operative TNT (OR:2.35; 95% CI 1.06-5.25; p = 0.03) was predictive of pCR, while extramural vascular invasion (EMVI) was a predictor for lower rates of pCR (OR: 0.28; 95% CI 0.09-0.9; p = 0.03). CONCLUSION: Rectal cancer patients undergoing TNT prior to surgery have a higher chance of developing a complete pathologic response. Evaluation of this therapy should be continued and extended to larger numbers of patients to see if the differences we observed are real.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Humans , Retrospective Studies , Chemoradiotherapy , Neoplasm Staging , Treatment Outcome , Rectal Neoplasms/surgery , Rectal Neoplasms/pathology
12.
Sci Adv ; 8(8): eabi6110, 2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35050692

ABSTRACT

The spread of SARS-CoV-2 and ongoing COVID-19 pandemic underscores the need for new treatments. Here we report that cannabidiol (CBD) inhibits infection of SARS-CoV-2 in cells and mice. CBD and its metabolite 7-OH-CBD, but not THC or other congeneric cannabinoids tested, potently block SARS-CoV-2 replication in lung epithelial cells. CBD acts after viral entry, inhibiting viral gene expression and reversing many effects of SARS-CoV-2 on host gene transcription. CBD inhibits SARS-CoV-2 replication in part by up-regulating the host IRE1α RNase endoplasmic reticulum (ER) stress response and interferon signaling pathways. In matched groups of human patients from the National COVID Cohort Collaborative, CBD (100 mg/ml oral solution per medical records) had a significant negative association with positive SARS-CoV-2 tests. This study highlights CBD as a potential preventative agent for early-stage SARS-CoV-2 infection and merits future clinical trials. We caution against use of non-medical formulations including edibles, inhalants or topicals as a preventative or treatment therapy at the present time.


Subject(s)
Antiviral Agents/pharmacology , Cannabidiol/pharmacology , Host-Pathogen Interactions/drug effects , Immunity, Innate/drug effects , SARS-CoV-2/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , COVID-19/virology , Cannabidiol/chemistry , Cannabidiol/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Epithelial Cells/virology , Female , Gene Expression Regulation, Viral/drug effects , Host-Pathogen Interactions/physiology , Humans , Interferons/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , SARS-CoV-2/physiology , Vero Cells , Virus Internalization/drug effects , Virus Replication/drug effects , COVID-19 Drug Treatment
13.
Dev Cell ; 56(12): 1727-1741.e7, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34004159

ABSTRACT

Rank signaling enhances stemness in mouse and human mammary epithelial cells (MECs) and mediates mammary tumor initiation. Mammary tumors initiated by oncogenes or carcinogen exposure display high levels of Rank and Rank pathway inhibitors have emerged as a new strategy for breast cancer prevention and treatment. Here, we show that ectopic Rank expression in the mammary epithelia unexpectedly delays tumor onset and reduces tumor incidence in the oncogene-driven Neu and PyMT models. Mechanistically, we have found that ectopic expression of Rank or exposure to Rankl induces senescence, even in the absence of other oncogenic mutations. Rank leads to DNA damage and senescence through p16/p19. Moreover, RANK-induced senescence is essential for Rank-driven stemness, and although initially translates into delayed tumor growth, eventually promotes tumor progression and metastasis. We uncover a dual role for Rank in the mammary epithelia: Rank induces senescence and stemness, delaying tumor initiation but increasing tumor aggressiveness.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Mammary Neoplasms, Animal/genetics , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Aging/genetics , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , DNA Damage/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
14.
Breast Cancer Res ; 23(1): 42, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33785053

ABSTRACT

BACKGROUND: Around 15-20% of primary breast cancers are characterized by HER2 protein overexpression and/or HER2 gene amplification. Despite the successful development of anti-HER2 drugs, intrinsic and acquired resistance represents a major hurdle. This study was performed to analyze the RANK pathway contribution in HER2-positive breast cancer and anti-HER2 therapy resistance. METHODS: RANK and RANKL protein expression was assessed in samples from HER2-positive breast cancer patients resistant to anti-HER2 therapy and treatment-naive patients. RANK and RANKL gene expression was analyzed in paired samples from patients treated with neoadjuvant dual HER2-blockade (lapatinib and trastuzumab) from the SOLTI-1114 PAMELA trial. Additionally, HER2-positive breast cancer cell lines were used to modulate RANK expression and analyze in vitro the contribution of RANK signaling to anti-HER2 resistance and downstream signaling. RESULTS: RANK and RANKL proteins are more frequently detected in HER2-positive tumors that have acquired resistance to anti-HER2 therapies than in treatment-naive ones. RANK (but not RANKL) gene expression increased after dual anti-HER2 neoadjuvant therapy in the cohort from the SOLTI-1114 PAMELA trial. Results in HER2-positive breast cancer cell lines recapitulate the clinical observations, with increased RANK expression observed after short-term treatment with the HER2 inhibitor lapatinib or dual anti-HER2 therapy and in lapatinib-resistant cells. After RANKL stimulation, lapatinib-resistant cells show increased NF-κB activation compared to their sensitive counterparts, confirming the enhanced functionality of the RANK pathway in anti-HER2-resistant breast cancer. Overactivation of the RANK signaling pathway enhances ERK and NF-κB signaling and increases lapatinib resistance in different HER2-positive breast cancer cell lines, whereas RANK loss sensitizes lapatinib-resistant cells to the drug. Our results indicate that ErbB signaling is required for RANK/RANKL-driven activation of ERK in several HER2-positive cell lines. In contrast, lapatinib is not able to counteract the NF-κB activation elicited after RANKL treatment in RANK-overexpressing cells. Finally, we show that RANK binds to HER2 in breast cancer cells and that enhanced RANK pathway activation alters HER2 phosphorylation status. CONCLUSIONS: Our data support a physical and functional link between RANK and HER2 signaling in breast cancer and demonstrate that increased RANK signaling may contribute to the development of lapatinib resistance through NF-κB activation. Whether HER2-positive breast cancer patients with tumoral RANK expression might benefit from dual HER2 and RANK inhibition therapy remains to be elucidated.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor, ErbB-2/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lapatinib/therapeutic use , NF-kappa B/metabolism , Neoadjuvant Therapy , Protein Binding , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction , Trastuzumab/therapeutic use
15.
Clin Cancer Res ; 27(5): 1491-1504, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33262138

ABSTRACT

PURPOSE: Recurrent and/or metastatic unresectable cutaneous squamous cell carcinomas (cSCCs) are treated with chemotherapy or radiotherapy, but have poor clinical responses. A limited response (up to 45% of cases) to EGFR-targeted therapies was observed in clinical trials with patients with advanced and metastatic cSCC. Here, we analyze the molecular traits underlying the response to EGFR inhibitors, and the mechanisms responsible for cSCC resistance to EGFR-targeted therapy. EXPERIMENTAL DESIGN: We generated primary cell cultures and patient cSCC-derived xenografts (cSCC-PDXs) that recapitulate the histopathologic and molecular features of patient tumors. Response to gefitinib treatment was tested and gefitinib-resistant (GefR) cSCC-PDXs were developed. RNA sequence analysis was performed in matched untreated and GefR cSCC-PDXs to determine the mechanisms driving gefitinib resistance. RESULTS: cSCCs conserving epithelial traits exhibited strong activation of EGFR signaling, which promoted tumor cell proliferation, in contrast to mesenchymal-like cSCCs. Gefitinib treatment strongly blocked epithelial-like cSCC-PDX growth in the absence of EGFR and RAS mutations, whereas tumors carrying the E545K PIK3CA-activating mutation were resistant to treatment. A subset of initially responding tumors acquired resistance after long-term treatment, which was induced by the bypass from EGFR to FGFR signaling to allow tumor cell proliferation and survival upon gefitinib treatment. Pharmacologic inhibition of FGFR signaling overcame resistance to EGFR inhibitor, even in PIK3CA-mutated tumors. CONCLUSIONS: EGFR-targeted therapy may be appropriate for treating many epithelial-like cSCCs without PIK3CA-activating mutations. Combined EGFR- and FGFR-targeted therapy may be used to treat cSCCs that show intrinsic or acquired resistance to EGFR inhibitors.


Subject(s)
Drug Resistance, Neoplasm , Gefitinib/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms, Glandular and Epithelial/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Nat Commun ; 11(1): 6335, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303745

ABSTRACT

Most breast cancers exhibit low immune infiltration and are unresponsive to immunotherapy. We hypothesized that inhibition of the receptor activator of nuclear factor-κB (RANK) signaling pathway may enhance immune activation. Here we report that loss of RANK signaling in mouse tumor cells increases leukocytes, lymphocytes, and CD8+ T cells, and reduces macrophage and neutrophil infiltration. CD8+ T cells mediate the attenuated tumor phenotype observed upon RANK loss, whereas neutrophils, supported by RANK-expressing tumor cells, induce immunosuppression. RANKL inhibition increases the anti-tumor effect of immunotherapies in breast cancer through a tumor cell mediated effect. Comparably, pre-operative single-agent denosumab in premenopausal early-stage breast cancer patients from the Phase-II D-BEYOND clinical trial (NCT01864798) is well tolerated, inhibits RANK pathway and increases tumor infiltrating lymphocytes and CD8+ T cells. Higher RANK signaling activation in tumors and serum RANKL levels at baseline predict these immune-modulatory effects. No changes in tumor cell proliferation (primary endpoint) or other secondary endpoints are observed. Overall, our preclinical and clinical findings reveal that tumor cells exploit RANK pathway as a mechanism to evade immune surveillance and support the use of RANK pathway inhibitors to prime luminal breast cancer for immunotherapy.


Subject(s)
Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , Adult , Animals , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Chemokines/metabolism , Denosumab/pharmacology , Denosumab/therapeutic use , Female , Humans , Immunosuppression Therapy , Immunotherapy , Inflammation Mediators/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Middle Aged , Models, Biological , Myeloid Cells/immunology , Neoplasm Staging , Neutrophils/immunology , RANK Ligand/blood , RANK Ligand/metabolism
17.
Oncogene ; 38(45): 7106-7112, 2019 11.
Article in English | MEDLINE | ID: mdl-31409895

ABSTRACT

Human tumors show altered patterns of protein isoforms that can be related to the dysregulation of messenger RNA alternative splicing also observed in transformed cells. Although somatic mutations in core spliceosome components and their associated factors have been described in some cases, almost nothing is known about the contribution of distorted epigenetic patterns to aberrant splicing. Herein, we show that the splicing RNA-binding protein CELF2 is targeted by promoter hypermethylation-associated transcriptional silencing in human cancer. Focusing on the context of breast cancer, we also demonstrate that CELF2 restoration has growth-inhibitory effects and that its epigenetic loss induces an aberrant downstream pattern of alternative splicing, affecting key genes in breast cancer biology such as the autophagy factor ULK1 and the apoptotic protein CARD10. Furthermore, the presence of CELF2 hypermethylation in the clinical setting is associated with shorter overall survival of the breast cancer patients carrying this epigenetic lesion.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , CELF Proteins/genetics , DNA Methylation , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , RNA Splicing , Female , Gene Expression Regulation, Neoplastic , Humans , Spliceosomes/genetics , Tumor Cells, Cultured
18.
Mol Cancer Res ; 17(10): 2063-2076, 2019 10.
Article in English | MEDLINE | ID: mdl-31320385

ABSTRACT

Taxanes are standard therapy in clinical practice for metastatic breast cancer; however, primary or acquired chemoresistance are a common cause of mortality. Breast cancer patient-derived xenografts (PDX) are powerful tools for the study of cancer biology and drug treatment response. Specific DNA methylation patterns have been associated to different breast cancer subtypes but its association with chemoresistance remains unstudied. Aiming to elucidate docetaxel resistance mechanisms, we performed genome-wide DNA methylation in breast cancer PDX models, including luminal and triple-negative breast cancer (TNBC) models sensitive to docetaxel, their matched models after emergence of chemoresistance and residual disease after short-term docetaxel treatment. We found that DNA methylation profiles from breast cancer PDX models maintain the subtype-specific methylation patterns of clinical samples. Two main DNA methylation clusters were found in TNBC PDX and remain stable during the emergence of docetaxel resistance; however, some genes/pathways were differentially methylated according to docetaxel response. A DNA methylation signature of resistance able to segregate TNBC based on chemotherapy response was identified. Transcriptomic profiling of selected sensitive/resistant pairs and integrative analysis with methylation data demonstrated correlation between some differentially methylated and expressed genes in docetaxel-resistant TNBC PDX models. Multiple gene expression changes were found after the emergence of docetaxel resistance in TNBC. DNA methylation and transcriptional changes identified between docetaxel-sensitive and -resistant TNBC PDX models or residual disease may have predictive value for chemotherapy response in TNBC. IMPLICATIONS: Subtype-specific DNA methylation patterns are maintained in breast cancer PDX models. While no global methylation changes were found, we uncovered differentially DNA methylated and expressed genes/pathways associated with the emergence of docetaxel resistance in TNBC.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA Methylation/genetics , Docetaxel/therapeutic use , Transcriptome/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Docetaxel/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Mice , Xenograft Model Antitumor Assays
19.
Cancer Res ; 79(16): 4258-4270, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31213465

ABSTRACT

Taxanes are the mainstay of treatment in triple-negative breast cancer (TNBC), with de novo and acquired resistance limiting patient's survival. To investigate the genetic basis of docetaxel resistance in TNBC, exome sequencing was performed on matched TNBC patient-derived xenografts (PDX) sensitive to docetaxel and their counterparts that developed resistance in vivo upon continuous drug exposure. Most mutations, small insertions/deletions, and copy number alterations detected in the initial TNBC human metastatic samples were maintained after serial passages in mice and emergence of resistance. We identified a chromosomal amplification of chr12p in a human BRCA1-mutated metastatic sample and the derived chemoresistant PDX, but not in the matched docetaxel-sensitive PDX tumor. Chr12p amplification was validated in a second pair of docetaxel-sensitive/resistant BRCA1-mutated PDXs and after short-term docetaxel treatment in several TNBC/BRCA1-mutated PDXs and cell lines, as well as during metastatic recurrence in a patient with BRCA1-mutated breast cancer who had progressed on docetaxel treatment. Analysis of clinical data indicates an association between chr12p amplification and patients with TNBC/basal-like breast cancer, a BRCA1 mutational signature, and poor survival after chemotherapy. Detection of chr12p amplification in a cohort of TNBC PDX models was associated with an improved response to carboplatin. Our findings reveal tumor clonal dynamics during chemotherapy treatments and suggest that a preexisting population harboring chr12p amplification is associated with the emergence of docetaxel resistance and carboplatin responsiveness in TNBC/BRCA1-mutated tumors. SIGNIFICANCE: Chr12p copy number gains indicate rapid emergence of resistance to docetaxel and increased sensitivity to carboplatin, therefore sequential docetaxel/carboplatin treatment could improve survival in TNBC/BRCA1 patients. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/16/4258/F1.large.jpg.


Subject(s)
Carboplatin/pharmacology , Chromosomes, Human, Pair 12 , Docetaxel/pharmacology , Drug Resistance, Neoplasm/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Animals , BRCA1 Protein/genetics , Cell Line, Tumor , Exome , Female , Humans , Mice , Mutation , Treatment Outcome , Triple Negative Breast Neoplasms/mortality , Xenograft Model Antitumor Assays
20.
Cancer Treat Rev ; 76: 57-67, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31136850

ABSTRACT

Patients with solid tumours are at risk of impaired bone health from metastases and cancer therapy-induced bone loss (CTIBL). We review medical management of bone health in patients with solid tumours over the past 30 years, from first-generation bisphosphonates to the receptor activator of nuclear factor κB ligand (RANKL)-targeted monoclonal antibody, denosumab. In the 1980s, first-generation bisphosphonates were shown to reduce the incidence of skeletal-related events (SREs) in patients with breast cancer. Subsequently, more potent second- and third-generation bisphosphonates were developed, particularly zoledronic acid (ZA). Head-to-head studies showed that ZA was significantly more effective than pamidronate for reducing SREs in patients with breast and castrate-resistant prostate cancer (CRPC), becoming the standard of care for more than a decade. The RANKL inhibitor denosumab was licensed in 2010, and head-to-head studies and integrated analyses confirmed its superiority to ZA for preventing SREs, particularly in breast cancer and CRPC. Bisphosphonates and denosumab have also been investigated for prevention of CTIBL in patients receiving hormonal therapy for breast and prostate cancer, and denosumab is licensed in this indication. Despite advances in management of bone health, several issues remain, notably the optimal time to initiate therapy, duration of therapy, and dosing frequency, and how to avoid toxicity, particularly with long-term treatment. In summary, introduction of ZA and denosumab has protected patients with bone metastasis from serious bone complications and improved their quality of life. Ongoing research will hopefully guide the optimal use of these agents to help maintain bone health in patients with solid tumours.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/therapeutic use , Animals , Bone Resorption/drug therapy , Bone Resorption/etiology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Denosumab/therapeutic use , Female , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Zoledronic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...