1
|
Perioperative escape from dormancy of spontaneous micro-metastases: A role for malignant secretion of IL-6, IL-8, and VEGF, through adrenergic and prostaglandin signaling. Brain Behav Immun 2023; 109:175-187. [PMID: 36646396 DOI: 10.1016/j.bbi.2023.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/08/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
We recently showed that a minimally-invasive removal of MDA-MB-231HM primary tumors (PTs) and elimination of their secreted factors (including IL-6, IL-8, VEGF, EGF, PDGF-aa, MIF, SerpinE1, and M-CSF), caused regression of spontaneous micro-metastases into a non-growing dormant state. To explore the underlying mechanisms and potential clinical ramifications of this phenomenon, we herein used the MDA-MB-231HM human breast cancer cell-line, in-vitro, and in vivo following orthotopic implantation in immune-deficient BALB/C nu/nu mice. Employing bioluminescence imaging, we found that adding laparotomy to minimally-invasive removal of the PT caused an outbreak of micro-metastases. However, perioperative β-adrenergic and COX-2 inhibition, using propranolol + etodolac, maintained metastatic dormancy following laparotomy. In-vitro, β-adrenergic agonists (epinephrine or metaproterenol) and prostaglandin-E2 markedly increased MDA-MB-231HM secretion of the pro-metastatic factors IL-6, IL-8, and VEGF, whereas cortisol reduced their secretion, effects that were maintained even 12 h after the washout of these agonists. In-vivo, laparotomy elevated IL-6 and IL-8 levels in both plasma and ex-vivo PT spontaneous secretion, whereas perioperative propranolol + etodolac administration blocked these effects. Similar trends were evident for EGF and MIF. Promoter-based bioinformatics analyses of excised PT transcriptomes implicated elevated NF-kB activity and reduced IRF1 activity in the gene regulatory effects of laparotomy, and these effects were inhibited by pre-surgical propranolol + etodolac. Taken together, our findings suggest a novel mechanism of post-operative metastatic outbreak, where surgery-induced adrenergic and prostanoid signaling increase the secretion of pro-metastatic factors, including IL-6, IL-8, and VEGF, from PT and possibly residual malignant tissue, and thereby prevent residual disease from entering dormancy.
Collapse
|
2
|
OUP accepted manuscript. Clin Chem 2022; 68:973-983. [DOI: 10.1093/clinchem/hvac073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022]
|
3
|
Bushnell GG, Deshmukh AP, den Hollander P, Luo M, Soundararajan R, Jia D, Levine H, Mani SA, Wicha MS. Breast cancer dormancy: need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer 2021; 7:66. [PMID: 34050189 PMCID: PMC8163741 DOI: 10.1038/s41523-021-00269-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/08/2021] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in the USA. Although advances in treatment over the past several decades have significantly improved the outlook for this disease, most women who are diagnosed with estrogen receptor positive disease remain at risk of metastatic relapse for the remainder of their life. The cellular source of late relapse in these patients is thought to be disseminated tumor cells that reactivate after a long period of dormancy. The biology of these dormant cells and their natural history over a patient's lifetime is largely unclear. We posit that research on tumor dormancy has been significantly limited by the lack of clinically relevant models. This review will discuss existing dormancy models, gaps in biological understanding, and propose criteria for future models to enhance their clinical relevance.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Abhijeet P Deshmukh
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Luo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics and Departments of Physics and Bioengineering, Northeastern University, Boston, MA, USA.
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Akkoc Y, Peker N, Akcay A, Gozuacik D. Autophagy and Cancer Dormancy. Front Oncol 2021; 11:627023. [PMID: 33816262 PMCID: PMC8017298 DOI: 10.3389/fonc.2021.627023] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Metastasis and relapse account for the great majority of cancer-related deaths. Most metastatic lesions are micro metastases that have the capacity to remain in a non-dividing state called “dormancy” for months or even years. Commonly used anticancer drugs generally target actively dividing cancer cells. Therefore, cancer cells that remain in a dormant state evade conventional therapies and contribute to cancer recurrence. Cellular and molecular mechanisms of cancer dormancy are not fully understood. Recent studies indicate that a major cellular stress response mechanism, autophagy, plays an important role in the adaptation, survival and reactivation of dormant cells. In this review article, we will summarize accumulating knowledge about cellular and molecular mechanisms of cancer dormancy, and discuss the role and importance of autophagy in this context.
Collapse
Affiliation(s)
- Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Nesibe Peker
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Arzu Akcay
- Yeni Yüzyıl University, School of Medicine, Private Gaziosmanpaşa Hospital, Department of Pathology, Istanbul, Turkey
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Koç University School of Medicine, Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| |
Collapse
|
5
|
Werner S, Brors B, Eick J, Marques E, Pogenberg V, Parret A, Kemming D, Wood AW, Edgren H, Neubauer H, Streichert T, Riethdorf S, Bedi U, Baccelli I, Jücker M, Eils R, Fehm T, Trumpp A, Johnsen SA, Klefström J, Wilmanns M, Müller V, Pantel K, Wikman H. Suppression of early hematogenous dissemination of human breast cancer cells to bone marrow by retinoic Acid-induced 2. Cancer Discov 2015; 5:506-19. [PMID: 25716347 DOI: 10.1158/2159-8290.cd-14-1042] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/18/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Regulatory pathways that drive early hematogenous dissemination of tumor cells are insufficiently defined. Here, we used the presence of disseminated tumor cells (DTC) in the bone marrow to define patients with early disseminated breast cancer and identified low retinoic acid-induced 2 (RAI2) expression to be significantly associated with DTC status. Low RAI2 expression was also shown to be an independent poor prognostic factor in 10 different cancer datasets. Depletion of RAI2 protein in luminal breast cancer cell lines resulted in dedifferentiation marked by downregulation of ERα, FOXA1, and GATA3, together with increased invasiveness and activation of AKT signaling. Functional analysis of the previously uncharacterized RAI2 protein revealed molecular interaction with CtBP transcriptional regulators and an overlapping function in controlling the expression of a number of key target genes involved in breast cancer. These results suggest that RAI2 is a new metastasis-associated protein that sustains differentiation of luminal breast epithelial cells. SIGNIFICANCE We identified downregulation of RAI2 as a novel metastasis-associated genetic alteration especially associated with early occurring bone metastasis in ERα-positive breast tumors. We specified the role of the RAI2 protein to function as a transcriptional regulator that controls the expression of several key regulators of breast epithelial integrity and cancer.
Collapse
Affiliation(s)
- Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics (G200), German Cancer Research Center (DKFZ), Heidelberg, Germany. National Center for Tumor Diseases (NCT), Heidelberg, Germany. German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Julia Eick
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elsa Marques
- Translational Cancer Biology Research Program and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | | | | | - Dirk Kemming
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. European Laboratory Association, Ibbenbüren, Germany
| | - Antony W Wood
- Cell Signaling Technology, Inc., Danvers, Massachusetts
| | - Henrik Edgren
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hans Neubauer
- Department of Obstetrics and Gynecology, University of Duesseldorf, Duesseldorf, Germany. Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Thomas Streichert
- Department of Clinical Chemistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Upasana Bedi
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irène Baccelli
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Eils
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), Heidelberg, Germany. Institute of Pharmacy and Molecular Biotechnology, and Bioquant Center, University of Heidelberg, Heidelberg, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, University of Duesseldorf, Duesseldorf, Germany. Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steven A Johnsen
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juha Klefström
- Translational Cancer Biology Research Program and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | | | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
6
|
|
7
|
Tumor dormancy and cancer stem cells: two sides of the same coin? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:145-79. [PMID: 23143979 DOI: 10.1007/978-1-4614-1445-2_8] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Increasing evidence suggests that tumor dormancy represents an important mechanism underlying the observed failure of existing therapeutic modalities to fully eradicate cancers. In addition to its more established role in maintaining minimal residual disease after treatment, dormancy might also critically contribute to early stages of tumor development and the formation of clinically undetectable micrometastatic foci. There are striking parallels between the concept of tumor dormancy and the cancer stem cell (CSC) theory of tumor propagation. For instance, the CSC hypothesis similarly predicts that a subset of self-renewing cancer cells-that is CSCs-is responsible for tumor initiation, bears the preferential ability to survive tumor therapy, and persists long term to ultimately cause delayed cancer recurrence and metastatic progression. Additionally, many of the biological mechanisms involved in controlling the dormant state of a tumor can also govern CSC behavior, including cell cycle modifications, alteration of angiogenic processes, and modulation of antitumor immune responses. In fact, quiescence and immune escape are emerging hallmark features of at least some CSCs, indicating significant overlap between dormant cancer populations and CSCs. Herein, we crucially dissect whether CSCs occupy specific roles in orchestrating the switch between dormancy and exuberant tumor growth. We elucidate how recently uncovered CSC biological features could enable these cells to evade immunologic clearance and regulate cancer expansion, relapse, and progression. We propose that the study of CSC immunobiological pathways holds the promise to critically advance our understanding of the processes mediating tumor dormancy. Ultimately, such research endeavors could unravel novel therapeutic avenues that efficiently target both proliferating and dormant CSCs to minimize the risk of tumor recurrence in cancer patients.
Collapse
|
8
|
Kordon EC. MMTV-induced pregnancy-dependent mammary tumors : early history and new perspectives. J Mammary Gland Biol Neoplasia 2008; 13:289-97. [PMID: 18661103 DOI: 10.1007/s10911-008-9091-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 07/04/2008] [Indexed: 11/28/2022] Open
Abstract
Almost 60 years ago, Foulds carefully described for the first time a particular type of mouse mammary tumor that appeared in the glands of pregnant females and disappeared shortly after delivery. Since then, the attention that researchers paid to the Mouse Mammary Tumor Virus (MMTV)-induced pregnancy-dependent tumors has not vanished through the years. This was because the information obtained from mice carrying MMTV variants that were able to induce pregnancy-dependent tumors was meaningful for studying different aspects of mammary tumor biology. In addition, mice infected with these viral variants provided some of the few chances to use fully hormone-dependent estrogen receptor positive breast cancer models in the mouse. In the analysis of the association between tumor morphology and behavior, the mechanisms underlying progression towards autonomy, the impact of different genes during cancer initiation and development, and the relevance of host genetic background for tumor incidence and hormone-dependence, mouse strains carrying these MMTV variants have been very important tools that could not have been replaced with any other available model. The goal of this article is to provide a succinct chronicle of the experiments and observations made in the MMTV-induced pregnancy-dependent models that most significantly contributed to the mouse mammary tumor biology field. In addition, the possibility to use these MMTV variants as alternative models for analyzing mammary tumor stem cells and pregnancy-associated breast cancer in women is discussed.
Collapse
Affiliation(s)
- Edith C Kordon
- Institute of Physiology, Molecular Biology and Neurosciences (IFIBYNE-CONICET), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Roth TM, Petty EM, Barald KF. The role of steroid hormones in the NF1 phenotype: focus on pregnancy. Am J Med Genet A 2008; 146A:1624-33. [PMID: 18481270 DOI: 10.1002/ajmg.a.32301] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Neurofibromatosis Type 1 (NF1) gene functions as a tumor suppressor gene. Loss of its protein, neurofibromin, in the autosomal dominant disorder NF1 is associated with peripheral nervous system tumors, particularly neurofibromas, benign lesions in which the major cell type is the Schwann Cell (SC). Benign and malignant human tumors found in NF1 patients are heterogeneous with respect to their cellular composition. The number and size of neurofibromas in NF1 patients has been shown to increase during pregnancy, with, in some cases, post-partum regression, which suggests hormonal involvement in this increase. However, in this review, we consider evidence from the literature that both direct hormonal influence on tumor growth and on angiogenesis may contribute to these effects.
Collapse
Affiliation(s)
- Therese M Roth
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | |
Collapse
|
10
|
Roth TM, Ramamurthy P, Muir D, Wallace MR, Zhu Y, Chang L, Barald KF. Influence of hormones and hormone metabolites on the growth of Schwann cells derived from embryonic stem cells and on tumor cell lines expressing variable levels of neurofibromin. Dev Dyn 2008; 237:513-24. [PMID: 18213578 DOI: 10.1002/dvdy.21430] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Loss of neurofibromin, the protein product of the tumor suppressor gene neurofibromatosis type 1 (NF1), is associated with neurofibromas, composed largely of Schwann cells. The number and size of neurofibromas in NF1 patients have been shown to increase during pregnancy. A mouse embryonic stem cell (mESC) model was used, in which mESCs with varying levels of neurofibromin were differentiated into Schwann-like cells. NF1 cell lines derived from a malignant and a benign human tumor were used to study proliferation in response to hormones. Estrogen and androgen receptors were not expressed or expressed at very low levels in the NF1+/+ cells, at low levels in NF1+/-cells, and robust levels in NF1-/-cells. A 17beta-estradiol (E2) metabolite, 2-methoxy estradiol (2ME2) is cytotoxic to the NF1-/- malignant tumor cell line, and inhibits proliferation in the other cell lines. 2ME2 or its derivatives could provide new treatment avenues for NF1 hormone-sensitive tumors at times of greatest hormonal influence.
Collapse
Affiliation(s)
- Therese M Roth
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Quaglino A, Schere-Levy C, Romorini L, Meiss RP, Kordon EC. Mouse mammary tumors display Stat3 activation dependent on leukemia inhibitory factor signaling. Breast Cancer Res 2008; 9:R69. [PMID: 17925034 PMCID: PMC2242666 DOI: 10.1186/bcr1777] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Revised: 09/07/2007] [Accepted: 10/10/2007] [Indexed: 11/24/2022] Open
Abstract
Introduction It has been demonstrated that leukemia inhibitory factor (LIF) induces epithelium apoptosis through Stat3 activation during mouse mammary gland involution. In contrast, it has been shown that this transcription factor is commonly activated in breast cancer cells, although what causes this effect remains unknown. Here we have tested the hypothesis that locally produced LIF can be responsible for Stat3 activation in mouse mammary tumors. Methods The studies were performed in different tumorigenic and non-tumorigenic mammary cells. The expression of LIF and LIF receptor was tested by RT-PCR analysis. In tumors, LIF and Stat3 proteins were analyzed by immunohistochemistry, whereas Stat3 and extracellular signal-regulated kinase (ERK)1/2 expression and phosphorylation were studied by Western blot analysis. A LIF-specific blocking antibody was used to determine whether this cytokine was responsible for Stat3 phosphorylation induced by conditioned medium. Specific pharmacological inhibitors (PD98059 and Stat3ip) that affect ERK1/2 and Stat3 activation were used to study their involvement in LIF-induced effects. To analyze cell survival, assays with crystal violet were performed. Results High levels of LIF expression and activated Stat3 were found in mammary tumors growing in vivo and in their primary cultures. We found a single mouse mammary tumor cell line, LM3, that showed low levels of activated Stat3. Incidentally, these cells also showed very little expression of LIF receptor. This suggested that autocrine/paracrine LIF would be responsible for Stat3 activation in mouse mammary tumors. This hypothesis was confirmed by the ability of conditioned medium of mammary tumor primary cultures to induce Stat3 phosphorylation, activity that was prevented by pretreatment with LIF-blocking antibody. Besides, we found that LIF increased tumor cell viability. Interestingly, blocking Stat3 activation enhanced this effect in mammary tumor cells. Conclusion LIF is overexpressed in mouse mammary tumors, where it acts as the main Stat3 activator. Interestingly, the positive LIF effect on tumor cell viability is not dependent on Stat3 activation, which inhibits tumor cell survival as it does in normal mammary epithelium.
Collapse
Affiliation(s)
- Ana Quaglino
- IFIBYNE (CONICET), Facultad de Ciencias Exactas y Naturales, University of Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
12
|
Abstract
Patients with cancer can develop recurrent metastatic disease with latency periods that range from years even to decades. This pause can be explained by cancer dormancy, a stage in cancer progression in which residual disease is present but remains asymptomatic. Cancer dormancy is poorly understood, resulting in major shortcomings in our understanding of the full complexity of the disease. Here, I review experimental and clinical evidence that supports the existence of various mechanisms of cancer dormancy including angiogenic dormancy, cellular dormancy (G0-G1 arrest) and immunosurveillance. The advances in this field provide an emerging picture of how cancer dormancy can ensue and how it could be therapeutically targeted.
Collapse
Affiliation(s)
- Julio A Aguirre-Ghiso
- Department of Biomedical Sciences, School of Public Health and Center for Excellence in Cancer Genomics, University at Albany, State University of New York, One Discovery Drive, Rensselaer, New York 12144-3456, USA.
| |
Collapse
|
13
|
Kumar MJM, Ponvijay KS, Nandhini R, Nagarajan RS, Jose J, Srinivas G, Nagarajan P, Venkatesan R, Kumar K, Singh S. A mouse model for luminal epithelial like ER positive subtype of human breast cancer. BMC Cancer 2007; 7:180. [PMID: 17880731 PMCID: PMC2094712 DOI: 10.1186/1471-2407-7-180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 09/20/2007] [Indexed: 11/10/2022] Open
Abstract
Background Generation of novel spontaneous ER positive mammary tumor animal model from heterozygous NIH nude mice. Methods Using brother-sister mating with pedigree expansion system, we derived a colony of heterozygous breeding females showing ER-Positive tumors around the age of 6 months. Complete blood picture, differential leukocyte count, and serum levels of Estrogen, Alanine amino transferase (SGPT), Aspartate amino transferase (SGOT), total protein and albumin were estimated. Aspiration biopsies and microbiology were carried out. Gross pathology of the tumors and their metastatic potential were assessed. The tumors were excised and further characterized using histopathology, cytology, electron microscopy (EM), molecular markers and Mouse mammary Tumor Virus – Long Terminal Repeats (MMTV LTR) specific RT-PCR. Results The tumors originated from 2ndor 5thor both the mammary glands and were multi-nodulated with variable central necrosis accompanied with an accumulation of inflammatory exudate. Significant increases in estrogen, SGPT, SGOT and neutrophils levels were noticed. Histopathologically, invasive nodular masses of pleomorphic tubular neoplastic epithelial cells invaded fibro-vascular stroma, adjacent dermis and subcutaneous tissue. Metastatic spread through hematogenous and regional lymph nodes, into liver, lungs, spleen, heart and dermal lymphatics was observed. EM picture revealed no viral particles and MMTV-negativity was confirmed through MMTV LTR-specific RT-PCR. High expression of ER α, moderate to high expression of proliferating cell nuclear antigen (PCNA), moderate expression of vimentin and Cytokeratin 19 (K19) and low expression of p53 were observed in tumor sections, when compared with that of the normal mammary gland. Conclusion Since 75% of human breast cancer were classified ER-positive and as our model mimics (in most of the characteristics, such as histopathology, metastasis, high estrogen levels) the ER-positive luminal epithelial-like human breast cancer, this model will be an attractive tool to understand the biology of estrogen-dependant breast cancer in women. To our knowledge, this is the first report of a spontaneous mammary model displaying regional lymph node involvement with both hematogenous and lymphatic spread to liver, lung, heart, spleen and lymph nodes.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/metabolism
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/secondary
- Disease Models, Animal
- Estrogens/metabolism
- Female
- Heart Neoplasms/metabolism
- Heart Neoplasms/secondary
- Heterozygote
- Humans
- Immunohistochemistry
- Inbreeding
- Keratin-19/metabolism
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphatic Metastasis
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Proliferating Cell Nuclear Antigen/metabolism
- Receptors, Estrogen/metabolism
- Splenic Neoplasms/metabolism
- Splenic Neoplasms/secondary
- Tumor Suppressor Protein p53/metabolism
- Vimentin/metabolism
Collapse
Affiliation(s)
- MJ Mahesh Kumar
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - KS Ponvijay
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - R Nandhini
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - RS Nagarajan
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - J Jose
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - G Srinivas
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - P Nagarajan
- National Institute of Immunology, New Delhi 110067, India
| | - R Venkatesan
- National Institute of Immunology, New Delhi 110067, India
| | - Kishor Kumar
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - S Singh
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| |
Collapse
|
14
|
Gattelli A, Zimberlin MN, Meiss RP, Castilla LH, Kordon EC. Selection of early-occurring mutations dictates hormone-independent progression in mouse mammary tumor lines. J Virol 2006; 80:11409-15. [PMID: 16971449 PMCID: PMC1642155 DOI: 10.1128/jvi.00234-06] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Mice harboring three mouse mammary tumor virus (MMTV) variants develop pregnancy-dependent (PD) tumors that progress to pregnancy-independent (PI) behavior through successive passages. Herein, we identified 10 predominant insertions in PI transplants from 8 independent tumor lines. These mutations were also detected in small cell populations in the early PD passages. In addition, we identified a new viral insertion upstream of the gene Rspo3, which is overexpressed in three of the eight independent tumor lines and codes for a protein very similar to the recently described protein encoded by Int7. This study suggests that during progression towards hormone independence, clonal expansion of cells with specific mutations might be more relevant than the occurrence of new MMTV insertions.
Collapse
Affiliation(s)
- Albana Gattelli
- IFIBYNE-CONICET, Facultad de Ciencias Exactas y Naturales, UBA Ciudad Universitaria, Pabellón 2, 2do piso, (1428) Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
15
|
Epstein RJ. Maintenance Therapy to Suppress Micrometastasis: The New Challenge for Adjuvant Cancer Treatment. Clin Cancer Res 2005; 11:5337-41. [PMID: 16061845 DOI: 10.1158/1078-0432.ccr-05-0437] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The palliative efficacy of cytotoxic drugs is routinely assessed using tumor shrinkage (response) rates shown in clinical trials. Although adjuvant drug therapy has a goal distinct from that of palliative therapy (i.e., to prolong survival by inhibiting progression of micrometastatic disease), it is widely assumed that the adjuvant efficacy of a drug will parallel its response rate ("activity") in advanced stages of the disease. Reconsideration of this assumption seems timely in view of recent developments: the realization that many predictors of short-term tumor response correlate inversely with long-term survival outcomes; the characterization of tumor progression as a discontinuous process that may include dormant phases; the understanding that micrometastasis is therapeutically suppressible by a variety of mechanisms including direct tumor cell kill, cytotoxic disruption of paracrine growth signals from normal tissues, and targeted inhibition of prometastatic pathways; the recognition that tumor dormancy not only blocks the antimetastatic efficacy of cytotoxic drugs but also represents a therapeutic end point for metastasis-suppressive noncytotoxic drugs such as hormone inhibitors; and the insight that optimal adjuvant drug therapy is likely to include both induction and maintenance components. The traditional view of cytoreductive response as a prerequisite for adjuvant drug efficacy thus merits reappraisal, with a view to accelerating incorporation of novel noncytotoxic maintenance therapies into controlled studies.
Collapse
Affiliation(s)
- Richard J Epstein
- Department of Medicine, University of Hong Kong, Professorial Block, Queen Mary Hospital, Pokfulam, Hong Kong.
| |
Collapse
|
16
|
Zhang X, Podsypanina K, Huang S, Mohsin SK, Chamness GC, Hatsell S, Cowin P, Schiff R, Li Y. Estrogen receptor positivity in mammary tumors of Wnt-1 transgenic mice is influenced by collaborating oncogenic mutations. Oncogene 2005; 24:4220-31. [PMID: 15824740 DOI: 10.1038/sj.onc.1208597] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The majority (75%) of human breast cancers express estrogen receptor (ER). Although ER-positive tumors usually respond to antiestrogen therapies, 30% of them do not. It is not known what controls the ER status of breast cancers or their responsiveness to antihormone interventions. In this report, we document that transgenic (TG) expression of Wnt-1 in mice induces ER-positive tumors. Loss of Pten or gain of Ras mutations during the evolution of tumors in Wnt-1 TG mice has no effect on the expression of ER, but overexpression of Neu or loss of p53 leads to ER-negative tumors. Thus, our results provide compelling evidence that expression of ER in breast cancer may be influenced by specific genetic changes that promote cancer progression. These findings constitute a first step to explore the molecular mechanisms leading to ER-positive or ER-negative mammary tumors. In addition, we find that ER-positive tumors arising in Wnt-1 TG mice are refractory to both ovariectomy and the ER antagonist tamoxifen, but lose ER expression with tamoxifen, suggesting that antiestrogen selects for ER-negative tumor cells and that the ER-positive cell fraction is dispensable for growth of these tumors. This is a first report of a mouse model of antiestrogen-resistant ER-positive breast cancers, and could provide a powerful tool to study the molecular mechanisms that control antiestrogen resistance.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Breast Neoplasms/genetics
- Disease Models, Animal
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Genes, p53
- Humans
- Immunohistochemistry
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/pharmacology
- Mammary Neoplasms, Animal/genetics
- Mice
- Mice, Transgenic
- Mitogens
- Ovariectomy/veterinary
- PTEN Phosphohydrolase
- Phosphoric Monoester Hydrolases/genetics
- Protein-Tyrosine Kinases
- Receptor, ErbB-2/genetics
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/physiology
- Signal Transduction
- Tamoxifen/pharmacology
- Tumor Suppressor Proteins/genetics
- Wnt Proteins
- Wnt1 Protein
Collapse
Affiliation(s)
- Xiaomei Zhang
- Breast Center, Baylor College of Medicine, One Baylor Plaza, N1210.03, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sun HC, Tang ZY, Ma ZC, Qin LX, Wang L, Ye QH, Fan J, Wu ZQ, Zhou XD. The prognostic factor for outcome following second resection for intrahepatic recurrence of hepatocellular carcinoma with a hepatitis B virus infection background. J Cancer Res Clin Oncol 2005; 131:284-8. [PMID: 15662524 DOI: 10.1007/s00432-004-0645-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2004] [Accepted: 10/25/2004] [Indexed: 12/28/2022]
Abstract
PURPOSE Second resection has been proved to be a safe and effective treatment for patients with intrahepatic recurrent HCC after primary resection; however, preoperative prognostic factors for outcome following second resection in patients with a hepatitis B virus (HBV) infection background remains to be clarified. METHODS Fifty-seven patients with intrahepatic recurrent an HCC and HBV infection background received second resection from 1997 to 2003 in our institute. All of them were negative for anti-hepatitis C virus (HCV) and positive regarding HBV profile. Patient and tumor factors were analyzed. RESULTS At the time of preparing this paper, 31 had re-recurrence and 21 patients had died. No postoperative mortality was noted. The 1-, 3-, and 5-year overall survival after second resection were 69.9%, 61.2%, and 30.6%, respectively. Univariate and multivariate analysis showed that vascular invasion and time to recurrence were the independent prognostic factors for overall survival following second resection. The 3- and 4-year overall survival after second resection were 57.7% and 46.6% in patients with the presence of any of two risk factors (n = 46), and 100% and 100% in those with absence of both risk factors (n = 11, P = 0.008). CONCLUSIONS Vascular invasion and time to recurrence were the prognostic factors for overall survival following second resection of intrahepatic recurrent HCC.
Collapse
Affiliation(s)
- Hui-Chuan Sun
- Liver Cancer Institute and Zhong Shan Hospital, Fudan University, 200032 Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|