1
|
Iwase T, Wang X, Thi Hanh Phi L, Sridhar N, Ueno NT, Lee J. Advances in targets in inflammatory breast cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 384:125-152. [PMID: 38637096 DOI: 10.1016/bs.ircmb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States.
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lan Thi Hanh Phi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nithya Sridhar
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
2
|
Najm M, Cornet M, Albergante L, Zinovyev A, Sermet-Gaudelus I, Stoven V, Calzone L, Martignetti L. Representation and quantification of module activity from omics data with rROMA. NPJ Syst Biol Appl 2024; 10:8. [PMID: 38242871 PMCID: PMC10799004 DOI: 10.1038/s41540-024-00331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
The efficiency of analyzing high-throughput data in systems biology has been demonstrated in numerous studies, where molecular data, such as transcriptomics and proteomics, offers great opportunities for understanding the complexity of biological processes. One important aspect of data analysis in systems biology is the shift from a reductionist approach that focuses on individual components to a more integrative perspective that considers the system as a whole, where the emphasis shifted from differential expression of individual genes to determining the activity of gene sets. Here, we present the rROMA software package for fast and accurate computation of the activity of gene sets with coordinated expression. The rROMA package incorporates significant improvements in the calculation algorithm, along with the implementation of several functions for statistical analysis and visualizing results. These additions greatly expand the package's capabilities and offer valuable tools for data analysis and interpretation. It is an open-source package available on github at: www.github.com/sysbio-curie/rROMA . Based on publicly available transcriptomic datasets, we applied rROMA to cystic fibrosis, highlighting biological mechanisms potentially involved in the establishment and progression of the disease and the associated genes. Results indicate that rROMA can detect disease-related active signaling pathways using transcriptomic and proteomic data. The results notably identified a significant mechanism relevant to cystic fibrosis, raised awareness of a possible bias related to cell culture, and uncovered an intriguing gene that warrants further investigation.
Collapse
Affiliation(s)
- Matthieu Najm
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Matthieu Cornet
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Luca Albergante
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Andrei Zinovyev
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Isabelle Sermet-Gaudelus
- Faculté de Médecine, Université de Paris, Paris, France
- Institut Necker Enfants Malades, INSERM U1151, Paris, France
- AP-HP. Centre - Université Paris Cité; Hôpital Necker Enfants Malades, Centre de Référence Maladie Rare - Mucoviscidose, Paris, France
| | - Véronique Stoven
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Laurence Calzone
- INSERM U900, 75428, Paris, France
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France
- Institut Curie, PSL Research University, 75248, Paris, France
| | - Loredana Martignetti
- INSERM U900, 75428, Paris, France.
- Center for Computational Biology, Mines ParisTech, PSL Research University, 75006, Paris, France.
- Institut Curie, PSL Research University, 75248, Paris, France.
| |
Collapse
|
3
|
Cereser B, Yiu A, Tabassum N, Del Bel Belluz L, Zagorac S, Ancheta KRZ, Zhong R, Miere C, Jeffries-Jones AR, Moderau N, Werner B, Stebbing J. The mutational landscape of the adult healthy parous and nulliparous human breast. Nat Commun 2023; 14:5136. [PMID: 37673861 PMCID: PMC10482899 DOI: 10.1038/s41467-023-40608-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/03/2023] [Indexed: 09/08/2023] Open
Abstract
The accumulation of somatic mutations in healthy human tissues has been extensively characterized, but the mutational landscape of the healthy breast is still poorly understood. Our analysis of whole-genome sequencing shows that in line with other healthy organs, the healthy breast during the reproduction years accumulates mutations with age, with the rate of accumulation in the epithelium of 15.24 ± 5 mutations/year. Both epithelial and stromal compartments contain mutations in breast-specific driver genes, indicative of subsequent positive selection. Parity- and age-associated differences are evident in the mammary epithelium, partly explaining the observed difference in breast cancer risk amongst women of different childbearing age. Parity is associated with an age-dependent increase in the clone size of mutated epithelial cells, suggesting that older first-time mothers have a higher probability of accumulating oncogenic events in the epithelium compared to younger mothers or nulliparous women. In conclusion, we describe the reference genome of the healthy female human breast during reproductive years and provide evidence of how parity affects the genomic landscape of the mammary gland.
Collapse
Grants
- British Heart Foundation
- British Heart Foundation (BHF)
- The work is funded by Action Against Cancer (grants P62625, BC; P66683, NT; P66814, LDDB; P63015, SZ; P71728, NM), UKRI-IBIN (grant P82771, NM), UKRI-OOACTN (grant P91025, NM), British Heart Foundation (grant F36083, AY), Barts Charity Lectureship (grant MGU045, BW).
Collapse
Affiliation(s)
- Biancastella Cereser
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK.
| | - Angela Yiu
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Neha Tabassum
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Lisa Del Bel Belluz
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Sladjana Zagorac
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Kenneth Russell Zapanta Ancheta
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
- Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, UK
| | - Rongrong Zhong
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Cristian Miere
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Nina Moderau
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Benjamin Werner
- Evolutionary Dynamics Group, Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Justin Stebbing
- Cancer Genetics Group, Department of Surgery and Cancer, Imperial College London, London, UK.
- Department of Life Sciences, Anglia Ruskin University (ARU), Cambridge, UK.
| |
Collapse
|
4
|
Balamurugan K, Poria DK, Sehareen SW, Krishnamurthy S, Tang W, McKennett L, Padmanaban V, Czarra K, Ewald AJ, Ueno NT, Ambs S, Sharan S, Sterneck E. Stabilization of E-cadherin adhesions by COX-2/GSK3β signaling is a targetable pathway in metastatic breast cancer. JCI Insight 2023; 8:156057. [PMID: 36757813 PMCID: PMC10070121 DOI: 10.1172/jci.insight.156057] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Metastatic progression of epithelial cancers can be associated with epithelial-mesenchymal transition (EMT) including transcriptional inhibition of E-cadherin (CDH1) expression. Recently, EM plasticity (EMP) and E-cadherin-mediated, cluster-based metastasis and treatment resistance have become more appreciated. However, the mechanisms that maintain E-cadherin expression in this context are less understood. Through studies of inflammatory breast cancer (IBC) and a 3D tumor cell "emboli" culture paradigm, we discovered that cyclooxygenase 2 (COX-2; PTGS2), a target gene of C/EBPδ (CEBPD), or its metabolite prostaglandin E2 (PGE2) promotes protein stability of E-cadherin, β-catenin, and p120 catenin through inhibition of GSK3β. The COX-2 inhibitor celecoxib downregulated E-cadherin complex proteins and caused cell death. Coexpression of E-cadherin and COX-2 was seen in breast cancer tissues from patients with poor outcome and, along with inhibitory GSK3β phosphorylation, in patient-derived xenografts (PDX) including triple negative breast cancer (TNBC).Celecoxib alone decreased E-cadherin protein expression within xenograft tumors, though CDH1 mRNA levels increased, and reduced circulating tumor cell (CTC) clusters. In combination with paclitaxel, celecoxib attenuated or regressed lung metastases. This study has uncovered a mechanism by which metastatic breast cancer cells can maintain E-cadherin-mediated cell-to-cell adhesions and cell survival, suggesting that some patients with COX-2+/E-cadherin+ breast cancer may benefit from targeting of the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Kuppusamy Balamurugan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Dipak K Poria
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Saadiya W Sehareen
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Lois McKennett
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Veena Padmanaban
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelli Czarra
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Shikha Sharan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Esta Sterneck
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| |
Collapse
|
5
|
Ruiz TFR, Colleta SJ, Dos Santos DD, Castro NFC, Cabral ÁS, Calmon MF, Rahal P, Gil CD, Girol AP, Vilamaior PSL, Leonel ECR, Taboga SR. Bisphenol A disruption promotes mammary tumor microenvironment via phenotypic cell polarization and inflammatory response. Cell Biol Int 2023; 47:1136-1146. [PMID: 36906806 DOI: 10.1002/cbin.12007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/04/2023] [Accepted: 02/20/2023] [Indexed: 03/13/2023]
Abstract
Inflammation in the established tumor microenvironment (TME) is often associated with a poor prognosis of breast cancer. Bisphenol A (BPA) is an endocrine-disrupting chemical that acts as inflammatory promoter and tumoral facilitator in mammary tissue. Previous studies demonstrated the onset of mammary carcinogenesis at aging when BPA exposure occurred in windows of development/susceptibility. We aim to investigate the inflammatory repercussions of BPA in TME in mammary gland (MG) during neoplastic development in aging. Female Mongolian gerbils were exposed to low (50 µg/kg) or high BPA (5000 µg/kg) doses during pregnancy and lactation. They were euthanized at 18 months of age (aging) and the MG were collected for inflammatory markers and histopathological analysis. Contrarily to control MG, BPA induced carcinogenic development mediated by COX-2 and p-STAT3 expression. BPA was also able to promote macrophage and mast cell (MC) polarization in tumoral phenotype, evidenced by pathways for recruitment and activation of these inflammatory cells and tissue invasiveness triggered by tumor necrosis factor-alpha and transforming growth factor-beta 1 (TGF-β1). Increase of tumor-associated macrophages, M1 (CD68 + iNOS+) and M2 (CD163+) expressing pro-tumoral mediators and metalloproteases was observed; this aspect greatly contributed to stromal remodeling and invasion of neoplastic cells. In addition, the MC population drastically increased in BPA-exposed MG. Tryptase-positive MCs increased in disrupted MG and expressed TGF-β1, contributing to EMT process during carcinogenesis mediated by BPA. BPA exposure interfered in inflammatory response by releasing and enhancing the expression of mediators that contribute to tumor growth and recruitment of inflammatory cells that promote a malignant profile.
Collapse
Affiliation(s)
- Thalles F R Ruiz
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Simone J Colleta
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Diego D Dos Santos
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Morphology and Genetics, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Nayara F C Castro
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Ágata S Cabral
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Marilia F Calmon
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Paula Rahal
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Cristiane D Gil
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Morphology and Genetics, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Ana Paula Girol
- Department of Basics Sciences, University Center Padre Albino (UNIFIPA), Catanduva, São Paulo, Brazil
| | - Patricia S L Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Ellen C R Leonel
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences (ICB III), Federal University of Goiás (UFG), Goiânia, Goiás, Brazil
| | - Sebastião R Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
6
|
Belpaire M, Taminiau A, Geerts D, Rezsohazy R. HOXA1, a breast cancer oncogene. Biochim Biophys Acta Rev Cancer 2022; 1877:188747. [PMID: 35675857 DOI: 10.1016/j.bbcan.2022.188747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
More than 25 years ago, the first literature records mentioned HOXA1 expression in human breast cancer. A few years later, HOXA1 was confirmed as a proper oncogene in mammary tissue. In the following two decades, molecular data about the mode of action of the HOXA1 protein, the factors contributing to activate and maintain HOXA1 gene expression and the identity of its target genes have accumulated and provide a wider view on the association of this transcription factor to breast oncogenesis. Large-scale transcriptomic data gathered from wide cohorts of patients further allowed refining the relationship between breast cancer type and HOXA1 expression. Several recent reports have reviewed the connection between cancer hallmarks and the biology of HOX genes in general. Here we take HOXA1 as a paradigm and propose an extensive overview of the molecular data centered on this oncoprotein, from what its expression modulators, to the interactors contributing to its oncogenic activities, and to the pathways and genes it controls. The data converge to an intricate picture that answers questions on the multi-modality of its oncogene activities, point towards better understanding of breast cancer aetiology and thereby provides an appraisal for treatment opportunities.
Collapse
Affiliation(s)
- Magali Belpaire
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Dirk Geerts
- Heart Failure Research Center, Amsterdam University Medical Center (AMC), Universiteit van Amsterdam, Amsterdam, the Netherlands.
| | - René Rezsohazy
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium.
| |
Collapse
|
7
|
Management of Locally Advanced Breast Cancer. Breast Cancer 2022. [DOI: 10.1007/978-981-16-4546-4_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
8
|
Rogic A, Pant I, Grumolato L, Fernandez-Rodriguez R, Edwards A, Das S, Sun A, Yao S, Qiao R, Jaffer S, Sachidanandam R, Akturk G, Karlic R, Skobe M, Aaronson SA. High endogenous CCL2 expression promotes the aggressive phenotype of human inflammatory breast cancer. Nat Commun 2021; 12:6889. [PMID: 34824220 PMCID: PMC8617270 DOI: 10.1038/s41467-021-27108-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory Breast Cancer (IBC) is a highly aggressive malignancy with distinct clinical and histopathological features whose molecular basis is unresolved. Here we describe a human IBC cell line, A3250, that recapitulates key IBC features in a mouse xenograft model, including skin erythema, diffuse tumor growth, dermal lymphatic invasion, and extensive metastases. A3250 cells express very high levels of the CCL2 chemokine and induce tumors enriched in macrophages. CCL2 knockdown leads to a striking reduction in macrophage densities, tumor proliferation, skin erythema, and metastasis. These results establish IBC-derived CCL2 as a key factor driving macrophage expansion, and indirectly tumor growth, with transcriptomic analysis demonstrating the activation of multiple inflammatory pathways. Finally, primary human IBCs exhibit macrophage infiltration and an enriched macrophage RNA signature. Thus, this human IBC model provides insight into the distinctive biology of IBC, and highlights potential therapeutic approaches to this deadly disease. Inflammatory breast cancer (IBC) is an aggressive form of breast cancer with a poor prognosis. Here the authors report the characterization of a human IBC cell line recapitulating the clinical and histopathological features of the human disease, and implicating its high level of CCL2 in macrophage infiltration and tumor progression.
Collapse
Affiliation(s)
- Anita Rogic
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ila Pant
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Luca Grumolato
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
| | - Ruben Fernandez-Rodriguez
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew Edwards
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Suvendu Das
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Institute of Advanced Research, Department of Biological Sciences and Biotechnology, Koba Institutional, Area, Gandhinagar 382 426, Gujarat, India
| | - Aaron Sun
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shen Yao
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rui Qiao
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shabnam Jaffer
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ravi Sachidanandam
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Guray Akturk
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rosa Karlic
- Bioinformatics group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Mihaela Skobe
- Laboratory of Cancer Lymphangiogenesis, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA. .,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA. .,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
9
|
Bertucci F, Boudin L, Finetti P, Van Berckelaer C, Van Dam P, Dirix L, Viens P, Gonçalves A, Ueno NT, Van Laere S, Birnbaum D, Mamessier E. Immune landscape of inflammatory breast cancer suggests vulnerability to immune checkpoint inhibitors. Oncoimmunology 2021; 10:1929724. [PMID: 34104544 PMCID: PMC8158040 DOI: 10.1080/2162402x.2021.1929724] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background. Anti-PD1/PDL1 immune checkpoint inhibitors (ICIs) showed promising results in breast cancer, and exploration of additional actionable immune checkpoints is ongoing. Inflammatory breast cancer (IBC) is an aggressive form of disease, the immune tumor microenvironment (TME) of which is poorly known. We aimed at providing the first comprehensive immune portrait of IBCs. Methods. From the gene expression profiles of 137 IBC and 252 non-IBC clinical samples, we measured the fractions of 22 immune cell types, expression of signatures associated with tertiary lymphoid structures (TLS) and with the response to ICIs (T cell-inflamed signature: TIS) and of 18 genes coding for major actionable immune checkpoints. The IBC/non-IBC comparison was adjusted upon the clinicopathological variables. Results. The immune profiles of IBCs were heterogeneous. CIBERSORT analysis showed profiles rich in macrophages, CD8+ and CD4 + T-cells, with remarkable similarity with melanoma TME. The comparison with non-IBCs showed significant enrichment in M1 macrophages, γδ T-cells, and memory B-cells. IBCs showed higher expression of TLS and TIS signatures. The TIS signature displayed values in IBCs close to those observed in other cancers sensitive to ICIs. Two-thirds of actionable immune genes (HAVCR2/TIM3, CD27, CD70, CTLA4, ICOS, IDO1, LAG3, PDCD1, TNFRSF9, PVRIG, CD274/PDL1, and TIGIT) were overexpressed in IBCs as compared to normal breast and two-thirds were overexpressed in IBCs versus non-IBCs, with very frequent co-overexpression. For most of them, the overexpression was associated with better pathological response to chemotherapy. Conclusion. Our results suggest the potential higher vulnerability of IBC to ICIs. Clinical trials.
Collapse
Affiliation(s)
- François Bertucci
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Laurys Boudin
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Christophe Van Berckelaer
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Peter Van Dam
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium.,Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Patrice Viens
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Anthony Gonçalves
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Naoto T Ueno
- Breast Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium.,Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Emilie Mamessier
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| |
Collapse
|
10
|
Gadde M, Phillips C, Ghousifam N, Sorace AG, Wong E, Krishnamurthy S, Syed A, Rahal O, Yankeelov TE, Woodward WA, Rylander MN. In vitro vascularized tumor platform for modeling tumor-vasculature interactions of inflammatory breast cancer. Biotechnol Bioeng 2020; 117:3572-3590. [PMID: 32648934 DOI: 10.1002/bit.27487] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/24/2020] [Accepted: 07/08/2020] [Indexed: 12/26/2022]
Abstract
Inflammatory breast cancer (IBC), a rare form of breast cancer associated with increased angiogenesis and metastasis, is largely driven by tumor-stromal interactions with the vasculature and the extracellular matrix (ECM). However, there is currently a lack of understanding of the role these interactions play in initiation and progression of the disease. In this study, we developed the first three-dimensional, in vitro, vascularized, microfluidic IBC platform to quantify the spatial and temporal dynamics of tumor-vasculature and tumor-ECM interactions specific to IBC. Platforms consisting of collagen type 1 ECM with an endothelialized blood vessel were cultured with IBC cells, MDA-IBC3 (HER2+) or SUM149 (triple negative), and for comparison to non-IBC cells, MDA-MB-231 (triple negative). Acellular collagen platforms with endothelialized blood vessels served as controls. SUM149 and MDA-MB-231 platforms exhibited a significantly (p < .05) higher vessel permeability and decreased endothelial coverage of the vessel lumen compared to the control. Both IBC platforms, MDA-IBC3 and SUM149, expressed higher levels of vascular endothelial growth factor (p < .05) and increased collagen ECM porosity compared to non-IBCMDA-MB-231 (p < .05) and control (p < .01) platforms. Additionally, unique to the MDA-IBC3 platform, we observed progressive sprouting of the endothelium over time resulting in viable vessels with lumen. The newly sprouted vessels encircled clusters of MDA-IBC3 cells replicating a key feature of in vivo IBC. The IBC in vitro vascularized platforms introduced in this study model well-described in vivo and clinical IBC phenotypes and provide an adaptable, high throughput tool for systematically and quantitatively investigating tumor-stromal mechanisms and dynamics of tumor progression.
Collapse
Affiliation(s)
- Manasa Gadde
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Caleb Phillips
- Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Enoch Wong
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Anum Syed
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Omar Rahal
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas.,Department of Oncology, The University of Texas at Austin, Austin, Texas.,Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Wendy A Woodward
- M.D. Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Marissa N Rylander
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, Texas.,Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
11
|
Guo Y, Chen B, Pei X, Zhang D. Radix Stephaniae Tetrandrine: An Emerging Role for Management of Breast Cancer. Curr Pharm Des 2020; 26:25-36. [DOI: 10.2174/1381612826666200110143706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/02/2019] [Indexed: 11/22/2022]
Abstract
Background:
Radix Stephaniae Tetrandrine (RST), known as FangJi (Pinyin name) in Chinese, is the
dried root of Stephania tetrandra S.Moore, and has been prescribed in combination with other herbs to treat cardiovascular
diseases and breast cancer in traditional Chinese medicine (TCM) clinical trials.
Objective:
The aim of the review is to provide a comprehensive evaluation about the application of RST in breast
cancer management in TCM clinical trials, its ingredients, and its action on preventing the development of breast
cancer in vitro and in vivo studies.
Methods:
Literature sources used were Pubmed, CNKI.net, Cqvip.com, and the Web of Science. For the inquiry,
keywords such as Fangji, breast cancer, clinical trials, Radix Stephaniae Tetrandrine, tetrandrine, and fangchinoline
were used in various combinations. About 150 research papers and reviews were consulted.
Results:
In TCM, RST exhibited the anti-tumor ability through its action on the bladder and lungs through dispersing
phlegm and blood stasis. 10 clinical trials were identified which used RST in combination with other
herbs to treat breast cancer. On average, the trials were characterized by high efficacy (>85%) and low toxicity.
However, most of the clinical trials are characterized as small patient samples, poor design, and different combinations
of herbs in prescriptions. To date, more and more compounds have been isolated from this plant. RST
exhibited anti-tumor activities by targeting reversing multidrug resistance, inhibiting cell proliferation, inducing
apoptosis, preventing tumor angiogenesis, anti-oxidation, anti-inflammation, and enhancing the sensitization and
attenuating the toxicity of radiotherapy.
Conclusion:
The successful applications of RST in TCM clinical trials and preclinical experiments to beating
breast cancer will provide potent lead compounds in the identification of novel anti-cancer drugs, which further
contributes to the scientific exploration of functions of RST in TCM.
Collapse
Affiliation(s)
- Yubo Guo
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese medicine, Beijing 100029, China
| | - Beibei Chen
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese medicine, Beijing 100029, China
| | - Xiaohua Pei
- The Fangshan Hospital of Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese medicine, Beijing 100029, China
| |
Collapse
|
12
|
Linhares S, Alrammah T, Alghamdi HA, Möller MG. Inflammatory Breast Cancer in Pregnancy and Lactation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1252:143-151. [PMID: 32816275 DOI: 10.1007/978-3-030-41596-9_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammatory breast cancer (IBC) represents only 1% to 5% of all breast malignancies and is an extremely aggressive subtype. At time of diagnosis, up to 85% of patients will present with regional nodal metastases and up to 30 % will have metastasis to distant organs. There is limited medical literature describing treatment guidelines for IBC during gestation. The best diagnostic tools are core needle and full-thickness skin punch biopsies to assess presence of dermal lymphatic invasion. Breast Ultrasound is preferred to mammogram, but mammography could still be done with proper fetal shielding. Ultrasound and Magnetic resonance imaging are used for staging. Pregnant patients should be managed with special attention to the health of the fetus by a multidisciplinary team. Treatment based on current guidelines consist of a sequence of systemic chemotherapy followed by mastectomy with axillary dissection (modified radical mastectomy), and even if good clinical nodal response to neoadjuvant therapy is obtained, sentinel node biopsy is not recommended. Radiation therapy is to be given once the baby has been delivered. Chemotherapy is not recommended in the first trimester, and anti-estrogen hormonal therapy, as well as targeted Her2-neu therapies are contraindicated during the length of the pregnancy. There is no evidence that early termination improves the outcome. However, given the poor prognosis of IBC, patients should be fully counseled on the risks and benefits of continuing or terminating an early pregnancy.
Collapse
Affiliation(s)
| | - Tamrah Alrammah
- Department of Surgery, Division of Surgical Oncology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Hattan A Alghamdi
- Department of Surgery, Division of Surgical Oncology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Mecker G Möller
- Department of Surgery, Division of Surgical Oncology, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
13
|
Liu X, Yang B, Li Y, Yang Z, Xue L, Zhang M, Chen G, Chinnathambi A, Alahmadi T. Anti-inflammatory and anti-cell proliferative effects of dieckol in the prevention and treatment of colon cancer induced by 1,2-dimethyl hydrazine in experimental animals. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_165_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
14
|
Malik DES, David RM, Gooderham NJ. Interleukin-6 selectively induces drug metabolism to potentiate the genotoxicity of dietary carcinogens in mammary cells. Arch Toxicol 2019; 93:3005-3020. [PMID: 31515600 DOI: 10.1007/s00204-019-02558-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022]
Abstract
Breast cancer is the most commonly diagnosed malignancy in females, the etiology being multifactorial and includes the role of lifestyle exposure to DNA-damaging chemicals such as dietary carcinogens benzo (a) pyrene (BaP) and 2-amino-1-methyl-6-phenylimidazo [4, 5-b] pyridine (PhIP). Both compounds require cytochrome P450 (CYP)-mediated metabolic activation to DNA-damaging species, and both induce transcriptional responses through the nuclear receptors Aryl hydrocarbon receptor (AhR) and estrogen receptor α (ERα). BaP and PhIP are mammary carcinogens in rodents. Clinically, circulating IL-6 expression is linked with poor prognosis of cancer and 35% of the deaths in breast cancer are linked with inflammation. The objective of this work was to investigate the molecular toxicology and local activation of BaP and PhIP in the presence of IL-6. Our laboratory has previously reported that miR27b can regulate CYP1B1 expression in colorectal cells, here we have investigated if this mechanism is working in mammary cell models, MCF-7 and MDA-MB-231 cells. Treatment (24 h) of cells with BaP (10 nM-10 µM) and PhIP (100 nM-100 µM) significantly induced genetic damage (micronuclei formation) in a dose-dependent manner in both cell lines. This effect was potentiated in the presence of human IL-6 at concentrations reported to be expressed in clinical breast cancer. On its own, IL-6 treatment failed to induce micronuclei frequency above the control levels in these cells. Compared to BaP or PhIP treatment alone, IL-6 plus BaP or PhIP selectively induced CYP1B1 significantly in both cell lines. Additionally, miR27b expression was downregulated by IL-6 treatments and transfection with miR27b inhibitor confirmed that miR27b is a regulator of CYP1B1 in both cell lines. These data show that BaP- and PhIP-induced DNA damage in mammary cells is potentiated by the inflammatory cytokine IL-6 and that inflammation-induced CYP expression, specifically CYP1B1 via miR27b, is responsible for this effect.
Collapse
Affiliation(s)
- Durr-E-Shahwar Malik
- Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Rhiannon M David
- Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
- Genetic Toxicology, Discovery Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Nigel J Gooderham
- Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
15
|
Reddy SM, Reuben A, Barua S, Jiang H, Zhang S, Wang L, Gopalakrishnan V, Hudgens CW, Tetzlaff MT, Reuben JM, Tsujikawa T, Coussens LM, Wani K, He Y, Villareal L, Wood A, Rao A, Woodward WA, Ueno NT, Krishnamurthy S, Wargo JA, Mittendorf EA. Poor Response to Neoadjuvant Chemotherapy Correlates with Mast Cell Infiltration in Inflammatory Breast Cancer. Cancer Immunol Res 2019; 7:1025-1035. [PMID: 31043414 DOI: 10.1158/2326-6066.cir-18-0619] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/18/2018] [Accepted: 04/22/2019] [Indexed: 12/28/2022]
Abstract
Our understanding is limited concerning the tumor immune microenvironment of inflammatory breast cancer (IBC), an aggressive form of primary cancer with low rates of pathologic complete response to current neoadjuvant chemotherapy (NAC) regimens. We retrospectively identified pretreatment (N = 86) and matched posttreatment tissue (N = 27) from patients with stage III or de novo stage IV IBC who received NAC followed by a mastectomy. Immune profiling was performed including quantification of lymphoid and myeloid infiltrates by IHC and T-cell repertoire analysis. Thirty-four of 86 cases in this cohort (39.5%) achieved a pathologic complete response. Characterization of the tumor microenvironment revealed that having a lower pretreatment mast cell density was significantly associated with achieving a pathologic complete response to NAC (P = 0.004), with responders also having more stromal tumor-infiltrating lymphocytes (P = 0.035), CD8+ T cells (P = 0.047), and CD20+ B cells (P = 0.054). Spatial analysis showed close proximity of mast cells to CD8+ T cells, CD163+ monocytes/macrophages, and tumor cells when pathologic complete response was not achieved. PD-L1 positivity on tumor cells was found in fewer than 2% of cases and on immune cells in 27% of cases, but with no correlation to response. Our results highlight the strong association of mast cell infiltration with poor response to NAC, suggesting a mechanism of treatment resistance and a potential therapeutic target in IBC. Proximity of mast cells to immune and tumor cells may suggest immunosuppressive or tumor-promoting interactions of these mast cells.
Collapse
Affiliation(s)
- Sangeetha M Reddy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Souptik Barua
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Hong Jiang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shaojun Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Courtney W Hudgens
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael T Tetzlaff
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas
| | - Takahiro Tsujikawa
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, Oregon.,Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Lisa M Coussens
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yan He
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lily Villareal
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas
| | - Anita Wood
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.,Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, Texas. .,Department of Breast Surgical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| |
Collapse
|
16
|
Brzezinska M, Dixon JM. Inflammatory breast cancer: no longer an absolute contraindication for breast conservation surgery following good response to neoadjuvant therapy. Gland Surg 2018; 7:520-524. [PMID: 30687625 DOI: 10.21037/gs.2018.08.04] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammatory breast cancer (IBC) is a rare form of breast cancer characterised by an erythematous swollen breast with extensive oedema and has in the past been associated with a very poor prognosis. After diagnosis by core biopsy of the cancer and any involved nodes patients in the Edinburgh Breast Unit (EBU) are primarily managed with neoadjuvant systemic therapy-chemotherapy or endocrine therapy. If the cancer is localised to one or a few well defined lesions then each of these lesions together with the lowest involved node are clipped. Response during neoadjuvant treatment is monitored clinically and by ultrasound plus mammography +/- magnetic resonance imaging (MRI). Following completion of neoadjuvant therapy, imaging is reviewed at a multidisciplinary meeting and patients with a localised single or multiple areas of cancer where all signs of erythema and oedema have settled are considered as to their suitability for breast conserving surgery and whole breast radiotherapy [breast conserving treatment (BCT)]. Here we discuss the results and outcomes of a selected group of patients with IBC who after obtaining a very good response to neoadjuvant chemotherapy or endocrine therapy were treated by BCT and we compare these with other recent publications on this topic. Our data show that patients treated by BCT did not have worse outcomes than patients treated with mastectomy. Importantly other series published recently support our conclusions. Another important observation is that response in estrogen receptor (ER) rich IBC is seen with neoadjuvant endocrine treatment and so not everyone with IBC needs to have neoadjuvant chemotherapy. In conclusion, patients with one or more well defined and localised breast masses and IBC may be suitable for BCT after a major response to neoadjuvant therapy and for these patients BCT should now be considered a viable option.
Collapse
Affiliation(s)
- Monika Brzezinska
- Edinburgh Breast Unit, NHS Lothian, Western General Hospital, Edinburgh, UK
| | - J Michael Dixon
- Edinburgh Breast Unit, NHS Lothian, Western General Hospital, Edinburgh, UK
| |
Collapse
|
17
|
Harano K, Wang Y, Lim B, Seitz RS, Morris SW, Bailey DB, Hout DR, Skelton RL, Ring BZ, Masuda H, Rao AUK, Laere SV, Bertucci F, Woodward WA, Reuben JM, Krishnamurthy S, Ueno NT. Rates of immune cell infiltration in patients with triple-negative breast cancer by molecular subtype. PLoS One 2018; 13:e0204513. [PMID: 30312311 PMCID: PMC6193579 DOI: 10.1371/journal.pone.0204513] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
In patients with triple-negative breast cancer (TNBC), tumor-infiltrating lymphocytes (TILs) are associated with improved survival. Lehmann et al. identified 4 molecular subtypes of TNBC [basal-like (BL) 1, BL2, mesenchymal (M), and luminal androgen receptor (LAR)], and an immunomodulatory (IM) gene expression signature indicates the presence of TILs and modifies these subtypes. The association between TNBC subtype and TILs is not known. Also, the association between inflammatory breast cancer (IBC) and the presence of TILs is not known. Therefore, we studied the IM subtype distribution among different TNBC subtypes. We retrospectively analyzed patients with TNBC from the World IBC Consortium dataset. The molecular subtype and the IM signature [positive (IM+) or negative (IM-)] were analyzed. Fisher’s exact test was used to analyze the distribution of positivity for the IM signature according to the TNBC molecular subtype and IBC status. There were 88 patients with TNBC in the dataset, and among them 39 patients (44%) had IBC and 49 (56%) had non-IBC. The frequency of IM+ cases differed by TNBC subtype (p = 0.001). The frequency of IM+ cases by subtype was as follows: BL1, 48% (14/29); BL2, 30% (3/10); LAR, 18% (3/17); and M, 0% (0/21) (in 11 patients, the subtype could not be determined). The frequency of IM+ cases did not differ between patients with IBC and non-IBC (23% and 33%, respectively; p = 0.35). In conclusion, the IM signature representing the underlying molecular correlate of TILs in the tumor may differ by TNBC subtype but not by IBC status.
Collapse
Affiliation(s)
- Kenichi Harano
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Pulmonology Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Ying Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bora Lim
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert S. Seitz
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
| | - Stephan W. Morris
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
| | - Daniel B. Bailey
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
| | - David R. Hout
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
| | - Rachel L. Skelton
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
| | - Brian Z. Ring
- Insight Genetics, Inc., Nashville, Tennessee, United States of America
- College of Life Science, Huazhong University of Science and Technology, Wuhan, China
| | - Hiroko Masuda
- Department of Breast Surgical Oncology, Showa University, Shinagawa-ku, Tokyo, Japan
| | - Arvind U. K. Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Steven Van Laere
- Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Francois Bertucci
- Predictive Oncology team, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Wendy A. Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - James M. Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (SK); (NTU)
| | - Naoto T. Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (SK); (NTU)
| |
Collapse
|
18
|
Mohammadi A, Sharifi A, Pourpaknia R, Mohammadian S, Sahebkar A. Manipulating macrophage polarization and function using classical HDAC inhibitors: Implications for autoimmunity and inflammation. Crit Rev Oncol Hematol 2018; 128:1-18. [DOI: 10.1016/j.critrevonc.2018.05.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/18/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
|
19
|
Andrieu GP, Shafran JS, Deeney JT, Bharadwaj KR, Rangarajan A, Denis GV. BET proteins in abnormal metabolism, inflammation, and the breast cancer microenvironment. J Leukoc Biol 2018; 104:265-274. [PMID: 29493812 PMCID: PMC6134394 DOI: 10.1002/jlb.5ri0917-380rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 02/10/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022] Open
Abstract
Obesity and its associated pathology Type 2 diabetes are two chronic metabolic and inflammatory diseases that promote breast cancer progression, metastasis, and poor outcomes. Emerging critical opinion considers unresolved inflammation and abnormal metabolism separately from obesity; settings where they do not co-occur can inform disease mechanism. In breast cancer, the tumor microenvironment is often infiltrated with T effector and T regulatory cells programmed by metabolic signaling. The pathways by which tumor cells evade immune surveillance, immune therapies, and take advantage of antitumor immunity are poorly understood, but likely depend on metabolic inflammation in the microenvironment. Immune functions are abnormal in metabolic disease, and lessons learned from preclinical studies in lean and metabolically normal environments may not translate to patients with obesity and metabolic disease. This problem is made more urgent by the rising incidence of breast cancer among women who are not obese but who have metabolic disease and associated inflammation, a phenotype common in Asia. The somatic BET proteins, comprising BRD2, BRD3, and BRD4, are new critical regulators of metabolism, coactivate transcription of genes that encode proinflammatory cytokines in immune cell subsets infiltrating the microenvironment, and could be important targets in breast cancer immunotherapy. These transcriptional coregulators are well known to regulate tumor cell progression, but only recently identified as critical for metabolism, metastasis, and expression of immune checkpoint molecules. We consider interrelationships among metabolism, inflammation, and breast cancer aggressiveness relevant to the emerging threat of breast cancer among women with metabolic disease, but without obesity.
Collapse
Affiliation(s)
| | - Jordan S. Shafran
- Cancer Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jude T. Deeney
- Department of Medicine, Section of Endocrinology, Obesity Research Center, Evans Biomedical Research Center; Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kishan R. Bharadwaj
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Gerald V. Denis
- Cancer Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Zare A, Petrova A, Agoumi M, Amstrong H, Bigras G, Tonkin K, Wine E, Baksh S. RIPK2: New Elements in Modulating Inflammatory Breast Cancer Pathogenesis. Cancers (Basel) 2018; 10:cancers10060184. [PMID: 29874851 PMCID: PMC6025367 DOI: 10.3390/cancers10060184] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/29/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer that is associated with significantly high mortality. In spite of advances in IBC diagnoses, the prognosis is still poor compared to non-IBC. Due to the aggressive nature of the disease, we hypothesize that elevated levels of inflammatory mediators may drive tumorigenesis and metastasis in IBC patients. Utilizing IBC cell models and patient tumor samples, we can detect elevated NF-κB activity and hyperactivation of non-canonical drivers of NF-κB (nuclear factor kappaB)-directed inflammation such as tyrosine phosphorylated receptor-interacting protein kinase 2 (pY RIPK2), when compared to non-IBC cells or patients. Interestingly, elevated RIPK2 activity levels were present in a majority of pre-chemotherapy samples from IBC patients at the time of diagnosis to suggest that patients at diagnosis had molecular activation of NF-κB via RIPK2, a phenomenon we define as “molecular inflammation”. Surprisingly, chemotherapy did cause a significant increase in RIPK2 activity and thus molecular inflammation suggesting that chemotherapy does not resolve the molecular activation of NF-κB via RIPK2. This would impact on the metastatic potential of IBC cells. Indeed, we can demonstrate that RIPK2 activity correlated with advanced tumor, metastasis, and group stage as well as body mass index (BMI) to indicate that RIPK2 might be a useful prognostic marker for IBC and advanced stage breast cancer.
Collapse
Affiliation(s)
- Alaa Zare
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Alexandra Petrova
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Mehdi Agoumi
- Anatomic Pathologist at DynalifeDx, Diagnostic Laboratory Services; Department of Laboratory Medicine and Pathology, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2R3, Canada.
| | - Heather Amstrong
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Gilbert Bigras
- Cross Cancer Institute Department of Laboratory Medicine and Pathology, University of Alberta, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada.
| | - Katia Tonkin
- Division of Medical Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada.
| | - Eytan Wine
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Shairaz Baksh
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Division of Medical Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada.
- Division of Experimental Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2R7, Canada.
- Women and Children's Health Research Institute, Edmonton Clinic Health Academy (ECHA), University of Alberta, 4-081 11405 87 Avenue NW Edmonton, AB T6G 1C9, Canada.
| |
Collapse
|
21
|
Eckhardt BL, Gagliardi M, Iles L, Evans K, Ivan C, Liu X, Liu CG, Souza G, Rao A, Meric-Bernstam F, Ueno NT, Bartholomeusz GA. Clinically relevant inflammatory breast cancer patient-derived xenograft-derived ex vivo model for evaluation of tumor-specific therapies. PLoS One 2018; 13:e0195932. [PMID: 29768500 PMCID: PMC5955489 DOI: 10.1371/journal.pone.0195932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/03/2018] [Indexed: 02/05/2023] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive presentation of invasive breast cancer with a 62% to 68% 5-year survival rate. It is the most lethal form of breast cancer, and early recognition and treatment is important for patient survival. Like non-inflammatory breast cancer, IBC comprises multiple subtypes, with the triple-negative subtype being overrepresented. Although the current multimodality treatment regime of anthracycline- and taxane-based neoadjuvant therapy, surgery, and radiotherapy has improved the outcome of patients with triple-negative IBC, overall survival continues to be worse than in patients with non-inflammatory locally advanced breast cancer. Translation of new therapies into the clinics to successfully treat IBC has been poor, in part because of the lack of in vitro preclinical models that can accurately predict the response of the original tumor to therapy. We report the generation of a preclinical IBC patient-derived xenograft (PDX)-derived ex vivo (PDXEx) model and show that it closely replicates the tissue architecture of the original PDX tumor harvested from mice. The gene expression profile of our IBC PDXEx model had a high degree of correlation to that of the original tumor. This suggests that the process of generating the PDXEx model did not significantly alter the molecular signature of the original tumor. We demonstrate a high degree of similarity in drug response profile between a PDX mouse model and our PDXEx model generated from the same original PDX tumor tissue and treated with the same panel of drugs, indicating that our PDXEx model had high predictive value in identifying effective tumor-specific therapies. Finally, we used our PDXEx model as a platform for a robotic-based high-throughput drug screen of a 386-drug anti-cancer compound library. The top candidates identified from this drug screen all demonstrated greater therapeutic efficacy than the standard-of-care drugs used in the clinic to treat triple-negative IBC, doxorubicin and paclitaxel. Our PDXEx model is simple, and we are confident that it can be incorporated into a PDX mouse system for use as a first-pass screening platform. This will permit the identification of effective tumor-specific therapies with high predictive value in a resource-, time-, and cost-efficient manner.
Collapse
Affiliation(s)
- Bedrich L. Eckhardt
- Department of Breast Medical Oncology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Maria Gagliardi
- Department of Breast Medical Oncology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - LaKesla Iles
- Department of Experimental Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Kurt Evans
- Department of Investigational Cancer Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiuping Liu
- Department of Experimental Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Glauco Souza
- Nano3D Biosciences, Houston, Texas, United States of America
- University of Texas Health Science Center, Houston, Texas, United States of America
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
| | - Geoffrey A. Bartholomeusz
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
- Department of Experimental Therapeutics, The University of Texas, MD, Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
22
|
Rhett JM, Yeh ES. The Potential for Connexin Hemichannels to Drive Breast Cancer Progression through Regulation of the Inflammatory Response. Int J Mol Sci 2018; 19:ijms19041043. [PMID: 29601539 PMCID: PMC5979453 DOI: 10.3390/ijms19041043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, connexin hemichannels have become recognized as major players in modulating the inflammatory response. Chronic inflammation is documented to promote tumorigenesis and is a critical component of tumor progression. Furthermore, inflammation is strongly linked to angiogenesis, immunotolerance, invasiveness, metastasis, and resistance in breast cancers. In this review, the literature on the role of connexin hemichannels in inflammation is summarized, and the potential role for hemichannel-mediated inflammation in driving breast cancer progression is discussed. Lastly, the potential for connexin-based therapeutics to modulate the inflammatory component of the tumor microenvironment as an avenue for the treatment of breast cancer is also discussed.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| |
Collapse
|
23
|
Cerón-Carrasco JP, Pérez-Sánchez H, Zúñiga J, Requena A. Antibodies as Carrier Molecules: Encapsulating Anti-Inflammatory Drugs inside Herceptine. J Phys Chem B 2018; 122:2064-2072. [DOI: 10.1021/acs.jpcb.7b10749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- José Pedro Cerón-Carrasco
- Bioinformatics
and High Performance Computing Research Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, 30107 Murcia, Spain
| | - Horacio Pérez-Sánchez
- Bioinformatics
and High Performance Computing Research Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, 30107 Murcia, Spain
| | - José Zúñiga
- Departamento
de Química Física, Universidad de Murcia, 30100 Murcia, Spain
| | - Alberto Requena
- Departamento
de Química Física, Universidad de Murcia, 30100 Murcia, Spain
| |
Collapse
|
24
|
Mohamed HT, El-Husseiny N, El-Ghonaimy EA, Ibrahim SA, Bazzi ZA, Cavallo-Medved D, Boffa MB, El-Shinawi M, Mohamed MM. IL-10 correlates with the expression of carboxypeptidase B2 and lymphovascular invasion in inflammatory breast cancer: The potential role of tumor infiltrated macrophages. Curr Probl Cancer 2018; 42:215-230. [PMID: 29459177 DOI: 10.1016/j.currproblcancer.2018.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/26/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022]
Abstract
Pro-carboxypeptidase B2 (pro-CPB2) or thrombin-activatable fibrinolysis inhibitor (TAFI) is a glycoprotein encoded by the CPB2 gene and deregulated in several cancer types, including breast cancer. Thrombin binding to thrombomodulin (TM), encoded by THBD, is important for TAFI activation. CPB2 gene expression is influenced by genetic polymorphism and cytokines such as interleukin 10 (IL-10). Our previous results showed that tumor infiltrating monocytes/macrophages (CD14+/CD16+) isolated from inflammatory breast cancer (IBC) patients' secrete high levels of IL-10. The aim of the present study is to test genetic polymorphism and expression of CPB2 in healthy breast tissues and carcinoma tissues of non-IBC and IBC patients. Furthermore, to investigate whether IL-10 modulates the expression of CPB2 and THBD in vivo and in-vitro. We tested CPB2 Thr325Ile polymorphism using restriction fragment length polymorphism, (RFLP) technique in healthy and carcinoma breast tissues. The mRNA expression of CPB2, THBD and IL10 were assessed by RT-qPCR. Infiltration of CD14+ cells was assessed by immunohistochemistry. In addition, we investigated the correlation between infiltration of CD14+ cells and expression of IL10 and CPB2. Furthermore, we correlated IL10 expression with the expression of both CPB2 and THBD in breast carcinoma tissues. Finally, we validated the role of recombinant IL-10 in regulating the expression of CPB2 and THBD using different breast cancer cell lines. Our results showed that CPB2 genotypes carrying the high-risk allele [Thr/Ile (CT) and Ile/Ile (TT)] were more frequent in both IBC and non-IBC patients compared to control group. CPB2 genotypes did not show any statistical correlation with CPB2 mRNA expression levels or patients' clinical pathological properties. Interestingly, CPB2 and IL10 expression were significantly higher and positively correlated with the incidence of CD14+ cells in carcinoma tissues of IBC as compared to non-IBC. On the other hand, THBD expression was significantly lower in IBC carcinoma versus non-IBC tissues. Based on molecular subtypes, CPB2 and IL10 expression were significantly higher in triple negative (TN) as compared to hormonal positive (HP) carcinoma tissues of IBC. Moreover, CPB2 expression was positively correlated with presence of lymphovascular invasion and the expression of IL10 in carcinoma tissues of IBC patients. Furthermore, recombinant human IL-10 stimulated CPB2 expression in SUM-149 (IBC cell line) but not in MDA-MB-231 (non-IBC cell line), while there was no significant effect THBD expression. In conclusion, carcinoma tissues of IBC patients are characterized by higher expression of CPB2 and lower expression of THBD. Moreover, CPB2 positively correlates with IL10 mRNA expression, incidence of CD14+ cells and lymphovascular invasion in IBC patients. IL-10 stimulated CPB2 expression in TN-IBC cell line suggests a relevant role of CPB2 in the aggressive phenotype of IBC.
Collapse
Affiliation(s)
| | - Noura El-Husseiny
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | | | - Zainab A Bazzi
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Dora Cavallo-Medved
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada
| | - Michael B Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Breast-Gynecological International Cancer Society, Cairo, Egypt
| | - Mona Mostafa Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt; Breast-Gynecological International Cancer Society, Cairo, Egypt.
| |
Collapse
|
25
|
Mehnath S, Arjama M, Rajan M, Jeyaraj M. Development of cholate conjugated hybrid polymeric micelles for FXR receptor mediated effective site-specific delivery of paclitaxel. NEW J CHEM 2018. [DOI: 10.1039/c8nj03251c] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of the present study was to explore the tumor targeting potential of a cholic acid (CA) conjugated polymeric micelle system for the effective delivery of paclitaxel (PTX).
Collapse
Affiliation(s)
- Sivaraj Mehnath
- Biomaterial and Nanomedicine Laboratory
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Guindy Campus
- Chennai
| | - Mukherjee Arjama
- Biomaterial and Nanomedicine Laboratory
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Guindy Campus
- Chennai
| | | | - Murugaraj Jeyaraj
- Biomaterial and Nanomedicine Laboratory
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Guindy Campus
- Chennai
| |
Collapse
|
26
|
Gutierrez Barrera AM, Fouad TM, Song J, Webster R, Elsayegh N, Wood AL, Demir A, Litton JK, Ueno NT, Arun BK. BRCA mutations in women with inflammatory breast cancer. Cancer 2017; 124:466-474. [PMID: 29044548 DOI: 10.1002/cncr.31069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/08/2017] [Accepted: 09/13/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Inflammatory breast cancer (IBC) often affects women at a relatively young age. To the authors' knowledge, the rate of BRCA variants among patients with IBC is not known. To determine the association between BRCA status and IBC, the authors evaluated its rate and compared the clinicopathologic characteristics of patients with IBC with those of patients with other breast cancers (non-IBC). METHODS Patients who presented at the study institution's cancer genetics program and who underwent BRCA genetic testing were included in the current study. The authors compared clinicopathologic data between patients with IBC and those with non-IBC using propensity score matching to identify predictors. RESULTS A total of 1789 patients who underwent BRCA genetic testing (1684 with non-IBC and 105 with IBC) were included. BRCA pathogenic variants were found in 27.3% of patients with non-IBC and 18.1% of patients with IBC (P = .0384). After propensity score matching, there were no significant differences noted between patients with IBC and those with non-IBC, including the rate of BRCA pathogenic variants (P = .5485). However, a subgroup analysis of the 479 patients with BRCA pathogenic variants demonstrated that patients with IBC (19 patients) were diagnosed at significantly younger ages compared with patients with non-IBC (P = .0244). CONCLUSIONS There was no clear association observed between BRCA pathogenic variants and IBC. However, among patients who tested positive for BRCA pathogenic variants, those with IBC were younger at the time of diagnosis compared with those with non-IBC breast cancers. These results confirm that genetic testing is important for patients with IBC who meet the current clinical criteria for genetic testing in breast cancer. Cancer 2018;124:466-74. © 2017 American Cancer Society.
Collapse
Affiliation(s)
| | - Tamer M Fouad
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Medical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rachel Webster
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nisreen Elsayegh
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita L Wood
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Atakan Demir
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Medical Oncology, Umraniye Research and Training Hospital, Umraniye, Istanbul, Turkey
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Banu K Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
27
|
Harano K, Kogawa T, Wu J, Yuan Y, Cohen EN, Lim B, Reuben JM, Ueno NT. Thrombocytosis as a prognostic factor in inflammatory breast cancer. Breast Cancer Res Treat 2017; 166:819-832. [PMID: 28831670 DOI: 10.1007/s10549-017-4463-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/12/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE Platelets are essential components of hemostasis and also play an important role in the tumor microenvironment. The purposes of our research were to examine the role of thrombocytosis in inflammatory breast cancer (IBC) and to know which cytokine drives thrombocytosis. METHODS We reviewed the medical records of 3654 patients with stage I-III breast cancer treated between 1998 and 2013, including 230 patients (6%) with IBC. We used Chi-squared test or Fisher's exact test to compare the variables between patients with and without thrombocytosis. Multivariate Cox regression models were used to determine the association of thrombocytosis with overall survival. We also examined baseline serum cytokine levels in 81 patients with primary IBC to determine the association of inflammatory cytokines with thrombocytosis. RESULTS We found that thrombocytosis was the only variable that predicted prognosis. Fifty-five patients (1.5%) had thrombocytosis. Thrombocytosis was more prevalent in patients with IBC than in those with non-IBC (3.4% vs. 1.4%, p = 0.015). In patients with IBC, thrombocytosis was associated with worse overall survival [hazard ratio 2.38, 95% confidence interval (CI) 1.05-5.4, p = 0.0378]. Circulating levels of growth-regulated oncogene (GRO) (odds ratio 1.003, 95% CI 1.001-1.005, p = 0.0019) and transforming growth factor β (TGF-β) (odds ratio 1.3, 95% CI 1.128-1.499, p = 0.0003) were associated with thrombocytosis. CONCLUSIONS Thrombocytosis was more prevalent in patients with IBC than in those with non-IBC and it was associated with poor prognosis. GRO and TGF-β were associated with thrombocytosis in IBC.
Collapse
Affiliation(s)
- Kenichi Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Medical Oncology, Nippon Medical School Musashikosugi Hospital, 1-396, Kosugi-cho, Nakahara-ku, Kawasaki City, Kanagawa, 211-8533, Japan
| | - Takahiro Kogawa
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jimin Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Evan N Cohen
- Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
|
29
|
Huang A, Cao S, Tang L. The tumor microenvironment and inflammatory breast cancer. J Cancer 2017; 8:1884-1891. [PMID: 28819386 PMCID: PMC5556652 DOI: 10.7150/jca.17595] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 04/01/2017] [Indexed: 01/25/2023] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and very aggressive subtype of breast cancer with clinical manifestations similar to acute inflammation. The prognosis of IBC is still poor even though combination therapy with surgery, chemotherapy, and target therapy, mainly due to a lack of fully understanding of the cellular and molecular mechanisms of IBC pathogenesis and progression. In the present article, we have comprehensively reviewed the connection of the pathogenesis of IBC and inflammation, immune reaction and cancer, particularly focused on the role and mechanism of tumor microenvironment related to IBC formation, tumor cell proliferation, migration, invasion and metastasis as well as the clinical manifestations of IBC. As the diverse cells including inflammatory cells, immune cells, and tumor cells and the soluble molecules produced by these cells in the microenvironment play an essential role in IBC development and progression. Therefore, anti-inflammatory therapy and immunotherapy with available agents warrant further investigation in the treatment of IBC.
Collapse
Affiliation(s)
- Aji Huang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shousong Cao
- Laboratory of Cancer Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lili Tang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
30
|
Anti-Inflammatory Mechanism Involved in Pomegranate-Mediated Prevention of Breast Cancer: the Role of NF-κB and Nrf2 Signaling Pathways. Nutrients 2017; 9:nu9050436. [PMID: 28452959 PMCID: PMC5452166 DOI: 10.3390/nu9050436] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Pomegranate (Punica granatum L.), a nutrient-rich unique fruit, has been used for centuries for the prevention and treatment of various inflammation-driven diseases. Based on our previous study, a characterized pomegranate emulsion (PE) exhibited a striking inhibition of dimethylbenz(a)anthracene (DMBA)-initiated rat mammary tumorigenesis via antiproliferative and apoptosis-inducing mechanisms. The objective of the present work is to investigate the anti-inflammatory mechanism of action of PE during DMBA rat mammary carcinogenesis by evaluating the expression of cyclooxygenase-2 (COX-2), heat shock protein 90 (HSP90), nuclear factor-κB (NF-κB) and nuclear factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2). Mammary tumor samples were harvested from our previous chemopreventive study in which PE (0.2–5.0 g/kg) was found to reduce mammary tumorigenesis in a dose-dependent manner. The expressions of COX-2, HSP90, NF-κB, inhibitory κBα (IκBα) and Nrf2 were detected by immunohistochemical techniques. PE decreased the expression of COX-2 and HSP90, prevented the degradation of IκBα, hindered the translocation of NF-κB from cytosol to nucleus and increased the expression and nuclear translocation of Nrf2 during DMBA-induced mammary tumorigenesis. These findings, together with our previous results, indicate that PE-mediated prevention of DMBA-evoked mammary carcinogenesis may involve anti-inflammatory mechanisms through concurrent but differential regulation of two interrelated molecular pathways, namely NF-κB and Nrf2 signaling.
Collapse
|
31
|
Eckhardt BL, Ueno NT. A target of potential RELAvance in inflammatory breast cancer. Oncotarget 2017; 8:25835-25836. [PMID: 28460487 PMCID: PMC5432219 DOI: 10.18632/oncotarget.17109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Bedrich L Eckhardt
- Department of Breast Medical Oncology, Section of Translational Breast Cancer Research, and The Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, Section of Translational Breast Cancer Research, and The Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Zhang Y, Guo Q, An S, Lu Y, Li J, He X, Liu L, Zhang Y, Sun T, Jiang C. ROS-Switchable Polymeric Nanoplatform with Stimuli-Responsive Release for Active Targeted Drug Delivery to Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2017; 9:12227-12240. [PMID: 28350451 DOI: 10.1021/acsami.6b16815] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Tumor microenvironment plays a vital role in the process of tumor development, proliferation, invasion, and metastasis. It is well acknowledged that reduction in pH, reactive oxygen species (ROS), and increased level of glucose transporter 1 (GLUT1) have become featured intracellular and extracellular biochemical markers of cancer owing to oncogenic transformation and abnormal metabolism. To establish a distinctive drug delivery system directed against the tumor microenvironment features, we develop a newly engineered polymeric nanoplatform for efficient doxorubicin (DOX) delivery with reduced systemic toxicity and high antitumor efficiency. A thioketal cross-linker is used to improve the formulation's stability during circulation and to foster quick intracellular drug release in response to tumor's ROS potential. Furthermore, the low drug loading efficiency of conventional micelles is ameliorated in this polymeric nanoplatform via a drug-conjugation strategy with an acid-labile chemical bond. The optimized formulation, MPLs-sB-DOX micelles, possesses a high drug-loading efficiency (31%) within nanosize diameter (37.8 nm). In addition, this formulation shows significant improvement in the pharmacokinetics and biodistribution profiles with a 2.69-fold increase of tumor accumulation, while with largely reduced systemic toxicity in comparison with free DOX. With advantages of efficient cellular uptake, preferential tumor accumulation, and controlled release behaviors, MPLs-sB-DOX micelles demonstrate good tumor-targeting ability with reduced systemic toxicity, proving to be a promising formulation for breast cancer therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Sai An
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Jianfeng Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Xi He
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
| | - Lisha Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education and Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
33
|
Fouad TM, Barrera AMG, Reuben JM, Lucci A, Woodward WA, Stauder MC, Lim B, DeSnyder SM, Arun B, Gildy B, Valero V, Hortobagyi GN, Ueno NT. Inflammatory breast cancer: a proposed conceptual shift in the UICC-AJCC TNM staging system. Lancet Oncol 2017; 18:e228-e232. [PMID: 28368261 PMCID: PMC6140765 DOI: 10.1016/s1470-2045(17)30192-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/22/2022]
Abstract
In the absence of histological criteria that distinguish between inflammatory and non-inflammatory breast cancer, diagnosis of inflammatory breast cancer relies entirely on the existence of clinical criteria as outlined by the TNM classification. This classification restricts patients presenting with clinical criteria characteristic of inflammatory breast cancer to subcategory T4d, which immediately relegates all patients with non-metastatic inflammatory breast cancer to stage 3, regardless of tumour size or nodal spread. Patients who present with metastatic disease are consigned to stage 4, and the TNM classification does not distinguish patients on the basis of the presence of inflammatory criteria. Evidence by our group and others suggests that patients with inflammatory breast cancer have significantly reduced overall survival among those who present with distant metastasis at diagnosis (stage 4). In light of these results, this Personal View addresses whether the current TNM staging classification accurately represents a distinction between patients with inflammatory and those with non-inflammatory breast cancer.
Collapse
Affiliation(s)
- Tamer M Fouad
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Medical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - James M Reuben
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anthony Lucci
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy A Woodward
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael C Stauder
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bora Lim
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah M DeSnyder
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu Arun
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Babiera Gildy
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
34
|
Zhao Y, Yang F, Li W, Xu C, Li L, Chen L, Liu Y, Sun P. miR-29a suppresses MCF-7 cell growth by downregulating tumor necrosis factor receptor 1. Tumour Biol 2017; 39:1010428317692264. [PMID: 28222663 DOI: 10.1177/1010428317692264] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor receptor 1 is the main receptor mediating many tumor necrosis factor-alpha-induced cellular events. Some studies have shown that tumor necrosis factor receptor 1 promotes tumorigenesis by activating nuclear factor-kappa B signaling pathway, while other studies have confirmed that tumor necrosis factor receptor 1 plays an inhibitory role in tumors growth by inducing apoptosis in breast cancer. Therefore, the function of tumor necrosis factor receptor 1 in breast cancer requires clarification. In this study, we first found that tumor necrosis factor receptor 1 was significantly increased in human breast cancer tissues and cell lines, and knockdown of tumor necrosis factor receptor 1 by small interfering RNA inhibited cell proliferation by arresting the cell cycle and inducing apoptosis. In addition, miR-29a was predicted as a regulator of tumor necrosis factor receptor 1 by TargetScan and was shown to be inversely correlated with tumor necrosis factor receptor 1 expression in human breast cancer tissues and cell lines. Luciferase reporter assay further confirmed that miR-29a negatively regulated tumor necrosis factor receptor 1 expression by binding to the 3' untranslated region. In our functional study, miR-29a overexpression remarkably suppressed cell proliferation and colony formation, arrested the cell cycle, and induced apoptosis in MCF-7 cell. Furthermore, in combination with tumor necrosis factor receptor 1 transfection, miR-29a significantly reversed the oncogenic role caused by tumor necrosis factor receptor 1 in MCF-7 cell. In addition, we demonstrated that miR-29a suppressed MCF-7 cell growth by inactivating the nuclear factor-kappa B signaling pathway and by decreasing cyclinD1 and Bcl-2/Bax protein levels. Taken together, our results suggest that miR-29a is an important regulator of tumor necrosis factor receptor 1 expression in breast cancer and functions as a tumor suppressor by targeting tumor necrosis factor receptor 1 to influence the growth of MCF-7 cell.
Collapse
Affiliation(s)
- Yiling Zhao
- 1 Department of Ultrasound, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Fenghua Yang
- 2 Department of Prevention and Health Statistics, Mudanjiang Medical University, Mudanjiang, China
| | - Wenyuan Li
- 3 Key Laboratory of Tumor Prevention and Treatment (Heilongjiang Higher Education Institutions), Mudanjiang Medical University, Mudanjiang, China
| | - Chunyan Xu
- 4 Department of Pathology, Tumor Hospital of Mudanjiang, Mudanjiang, China
| | - Li Li
- 3 Key Laboratory of Tumor Prevention and Treatment (Heilongjiang Higher Education Institutions), Mudanjiang Medical University, Mudanjiang, China
| | - Lifei Chen
- 5 Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yancui Liu
- 3 Key Laboratory of Tumor Prevention and Treatment (Heilongjiang Higher Education Institutions), Mudanjiang Medical University, Mudanjiang, China
| | - Ping Sun
- 3 Key Laboratory of Tumor Prevention and Treatment (Heilongjiang Higher Education Institutions), Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
35
|
Schairer C, Gadalla SM, Pfeiffer RM, Moore SC, Engels EA. Diabetes, Abnormal Glucose, Dyslipidemia, Hypertension, and Risk of Inflammatory and Other Breast Cancer. Cancer Epidemiol Biomarkers Prev 2017; 26:862-868. [PMID: 28087608 DOI: 10.1158/1055-9965.epi-16-0647] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Background: Obesity has been associated with substantially higher risk of inflammatory breast cancer (IBC) than other breast cancer. Here, we assess whether comorbidities of obesity, namely diabetes, abnormal glucose, dyslipidemia, and hypertension, are differentially related to risk of IBC and other breast cancers by tumor stage at diagnosis (localized/regional/distant/unstaged).Methods: We used linked SEER-Medicare data, with female breast cancer cases ages 66+ years identified by SEER registries (years 1992-2011). We divided first breast cancers into IBC (N = 2,306), locally advanced non-IBC (LABC; N = 10,347), and other (N = 197,276). We selected female controls (N = 200,000) from a stratified 5% random sample of Medicare recipients alive and breast cancer free. We assessed exposures until 12 months before diagnosis/selection using Medicare claims data. We estimated odds ratios (OR) and 99.9% confidence intervals (CI) using unconditional logistic regression.Results: Diabetes was associated with increased risk of distant IBC (98.5% of IBC cases; OR 1.44; 99.9% CI 1.21-1.71), distant (OR 1.24; 99.9% CI, 1.09-1.40) and regional (OR 1.29 (99.9% CI, 1.14-1.45) LABC, and distant (OR 1.23; 99.9% CI, 1.10-1.39) and unstaged (OR 1.32; 99.9% CI, 1.18-1.47) other breast cancers. Dyslipidemia was associated with reduced risk of IBC (OR 0.80; 95% CI, 0.67-0.94) and other breast cancers except localized disease. Results were similar by tumor estrogen receptor status. Abnormal glucose levels and hypertension had little association with risk of any tumor type.Conclusions: Associations with diabetes and dyslipidemia were similar for distant stage IBC and other advanced tumors.Impact: If confirmed, such findings could suggest avenues for prevention. Cancer Epidemiol Biomarkers Prev; 26(6); 862-8. ©2017 AACR.
Collapse
Affiliation(s)
- Catherine Schairer
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, NIH, Bethesda, Maryland.
| | - Shahinaz M Gadalla
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, NIH, Bethesda, Maryland
| | - Ruth M Pfeiffer
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, NIH, Bethesda, Maryland
| | - Steven C Moore
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, NIH, Bethesda, Maryland
| | - Eric A Engels
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, NIH, Bethesda, Maryland
| |
Collapse
|
36
|
Gutsche K, Randi EB, Blank V, Fink D, Wenger RH, Leo C, Scholz CC. Intermittent hypoxia confers pro-metastatic gene expression selectively through NF-κB in inflammatory breast cancer cells. Free Radic Biol Med 2016; 101:129-142. [PMID: 27717868 DOI: 10.1016/j.freeradbiomed.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/14/2016] [Accepted: 10/02/2016] [Indexed: 01/02/2023]
Abstract
Inflammatory breast cancer (IBC) is the most aggressive form of breast cancer. Treatment options are limited and the mechanisms underlying its aggressiveness are poorly understood. Intermittent hypoxia (IH) causes oxidative stress and is emerging as important regulator of tumor metastasis. Vessels in IBC tumors have been shown to be immature, which is a primary cause of IH. We therefore investigated the relevance of IH for the modulation of gene expression in IBC cells in order to assess IH as potential regulator of IBC aggressiveness. Gene array analysis of IBC cells following chronic IH (45-60 days) demonstrated increased expression of pro-metastatic genes of the extracellular matrix, such as tenascin-C (TNC; an essential factor of the metastatic niche) and matrix metalloproteinase 9 (MMP9), and of pro-inflammatory processes, such as cyclooxygenase-2 (COX-2). Investigating the oxidative stress-dependent regulation of TNC, we found a gradual sensitivity on mRNA and protein levels. Oxidative stress activated NF-E2-related factor 2 (Nrf2), c-Jun N-terminal kinase (JNK), c-Jun and nuclear factor κB (NF-κB), but TNC upregulation was only dependent on NF-κB activation. Pharmacological inhibition of inhibitor of NF-κB α (IκBα) phosphorylation as well as overexpression of IκBα prevented TNC, MMP9 and COX-2 induction, whereas the pro-inflammatory cytokine interleukin-1β (IL-1β) increased their expression levels. Analysis of the gene array data showed NF-κB binding sites for 64% of all upregulated genes, linking NF-κB with IH-dependent regulation of pro-metastatic gene expression in IBC cells. Our results provide a first link between intermittent hypoxia and pro-metastatic gene expression in IBC cells, revealing a putative novel mechanism for the high metastatic potential of IBC.
Collapse
Affiliation(s)
- Katrin Gutsche
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland; Department of Gynecology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Elisa B Randi
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Volker Blank
- Lady Davis Institute for Medical Research, Department of Medicine & Department of Physiology, McGill University, Montreal, Quebec, Canada H3T 1E2
| | - Daniel Fink
- Department of Gynecology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Cornelia Leo
- Department Women and Children, Cantonal Hospital Baden, 5404 Baden, Switzerland.
| | - Carsten C Scholz
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
37
|
Taminiau A, Draime A, Tys J, Lambert B, Vandeputte J, Nguyen N, Renard P, Geerts D, Rezsöhazy R. HOXA1 binds RBCK1/HOIL-1 and TRAF2 and modulates the TNF/NF-κB pathway in a transcription-independent manner. Nucleic Acids Res 2016; 44:7331-49. [PMID: 27382069 PMCID: PMC5009750 DOI: 10.1093/nar/gkw606] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/24/2016] [Indexed: 11/14/2022] Open
Abstract
HOX proteins define a family of key transcription factors regulating animal embryogenesis. HOX genes have also been linked to oncogenesis and HOXA1 has been described to be active in several cancers, including breast cancer. Through a proteome-wide interaction screening, we previously identified the TNFR-associated proteins RBCK1/HOIL-1 and TRAF2 as HOXA1 interactors suggesting that HOXA1 is functionally linked to the TNF/NF-κB signaling pathway. Here, we reveal a strong positive correlation between expression of HOXA1 and of members of the TNF/NF-κB pathway in breast tumor datasets. Functionally, we demonstrate that HOXA1 can activate NF-κB and operates upstream of the NF-κB inhibitor IκB. Consistently, we next demonstrate that the HOXA1-mediated activation of NF-κB is non-transcriptional and that RBCK1 and TRAF2 influences on NF-κB are epistatic to HOXA1. We also identify an 11 Histidine repeat and the homeodomain of HOXA1 to be required both for RBCK1 and TRAF2 interaction and NF-κB stimulation. Finally, we highlight that activation of NF-κB is crucial for HOXA1 oncogenic activity.
Collapse
Affiliation(s)
- Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Amandine Draime
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Janne Tys
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Barbara Lambert
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Julie Vandeputte
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Nathan Nguyen
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Patricia Renard
- Cellular Biology Research Unit, Université de Namur, Namur 5000, Belgium
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, Rotterdam 3015, The Netherlands
| | - René Rezsöhazy
- Animal Molecular and Cellular Biology Group (AMCB), Life Sciences Institute (ISV), Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| |
Collapse
|
38
|
Importance of hereditary and selected environmental risk factors in the etiology of inflammatory breast cancer: a case-comparison study. BMC Cancer 2016; 16:334. [PMID: 27229687 PMCID: PMC4881056 DOI: 10.1186/s12885-016-2369-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 05/18/2016] [Indexed: 12/18/2022] Open
Abstract
Background To assess the importance of heredity in the etiology of inflammatory breast cancer (IBC), we compared IBC patients to several carefully chosen comparison groups with respect to the prevalence of first-degree family history of breast cancer. Methods IBC cases (n = 141) were compared to non-inflammatory breast cancer cases (n = 178) ascertained through George Washington University (GWU) with respect to the prevalence of first-degree family history of breast cancer and selected environmental/lifestyle risk factors for breast cancer. Similar comparisons were conducted with subjects from three case–control studies: breast cancer cases (n = 1145) and unaffected controls (n = 1142) from the Cancer Genetic Markers of Susceptibility (CGEMS) study, breast cancer cases (n = 465) and controls (n = 9317) from the Women’s Health Initiative (WHI) study, and ovarian cancer cases (n = 260) and controls (n = 331) from a study by University of Toronto (UT). Results The frequency of first-degree breast cancer family history among IBC cases was 17.0 % compared to 24.4 % for GWU breast cancer cases, 23.9 % and 17.9 % for CGEMS breast cancer cases and controls, respectively, 16.9 % and 12.6 % for WHI breast cancer cases and controls, respectively, and 24.2 % and 11.2 % for UT ovarian cancer cases and controls, respectively. IBC cases had a significantly lower prevalence of parous women than WHI breast cancer cases (OR = 0.46, 95 % CI:0.27–0.81) and controls (OR = 0.31, 95 % CI:0.20–0.49). Oral contraceptive use was significantly higher among IBC cases compared to WHI breast cancer cases (OR = 7.77, 95 % CI:4.82–12.59) and controls (OR = 8.14, 95 % CI:5.28–12.61). IBC cases had a significantly higher frequency of regular alcohol consumption (≥1 drink per day) compared to WHI controls (OR = 1.84, 95 % CI:1.20–2.82) and UT controls (OR = 1.86, 95 % CI:1.07–3.22) and higher (statistically non-significant) prevalence (21.3 %) compared to breast cancer cases from GWU (18.2 %) and WHI (15.2 %). Conclusions The prevalence of first-degree breast cancer family history among IBC cases was lower compared to breast and ovarian cancer cases but higher than unaffected individuals. Our multiple-case inflammatory and non-inflammatory breast cancer families may reflect aggregation of common genetic and/or environmental factors predisposing to both types of breast cancer. Our findings that oral contraceptive use and regular alcohol consumption may be associated with IBC warrant further investigations.
Collapse
|
39
|
Shi CJ, Wen XS, Gao HF, Liu ZH, Xu XK, Li LF, Shen T, Xian CJ. Steamed root of Rehmannia glutinosa Libosch (Plantaginaceae) alleviates methotrexate-induced intestinal mucositis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 183:143-150. [PMID: 26934449 DOI: 10.1016/j.jep.2016.02.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/20/2016] [Accepted: 02/25/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intestinal mucositis induced by chemotherapy is a severe clinical problem in cancer patients that currently lack effective interventions. In traditional Chinese medicine, chemotherapeutic toxicity is diagnosed as Qi and Yin deficiency, and steamed rehmannia root (SRR) is frequently prescribed to these patients. Whether SRR can prevent the adverse effects remains to be confirmed experimentally. The present study used a rat model to investigate potential efficacy and action mechanisms of SRR in attenuating the adverse effects caused by chemotherapy. MATERIALS AND METHODS Intraperitoneal injection of a single dose of anti-metabolite methotrexate (MTX, 25mg/kg) was given to adult Wistar rats, which also received oral gavage of water or SRR (1.08g/kg twice daily 3 days before and 4 days after MTX treatment), or calcium folinate (CF, a clinically used MTX antidote as a comparison, at 1mg/kg twice daily 36h after MTX treatment), or SRR and CF in combination. Animals were sacrificed 4 days after MTX treatment. Complete blood cell counting was carried out. Jejunum was analyzed histologically for mucosal damage, immunohistochemically for proliferating cell nuclear antigen (PCNA), and biochemically for thiobarbituric acid reactive substances (TBARS) and reduced glutathione (GSH), as well as for tumor necrosis factor alpha (TNF-α). RESULTS MTX treatment led to weight loss, leucopenia, polycythemia, increase in large thrombocyte ratio, intestinal villus atrophy, crypt loss and reduction in PCNA positive crypt cells, increases in mucosal TBARS and TNF-α and decrease in GSH. All these alterations were inhibited by SRR administration except leucopenia, and the effects of CF or CF plus SRR supplementation were found to be inferior to those of SRR. CONCLUSIONS SRR can alleviate MTX-induced gut mucositis, which may be achieved by inhibiting MTX-induced oxidative stress and inflammatory response. These findings support the application of SRR in chemotherapy but not the combined application of SRR and CF.
Collapse
Affiliation(s)
- Cheng-Jin Shi
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xue-Sen Wen
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| | - Hui-Feng Gao
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zhi-Hua Liu
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xu-Kang Xu
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Li-Fen Li
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Tao Shen
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
40
|
Hamm CA, Moran D, Rao K, Trusk PB, Pry K, Sausen M, Jones S, Velculescu VE, Cristofanilli M, Bacus S. Genomic and Immunological Tumor Profiling Identifies Targetable Pathways and Extensive CD8+/PDL1+ Immune Infiltration in Inflammatory Breast Cancer Tumors. Mol Cancer Ther 2016; 15:1746-56. [PMID: 27196778 DOI: 10.1158/1535-7163.mct-15-0353] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 04/13/2016] [Indexed: 11/16/2022]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer that remains poorly understood at the molecular level. Comprehensive tumor profiling was performed to understand clinically actionable alterations in IBC. Targeted next-generation sequencing (NGS) and IHC were performed to identify activated pathways in IBC tumor tissues. siRNA studies examined the impact of IBC genomic variants in cellular models. IBC tumor tissues were further characterized for immune infiltration and immune checkpoint expression by IHC. Genomic analysis identified recurrent alterations in core biologic pathways, including activating and targetable variants in HER/PI3K/mTOR signaling. High rates of activating HER3 point mutations were discovered in IBC tumors. Cell line studies confirmed a role for mutant HER3 in IBC cell proliferation. Immunologic analysis revealed a subset of IBC tumors associated with high CD8(+)/PD-L1(+) lymphocyte infiltration. Immune infiltration positively correlated with an NGS-based estimate of neoantigen exposure derived from the somatic mutation rate and mutant allele frequency, iScore. Additionally, DNA mismatch repair alterations, which may contribute to higher iScores, occurred at greater frequency in tumors with higher immune infiltration. Our study identifies genomic alterations that mechanistically contribute to oncogenic signaling in IBC and provides a genetic basis for the selection of clinically relevant targeted and combination therapeutic strategies. Furthermore, an NGS-based estimate of neoantigen exposure developed in this study (iScore) may be a useful biomarker to predict immune infiltration in IBC and other cancers. The iScore may be associated with greater levels of response to immunotherapies, such as PD-L1/PD-1-targeted therapies. Mol Cancer Ther; 15(7); 1746-56. ©2016 AACR.
Collapse
Affiliation(s)
| | - Diarmuid Moran
- Translational R&D Oncology Group, Quintiles, Westmont, Illinois. Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois.
| | - Kakuturu Rao
- Translational R&D Oncology Group, Quintiles, Westmont, Illinois
| | | | - Karen Pry
- Translational R&D Oncology Group, Quintiles, Westmont, Illinois
| | - Mark Sausen
- Personal Genome Diagnostics, Inc., Baltimore, Maryland
| | - Siân Jones
- Personal Genome Diagnostics, Inc., Baltimore, Maryland
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Massimo Cristofanilli
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Sarah Bacus
- Translational R&D Oncology Group, Quintiles, Westmont, Illinois. Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
41
|
Whitfield M, Ouvrier A, Cadet R, Damon-Soubeyrand C, Guiton R, Janny L, Kocer A, Marceau G, Pons-Rejraji H, Trousson A, Drevet JR, Saez F. Liver X Receptors (LXRs) Alpha and Beta Play Distinct Roles in the Mouse Epididymis1. Biol Reprod 2016; 94:55. [DOI: 10.1095/biolreprod.115.133538] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/11/2016] [Indexed: 01/07/2023] Open
|
42
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
43
|
Hurwitz HI, Uppal N, Wagner SA, Bendell JC, Beck JT, Wade SM, Nemunaitis JJ, Stella PJ, Pipas JM, Wainberg ZA, Manges R, Garrett WM, Hunter DS, Clark J, Leopold L, Sandor V, Levy RS. Randomized, Double-Blind, Phase II Study of Ruxolitinib or Placebo in Combination With Capecitabine in Patients With Metastatic Pancreatic Cancer for Whom Therapy With Gemcitabine Has Failed. J Clin Oncol 2015; 33:4039-47. [PMID: 26351344 DOI: 10.1200/jco.2015.61.4578] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Patients with advanced pancreatic adenocarcinoma have a poor prognosis and limited second-line treatment options. Evidence suggests a role for the Janus kinase (JAK)/signal transducer and activator of transcription pathway in the pathogenesis and clinical course of pancreatic cancer. PATIENTS AND METHODS In this double-blind, phase II study, patients with metastatic pancreatic cancer who had experienced treatment failure with gemcitabine were randomly assigned 1:1 to the JAK1/JAK2 inhibitor ruxolitinib (15 mg twice daily) plus capecitabine (1,000 mg/m(2) twice daily) or placebo plus capecitabine. The primary end point was overall survival (OS); secondary end points included progression-free survival, clinical benefit response, objective response rate, and safety. Prespecified subgroup analyses evaluated treatment heterogeneity and efficacy in patients with evidence of inflammation. RESULTS In the intent-to-treat population (ruxolitinib, n = 64; placebo, n = 63), the hazard ratio was 0.79 (95% CI, 0.53 to 1.18; P = .25) for OS and was 0.75 (95% CI, 0.52 to 1.10; P = .14) for progression-free survival. In a prespecified subgroup analysis of patients with inflammation, defined by serum C-reactive protein levels greater than the study population median (ie, 13 mg/L), OS was significantly greater with ruxolitinib than with placebo (hazard ratio, 0.47; 95% CI, 0.26 to 0.85; P = .011). Prolonged survival in this subgroup was supported by post hoc analyses of OS that categorized patients by the modified Glasgow Prognostic Score, a systemic inflammation-based prognostic system. Grade 3 or greater adverse events were observed with similar frequency in the ruxolitinib (74.6%) and placebo (81.7%) groups. Grade 3 or greater anemia was more frequent with ruxolitinib (15.3%; placebo, 1.7%). CONCLUSION Ruxolitinib plus capecitabine was generally well tolerated and may improve survival in patients with metastatic pancreatic cancer and evidence of systemic inflammation.
Collapse
Affiliation(s)
- Herbert I Hurwitz
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE.
| | - Nikhil Uppal
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Stephanie A Wagner
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Johanna C Bendell
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - J Thaddeus Beck
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Seaborn M Wade
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - John J Nemunaitis
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Philip J Stella
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - J Marc Pipas
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Zev A Wainberg
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Robert Manges
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - William M Garrett
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Deborah S Hunter
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Jason Clark
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Lance Leopold
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Victor Sandor
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Richard S Levy
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| |
Collapse
|
44
|
Hurwitz HI, Uppal N, Wagner SA, Bendell JC, Beck JT, Wade SM, Nemunaitis JJ, Stella PJ, Pipas JM, Wainberg ZA, Manges R, Garrett WM, Hunter DS, Clark J, Leopold L, Sandor V, Levy RS. Randomized, Double-Blind, Phase II Study of Ruxolitinib or Placebo in Combination With Capecitabine in Patients With Metastatic Pancreatic Cancer for Whom Therapy With Gemcitabine Has Failed. J Clin Oncol 2015. [PMID: 26351344 DOI: 10.1200/jco.2015.61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Patients with advanced pancreatic adenocarcinoma have a poor prognosis and limited second-line treatment options. Evidence suggests a role for the Janus kinase (JAK)/signal transducer and activator of transcription pathway in the pathogenesis and clinical course of pancreatic cancer. PATIENTS AND METHODS In this double-blind, phase II study, patients with metastatic pancreatic cancer who had experienced treatment failure with gemcitabine were randomly assigned 1:1 to the JAK1/JAK2 inhibitor ruxolitinib (15 mg twice daily) plus capecitabine (1,000 mg/m(2) twice daily) or placebo plus capecitabine. The primary end point was overall survival (OS); secondary end points included progression-free survival, clinical benefit response, objective response rate, and safety. Prespecified subgroup analyses evaluated treatment heterogeneity and efficacy in patients with evidence of inflammation. RESULTS In the intent-to-treat population (ruxolitinib, n = 64; placebo, n = 63), the hazard ratio was 0.79 (95% CI, 0.53 to 1.18; P = .25) for OS and was 0.75 (95% CI, 0.52 to 1.10; P = .14) for progression-free survival. In a prespecified subgroup analysis of patients with inflammation, defined by serum C-reactive protein levels greater than the study population median (ie, 13 mg/L), OS was significantly greater with ruxolitinib than with placebo (hazard ratio, 0.47; 95% CI, 0.26 to 0.85; P = .011). Prolonged survival in this subgroup was supported by post hoc analyses of OS that categorized patients by the modified Glasgow Prognostic Score, a systemic inflammation-based prognostic system. Grade 3 or greater adverse events were observed with similar frequency in the ruxolitinib (74.6%) and placebo (81.7%) groups. Grade 3 or greater anemia was more frequent with ruxolitinib (15.3%; placebo, 1.7%). CONCLUSION Ruxolitinib plus capecitabine was generally well tolerated and may improve survival in patients with metastatic pancreatic cancer and evidence of systemic inflammation.
Collapse
Affiliation(s)
- Herbert I Hurwitz
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE.
| | - Nikhil Uppal
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Stephanie A Wagner
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Johanna C Bendell
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - J Thaddeus Beck
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Seaborn M Wade
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - John J Nemunaitis
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Philip J Stella
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - J Marc Pipas
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Zev A Wainberg
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Robert Manges
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - William M Garrett
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Deborah S Hunter
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Jason Clark
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Lance Leopold
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Victor Sandor
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| | - Richard S Levy
- Herbert I. Hurwitz, Duke University Medical Center, Durham, NC; Nikhil Uppal, New York University Langone Arena Oncology, Lake Success, NY; Stephanie A. Wagner, Indiana University Melvin and Bren Simon Cancer Center; Robert Manges, Investigative Clinical Research of Indiana, Indianapolis, IN; Johanna C. Bendell, Sarah Cannon Research Institute, Nashville, TN; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Seaborn M. Wade III, Virginia Cancer Institute, Richmond, VA; John J. Nemunaitis, Mary Crowley Medical Research Center, Dallas, TX; Philip J. Stella, St Joseph Mercy Health System, Alexander Cancer Care Center, Ann Arbor, MI; J. Marc Pipas, Dartmouth Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH; Zev A. Wainberg, University of California, Los Angeles, Los Angeles, CA; and William M. Garrett, Deborah S. Hunter, Jason Clark, Lance Leopold, Victor Sandor, and Richard S. Levy, Incyte Corporation, Wilmington, DE
| |
Collapse
|
45
|
Mandal A, Bishayee A. Trianthema portulacastrum Linn. displays anti-inflammatory responses during chemically induced rat mammary tumorigenesis through simultaneous and differential regulation of NF-κB and Nrf2 signaling pathways. Int J Mol Sci 2015; 16:2426-45. [PMID: 25622256 PMCID: PMC4346844 DOI: 10.3390/ijms16022426] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/13/2015] [Indexed: 12/19/2022] Open
Abstract
Trianthema portulacastrum, a medicinal and dietary plant, has gained substantial importance due to its various pharmacological properties, including anti-inflammatory and anticarcinogenic activities. We have recently reported that a characterized T. portulacastrum extract (TPE) affords a considerable chemoprevention of 7,12-dimethylbenz(a)anthracene (DMBA)-induced rat mammary tumorigenesis though the underlying mechanisms are not completely understood. The objective of this study was to investigate anti-inflammatory mechanisms of TPE during DMBA mammary carcinogenesis in rats by monitoring cyclooxygenase-2 (COX-2), heat shock protein 90 (HSP90), nuclear factor-kappaB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2). Mammary tumors were harvested from our previous study in which TPE (50-200 mg/kg) was found to inhibit mammary tumorigenesis in a dose-response manner. The expressions of intratumor COX-2, HSP90, NF-κB, inhibitory kappaB-alpha (IκBα) and Nrf2 were determined by immunohistochemistry. TPE downregulated the expression of COX-2 and HSP90, blocked the degradation of IκBα, hampered the translocation of NF-κB from cytosol to nucleus and upregulated the expression and nuclear translocation of Nrf2 during DMBA mammary carcinogenesis. These results in conjunction with our previous findings suggest that TPE prevents DMBA-induced breast neoplasia by anti-inflammatory mechanisms mediated through simultaneous and differential modulation of two interconnected molecular circuits, namely NF-κB and Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Animesh Mandal
- Cancer Therapeutics and Chemoprevention Group, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| | - Anupam Bishayee
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA.
| |
Collapse
|
46
|
Boshier PR, Sayers R, Hadjiminas DJ, Mackworth-Young C, Cleator S, Leff DR. Systemic inflammatory response syndrome in a patient diagnosed with high grade inflammatory triple negative breast cancer: a case report of a potentially rare paraneoplastic syndrome. Exp Hematol Oncol 2015; 5:16. [PMID: 27340609 PMCID: PMC4917943 DOI: 10.1186/s40164-016-0045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/09/2016] [Indexed: 11/10/2022] Open
Abstract
Background Inflammatory breast cancer is a complex pathological entity associated with poor outcomes. This loco-regional disease is characterised by a rapid clinical course in the presence breast erythema and infiltration of dermal lymphatics by tumours cells. Herein we describe a case of inflammatory breast cancer with a rare presentation and disease course defined by a profound systemic inflammatory response in the absence of an infective cause. Case presentation The patient presented with pyrexia and malaise following a recent tissue diagnosis of inflammatory breast cancer. At the time of admission the patient demonstrated clinical features of the systemic inflammatory response syndrome (SIRS) in the presence of a negative septic screen. Her condition deteriorated despite systemic broad spectrum intravenous antibiotics and she underwent surgical debulking of a 180 × 135 × 100 mm (821 g) primary tumour composed of oedematous, friable and haemorrhagic tissue (pT4,N1a,M0; oestrogen/progesterone/HER-2 receptor negative). Following surgery, the clinical picture dramatically improved with cessation of SIRS and normalisation of inflammatory markers. After 4 weeks the patient required readmission to hospital due to recurrent SIRS and negative septic screen. The patient received treatment with systemic chemotherapy showing transient clinical improvement and suppression of SIRS. Despite on going chemotherapy, systemic antibiotics and a trial of steroid therapy the patient died 5 months after her initial presentation to hospital. At the time of death she demonstrated persistent SIRS with elevated inflammatory markers. Conclusion This is the first case report of inflammatory breath cancer associated with SIRS in the absence of clinically confirmed infection. Important learning points highlighted by this case are: (a) recognition of the diagnostic and therapeutic uncertainties that still exist in the context of inflammatory breast cancer; (b) appreciation of the potential paraneoplastic systemic inflammatory manifestations of this disease, and finally; (c) the importance a multidisciplinary and multimodal approach to treatment.
Collapse
Affiliation(s)
- Piers R Boshier
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Rosie Sayers
- Department of Breast Surgery, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
| | - Dimitri J Hadjiminas
- Department of Breast Surgery, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
| | - Charles Mackworth-Young
- Department of Rheumatology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
| | - Susan Cleator
- Department of Oncology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
| | - Daniel R Leff
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
47
|
Xu S, Yang S, Sun G, Huang W, Zhang Y. Transforming growth factor-beta polymorphisms and serum level in the development of osteosarcoma. DNA Cell Biol 2014; 33:802-6. [PMID: 25098449 DOI: 10.1089/dna.2014.2527] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The transforming growth factor-beta (TGF-β) signaling pathway plays critical roles in the development of various diseases. The current study investigated whether TGF-β was involved in the pathogenesis of osteosarcoma from the genetic polymorphism perspective and serum level perspective. We first examined two TGF-β1 polymorphisms, rs1800469C/T and rs1800470T/C, in 202 osteosarcoma patients and 216 healthy controls. Data revealed that the prevalence of rs1800470TT genotype and T allele was significantly elevated in patients than in controls (odds ratio [OR]=2.28, 95% confidence interval [CI]: 1.30-3.98, p<0.001, and OR=1.49, 95% CI: 1.14-1.96, p<0.001). Function analyses showed that healthy controls carrying rs1800470TT genotype had a significantly higher serum level of TGF-β than those carrying the rs1800470CC genotype (191.1±15.7 pg/mL vs. 129.4±10.9 pg/mL, p=0.003). We then compared the serum level of TGF-β between osteosarcoma patients and healthy controls. Results demonstrated a significantly increased serum level of TGF-β in patients than in controls. Further analyses identified that patients with metastasis had augmented levels of serum TGF-β than those without metastasis. These data indicate that TGF-β may be closely involved in the pathogenesis of osteosarcoma.
Collapse
Affiliation(s)
- Shaogang Xu
- 1 Department of Orthopedics, Zhengzhou Orthopedics Hospital , Zhengzhou, Henan, China
| | | | | | | | | |
Collapse
|