81 results on '"Oktay MH"'
Search Results
2. Abstract P2-08-18: Tumor microenvironment of metastasis (TMEM) score in residual breast carcinoma post-neoadjuvant chemotherapy as an independent prognosticator of distant recurrence
- Author
-
Oktay, MH, primary, D'Alfonso, T, additional, Ginter, P, additional, Lanjewar, S, additional, Entenberg, D, additional, Pastoriza, JM, additional, Wang, Y, additional, Lin, Y, additional, Karagiannnis, GS, additional, Lin, J, additional, Ye, X, additional, Anampa, J, additional, Xue, X, additional, Rohan, TE, additional, Sparano, JA, additional, and Condeelis, JS, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P6-10-02: Black race is associated with worse distant relapse-free survival in breast cancer patients treated with neoadjuvant compared to adjuvant systemic chemotherapy
- Author
-
Pastoriza, JM, primary, Karagiannis, GS, additional, Xue, X, additional, Lin, J, additional, Condeelis, JS, additional, Sparanno, JA, additional, Rohan, TE, additional, and Oktay, MH, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract P2-05-06: Analytical and clinical validation of a fully automated tissue-based quantitative assay (MetaSite Breast™) to detect the likelihood of distant metastasis in hormone receptor (HR)-positive, HER2-negative early stage breast cancer (ESBC)
- Author
-
Donovan, MJ, primary, Jones, JG, additional, Entenberg, DR, additional, Condeelis, JS, additional, D'alfonso, TM, additional, Gustavson, M, additional, Molinaro, A, additional, Oktay, MH, additional, Xue, X, additional, Sparano, JA, additional, Peterson, MA, additional, Podznyakova, O, additional, Rohan, TE, additional, Shuber, AP, additional, Gertler, FB, additional, Ly, A, additional, Divelbiss, ME, additional, and Hamilton, DA, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract PD5-02: Paclitaxel induced mena- and TMEM-mediated pro-metastatic changes in the breast cancer microenvironment
- Author
-
Karagiannis, GS, primary, Pastoriza, JM, additional, Wang, Y, additional, Harney, AS, additional, Entenberg, D, additional, Pignatelli, J, additional, Jones, JG, additional, Anampa, J, additional, Sparano, JA, additional, Rohan, TE, additional, Condeelis, JS, additional, and Oktay, MH, additional
- Published
- 2017
- Full Text
- View/download PDF
6. Abstract S4-04: Tumor microenvironment of metastasis (TMEM) score is associated with early distant recurrence in hormone receptor (HR) positive, HER2-negative early stage breast cancer (ESBC)
- Author
-
Sparano, JA, primary, Gray, R, additional, Oktay, MH, additional, Entenberg, D, additional, Rohan, T, additional, Xue, X, additional, Donovan, M, additional, Peterson, M, additional, Shuber, A, additional, Hamilton, D, additional, D'Alfonso, T, additional, Goldstein, LJ, additional, Gerlter, F, additional, Davidson, N, additional, Condeelis, J, additional, and Jones, J, additional
- Published
- 2017
- Full Text
- View/download PDF
7. Abstract P2-11-03: A tumor microenvironment of metastasis (TMEM) biomarker in early breast cancer provides prognostic information that is complementary to IHC4: A prospective validation study
- Author
-
Sparano, JA, primary, Rohan, TE, additional, Xue, X, additional, Lin, H-M, additional, D'Alfonso, TM, additional, Ginter, PS, additional, Oktay, MH, additional, Robinson, BD, additional, Ginsberg, M, additional, Gertler, FB, additional, Glass, AG, additional, Condeelis, JS, additional, and Jones, JG, additional
- Published
- 2013
- Full Text
- View/download PDF
8. Abstract P6-02-04: TMEM (Tumor MicroEnvironment of Metastasis) in human breast cancer is a blood vessel associated intravasation microenvironment unrelated to lymphatics
- Author
-
Ginter, PS, primary, Robinson, BD, additional, D'Alfonso, TM, additional, Oktay, MH, additional, Gertler, FB, additional, Rohan, TE, additional, Condeelis, JS, additional, and Jones, JG, additional
- Published
- 2012
- Full Text
- View/download PDF
9. Phase Ib clinical and pharmacodynamic study of the TIE2 kinase inhibitor rebastinib with paclitaxel or eribulin in HER2-negative metastatic breast cancer.
- Author
-
Anampa JD, Flynn DL, Leary C, Oh S, Xue X, Oktay MH, Condeelis JS, and Sparano JA
- Abstract
Purpose: Breast cancer cells disseminate to distant sites via Tumor Microenvironment of Metastasis (TMEM) doorways. The TIE2 inhibitor rebastinib blocks TMEM doorway function in the PyMT mouse model of breast cancer. We aimed to assess the safety and pharmacodynamics of rebastinib plus paclitaxel or eribulin in patients with HER2-negative metastatic breast cancer (MBC)., Patients and Methods: This phase Ib trial enrolled 27 patients with MBC who received 50 mg or 100 mg of rebastinib PO BID in combination with weekly paclitaxel 80 mg/m2 (if ≤ 2 prior non-taxane regimens) or eribulin 1.4 mg/m2 on days 1 & 8 (if ≥ 1 prior regimen). Safety, tolerability and pharmacodynamic parameters indicating TIE2 kinase inhibition and TMEM doorway function were evaluated., Results: No dose-limiting toxicities in cycle 1 or 2 were observed among the first 12 patients at either rebastinib dose level. The most common treatment-emergent adverse events (AEs) were anemia (85%), fatigue (78%), anorexia (67%), leukopenia (67%), increased alanine aminotransferase (59%), hyperglycemia (56%), nausea (52%), and neutropenia (52%). AEs attributed to rebastinib include muscular weakness and myalgias. Intraocular pressure increased at the 100 mg rebastinib dose level, whereas angiopoietin-2 levels increased at both dose levels, providing pharmacodynamic evidence for TIE2 blockade. Circulating tumor cells decreased significantly with the combined treatment. Objective response occurred in 5/23 (22%) evaluable patients., Conclusions: In patients with MBC, the recommended phase 2 dose of rebastinib associated with pharmacodynamic evidence of TIE2 inhibition is either 50 or 100 mg PO BID in combination with paclitaxel or eribulin.
- Published
- 2024
- Full Text
- View/download PDF
10. Macrophages in tumor cell migration and metastasis.
- Author
-
Friedman-DeLuca M, Karagiannis GS, Condeelis JS, Oktay MH, and Entenberg D
- Subjects
- Humans, Animals, Epithelial-Mesenchymal Transition, Macrophages immunology, Tumor Microenvironment, Neoplasms pathology, Neoplasms etiology, Neoplasms immunology, Cell Movement, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Neoplasm Metastasis
- Abstract
Tumor-associated macrophages (TAMs) are a phenotypically diverse, highly plastic population of cells in the tumor microenvironment (TME) that have long been known to promote cancer progression. In this review, we summarize TAM ontogeny and polarization, and then explore how TAMs enhance tumor cell migration through the TME, thus facilitating metastasis. We also discuss how chemotherapy and host factors including diet, obesity, and race, impact TAM phenotype and cancer progression. In brief, TAMs induce epithelial-mesenchymal transition (EMT) in tumor cells, giving them a migratory phenotype. They promote extracellular matrix (ECM) remodeling, allowing tumor cells to migrate more easily. TAMs also provide chemotactic signals that promote tumor cell directional migration towards blood vessels, and then participate in the signaling cascade at the blood vessel that allows tumor cells to intravasate and disseminate throughout the body. Furthermore, while chemotherapy can repolarize TAMs to induce an anti-tumor response, these cytotoxic drugs can also lead to macrophage-mediated tumor relapse and metastasis. Patient response to chemotherapy may be dependent on patient-specific factors such as diet, obesity, and race, as these factors have been shown to alter macrophage phenotype and affect cancer-related outcomes. More research on how chemotherapy and patient-specific factors impact TAMs and cancer progression is needed to refine treatment strategies for cancer patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Friedman-DeLuca, Karagiannis, Condeelis, Oktay and Entenberg.)
- Published
- 2024
- Full Text
- View/download PDF
11. Intratumoral CXCL12 Gradients Contextualize Tumor Cell Invasion, Migration and Immune Suppression in Breast Cancer.
- Author
-
Anastasiadou DP, Couturier N, Goel S, Argyris DG, Vodopyanov S, Rivera-Sanchez L, Gonzalez E, Kreger J, Griffen A, Kazakov A, Burt J, Recoder N, Duran CL, Harney AS, Quesnel A, Filippou PS, Lenis VP, Shukla S, Entenberg D, Zintiridou A, Chen X, Eddy RJ, Oktay MH, Condeelis JS, Karagiannis NS, Briceno A, Guzik H, Alon R, DesMarais V, Ioannou G, Gnjatic S, Raynolds DM, Macedo R, Reshef R, Gil-Henn H, MacLean AL, Torres ER, LaFave LM, Lauvau G, and Karagiannis GS
- Abstract
Although the CXCL12/CXCR4 pathway has been prior investigated for its prometastatic and immuno- suppressive roles in the tumor microenvironment, evidence on the spatiotemporal regulation of these hallmarks has been lacking. Here, we demonstrate that CXCL12 forms a gradient specifically around cancer cell intravasation doorways, also known as Tumor Microenvironment of Metastasis (TMEM) doorways, thus facilitating the chemotactic translocation of prometastatic tumor cells expressing CXCR4 toward the perivascular TMEM doorways for subsequent entry into peripheral circulation. Fur- thermore, we demonstrate that the CXCL12-rich micro-environment around TMEM doorways may cre- ate immunosuppressive niches, whereby CD8
+ T cells, despite being attracted to these regions, often exhibit reduced effector functions, limiting their efficacy. While the CXCL12/CXCR4 pathway can mini- mally influence the overall composition of immune cell populations, it biases the distribution of CD8+ T cells away from TMEM doorways, justifying its prior-established role as immunosuppressive factor for CD8+ T cells. Our research suggests that the complex interactions between CXCL12 and the various tumor and immune cell types contributes not only to the completion of the initial steps of the metastatic cascade, but also offers an immunological "sanctuary" to prometastatic tumor cells homed around TMEM doorways. Overall, our study enhances our current understanding on the mechanisms, via which CXCL12 orchestrates tumor cell behavior and immune dynamics, potentially guiding future thera- peutic strategies to combat breast cancer metastasis and improve anti-tumor immunity.- Published
- 2024
- Full Text
- View/download PDF
12. Signaling events at TMEM doorways provide potential targets for inhibiting breast cancer dissemination.
- Author
-
Surve CR, Duran CL, Ye X, Chen X, Lin Y, Harney AS, Wang Y, Sharma VP, Stanley ER, Cox D, McAuliffe JC, Entenberg D, Oktay MH, and Condeelis JS
- Abstract
Tumor cell intravasation is essential for metastatic dissemination, but its exact mechanism is incompletely understood. We have previously shown that in breast cancer, the direct and stable association of a tumor cell expressing Mena, a Tie2
hi /VEGFhi macrophage, and a vascular endothelial cell, creates an intravasation portal, called a "tumor microenvironment of metastasis" (TMEM) doorway, for tumor cell intravasation, leading to dissemination to distant sites. The density of TMEM doorways, also called TMEM doorway score, is a clinically validated prognostic marker of distant metastasis in breast cancer patients. Although we know that tumor cells utilize TMEM doorway-associated transient vascular openings to intravasate, the precise signaling mechanisms involved in TMEM doorway function are only partially understood. Using two mouse models of breast cancer and an in vitro assay of intravasation, we report that CSF-1 secreted by the TMEM doorway tumor cell stimulates local secretion of VEGF-A from the Tie2hi TMEM doorway macrophage, leading to the dissociation of endothelial junctions between TMEM doorway associated endothelial cells, supporting tumor cell intravasation. Acute blockade of CSF-1R signaling decreases macrophage VEGF-A secretion as well as TMEM doorway-associated vascular opening, tumor cell trans-endothelial migration, and dissemination. These new insights into signaling events regulating TMEM doorway function should be explored further as treatment strategies for metastatic disease., Competing Interests: Competing interests: The authors declare no competing financial interests.- Published
- 2024
- Full Text
- View/download PDF
13. SKP2 Knockout in Rb1/p53-Deficient Mouse Models of Osteosarcoma Induces Immune Infiltration and Drives a Transcriptional Program with a Favorable Prognosis.
- Author
-
Ferrena A, Wang J, Zhang R, Karadal-Ferrena B, Al-Hardan W, Singh S, Borjihan H, Schwartz EL, Zhao H, Oktay MH, Yang R, Geller DS, Hoang BH, and Zheng D
- Subjects
- Animals, Humans, Mice, Disease Models, Animal, Mice, Knockout, Mice, Transgenic, Prognosis, S-Phase Kinase-Associated Proteins genetics, S-Phase Kinase-Associated Proteins metabolism, Tumor Microenvironment genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Bone Neoplasms genetics, Osteosarcoma genetics, Osteosarcoma pathology
- Abstract
Osteosarcoma is an aggressive bone malignancy with a poor prognosis. One putative proto-oncogene in osteosarcoma is SKP2, encoding a substrate recognition factor of the SCF E3 ubiquitin ligase. We previously demonstrated that Skp2 knockout in murine osteosarcoma improved survival and delayed tumorigenesis. Here, we performed RNA sequencing (RNA-seq) on tumors from a transgenic osteosarcoma mouse model with conditional Trp53 and Rb1 knockouts in the osteoblast lineage ("DKO": Osx1-Cre;Rb1lox/lox;p53lox/lox) and a triple-knockout model with additional Skp2 germline knockout ("TKO": Osx1-Cre;Rb1lox/lox;p53lox/lox;Skp2-/-), followed by qPCR and immunohistochemistry validation. To investigate the clinical implications of our results, we analyzed a human osteosarcoma patient cohort ("NCI-TARGET OS") with RNA-seq and clinical data. We found large differences in gene expression after SKP2 knockout. Surprisingly, we observed increased expression of genes related to immune microenvironment infiltration in TKO tumors, especially the signature genes for macrophages and to a lesser extent, T cells, B cells, and vascular cells. We also uncovered a set of relevant transcription factors that may mediate these changes. In osteosarcoma patient cohorts, high expression of genes upregulated in TKO was correlated with favorable overall survival, which was largely explained by the macrophage gene signatures. This relationship was further supported by our finding that SKP2 expression was negatively correlated with macrophage infiltration in the NCI-TARGET osteosarcoma and the TCGA Sarcoma cohorts. Overall, our findings indicate that SKP2 may mediate immune exclusion from the osteosarcoma tumor microenvironment, suggesting that SKP2 modulation in osteosarcoma may induce antitumor immune activation., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
14. A SOX9-B7x axis safeguards dedifferentiated tumor cells from immune surveillance to drive breast cancer progression.
- Author
-
Liu Y, John P, Nishitani K, Cui J, Nishimura CD, Christin JR, Couturier N, Ren X, Wei Y, Pulanco MC, Galbo PM Jr, Zhang X, Fu W, Cui W, Bartholdy BA, Zheng D, Lauvau G, Fineberg SA, Oktay MH, Zang X, and Guo W
- Subjects
- Animals, Female, Humans, Mice, CD8-Positive T-Lymphocytes, Immunosuppression Therapy, SOX9 Transcription Factor, V-Set Domain-Containing T-Cell Activation Inhibitor 1 metabolism, Breast Neoplasms
- Abstract
How dedifferentiated stem-like tumor cells evade immunosurveillance remains poorly understood. We show that the lineage-plasticity regulator SOX9, which is upregulated in dedifferentiated tumor cells, limits the number of infiltrating T lymphocytes in premalignant lesions of mouse basal-like breast cancer. SOX9-mediated immunosuppression is required for the progression of in situ tumors to invasive carcinoma. SOX9 induces the expression of immune checkpoint B7x/B7-H4 through STAT3 activation and direct transcriptional regulation. B7x is upregulated in dedifferentiated tumor cells and protects them from immunosurveillance. B7x also protects mammary gland regeneration in immunocompetent mice. In advanced tumors, B7x targeting inhibits tumor growth and overcomes resistance to anti-PD-L1 immunotherapy. In human breast cancer, SOX9 and B7x expression are correlated and associated with reduced CD8
+ T cell infiltration. This study, using mouse models, cell lines, and patient samples, identifies a dedifferentiation-associated immunosuppression mechanism and demonstrates the therapeutic potential of targeting the SOX9-B7x pathway in basal-like breast cancer., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. FAK family proteins regulate in vivo breast cancer metastasis via distinct mechanisms.
- Author
-
Genna A, Alter J, Poletti M, Meirson T, Sneh T, Gendler M, Saleev N, Karagiannis GS, Wang Y, Cox D, Entenberg D, Oktay MH, Korcsmaros T, Condeelis JS, and Gil-Henn H
- Abstract
Breast cancer is the most commonly diagnosed malignancy and the major leading cause of tumor-related deaths in women. It is estimated that the majority of breast tumor-related deaths are a consequence of metastasis, to which no cure exists at present. The FAK family proteins Proline-rich tyrosine kinase (PYK2) and focal adhesion kinase (FAK) are highly expressed in breast cancer, but the exact cellular and signaling mechanisms by which they regulate in vivo tumor cell invasiveness and consequent metastatic dissemination are mostly unknown. Using a PYK2 and FAK knockdown xenograft model we show here, for the first time, that ablation of either PYK2 or FAK decreases primary tumor size and significantly reduces Tumor MicroEnvironment of Metastasis (TMEM) doorway activation, leading to decreased intravasation and reduced spontaneous lung metastasis. Intravital imaging analysis further demonstrates that PYK2, but not FAK, regulates a motility phenotype switch between focal adhesion-mediated fast motility and invadopodia-dependent, ECM-degradation associated slow motility within the primary tumor. Furthermore, we validate our in vivo and intravital imaging results with integrated transcriptomic and proteomic data analysis from xenograft knockdown tumors and reveal new and distinct pathways by which these two homologous kinases regulate breast tumor cell invasiveness and consequent metastatic dissemination. Our findings identify PYK2 and FAK as novel mediators of mammary tumor progression and metastasis and as candidate therapeutic targets for breast cancer metastasis.
- Published
- 2023
- Full Text
- View/download PDF
16. Stabilized Window for Intravital Imaging of the Murine Pancreas.
- Author
-
Petersen J, Du W, Adkisson C, Gravekamp C, Oktay MH, Condeelis J, Panarelli NC, McAuliffe JC, and Entenberg D
- Subjects
- Humans, Animals, Mice, Acute Disease, Pancreas diagnostic imaging, Pancreas pathology, Intravital Microscopy methods, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms pathology, Pancreatitis pathology, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology
- Abstract
The physiology and pathophysiology of the pancreas are complex. Diseases of the pancreas, such as pancreatitis and pancreatic adenocarcinoma (PDAC) have high morbidity and mortality. Intravital imaging (IVI) is a powerful technique enabling the high-resolution imaging of tissues in both healthy and diseased states, allowing for real-time observation of cell dynamics. IVI of the murine pancreas presents significant challenges due to the deep visceral and compliant nature of the organ, which make it highly prone to damage and motion artifacts. Described here is the process of implantation of the Stabilized Window for Intravital imaging of the murine Pancreas (SWIP). The SWIP allows IVI of the murine pancreas in normal healthy states, during the transformation from the healthy pancreas to acute pancreatitis induced by cerulein, and in malignant states such as pancreatic tumors. In conjunction with genetically labeled cells or the administration of fluorescent dyes, the SWIP enables the measurement of single-cell and subcellular dynamics (including single-cell and collective migration) as well as serial imaging of the same region of interest over multiple days. The ability to capture tumor cell migration is of particular importance as the primary cause of cancer-related mortality in PDAC is the overwhelming metastatic burden. Understanding the physiological dynamics of metastasis in PDAC is a critical unmet need and crucial for improving patient prognosis. Overall, the SWIP provides improved imaging stability and expands the application of IVI in the healthy pancreas and malignant pancreas diseases.
- Published
- 2023
- Full Text
- View/download PDF
17. Tracking Tumor Cell Dissemination from Lung Metastases Using Photoconversion.
- Author
-
Friedman-DeLuca M, Patel PP, Karadal-Ferrena B, Barth ND, Duran CL, Ye X, Papanicolaou M, Condeelis JS, Oktay MH, Borriello L, and Entenberg D
- Subjects
- Humans, Animals, Mice, Neoplasm Metastasis, Quality of Life, Lung Neoplasms pathology
- Abstract
Metastasis - the systemic spread of cancer - is the leading cause of cancer-related deaths. Although metastasis is commonly thought of as a unidirectional process wherein cells from the primary tumor disseminate and seed metastases, tumor cells in existing metastases can also redisseminate and give rise to new lesions in tertiary sites in a process known as "metastasis-from-metastases" or "metastasis-to-metastasis seeding." Metastasis-to-metastasis seeding may increase the metastatic burden and decrease the patient's quality of life and survival. Therefore, understanding the processes behind this phenomenon is crucial to refining treatment strategies for patients with metastatic cancer. Little is known about metastasis-to-metastasis seeding, due in part to logistical and technological limitations. Studies on metastasis-to-metastasis seeding rely primarily on sequencing methods, which may not be practical for researchers studying the exact timing of metastasis-to-metastasis seeding events or what promotes or prevents them. This highlights the lack of methodologies that facilitate the study of metastasis-to-metastasis seeding. To address this, we have developed - and describe herein - a murine surgical protocol for the selective photoconversion of lung metastases, allowing specific marking and fate tracking of tumor cells redisseminating from the lung to tertiary sites. To our knowledge, this is the only method for studying tumor cell redissemination and metastasis-to-metastasis seeding from the lungs that does not require genomic analysis.
- Published
- 2023
- Full Text
- View/download PDF
18. Racial disparity in tumor microenvironment and distant recurrence in residual breast cancer after neoadjuvant chemotherapy.
- Author
-
Kim G, Karadal-Ferrena B, Qin J, Sharma VP, Oktay IS, Lin Y, Ye X, Asiry S, Pastoriza JM, Cheng E, Ladak N, Condeelis JS, Adler E, Ginter PS, D'Alfonso T, Entenberg D, Xue X, Sparano JA, and Oktay MH
- Abstract
Black, compared to white, women with residual estrogen receptor-positive (ER+) breast cancer after neoadjuvant chemotherapy (NAC) have worse distant recurrence-free survival (DRFS). Such racial disparity may be due to difference in density of portals for systemic cancer cell dissemination, called TMEM doorways, and pro-metastatic tumor microenvironment (TME). Here, we evaluate residual cancer specimens after NAC from 96 Black and 87 white women. TMEM doorways are visualized by triple immunohistochemistry, and cancer stem cells by immunofluorescence for SOX9. The correlation between TMEM doorway score and pro-metastatic TME parameters with DRFS is examined using log-rank and multivariate Cox regression. Black, compared to white, patients are more likely to develop distant recurrence (49% vs 34.5%, p = 0.07), receive mastectomy (69.8% vs 54%, p = 0.04), and have higher grade tumors (p = 0.002). Tumors from Black patients have higher TMEM doorway and macrophages density overall (p = 0.002; p = 0.002, respectively) and in the ER+/HER2- (p = 0.02; p = 0.02, respectively), but not in the triple negative disease. Furthermore, high TMEM doorway score is associated with worse DRFS. TMEM doorway score is an independent prognostic factor in the entire study population (HR, 2.02; 95%CI, 1.18-3.46; p = 0.01), with a strong trend in ER+/HER2- disease (HR, 2.38; 95%CI, 0.96-5.95; p = 0.06). SOX9 expression is not associated with racial disparity in TME or outcome. In conclusion, higher TMEM doorway density in residual breast cancer after NAC is associated with higher distant recurrence risk, and Black patients are associated with higher TMEM doorway density, suggesting that TMEM doorway density may contribute to racial disparities in breast cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. Cooperative NF-κB and Notch1 signaling promotes macrophage-mediated MenaINV expression in breast cancer.
- Author
-
Duran CL, Karagiannis GS, Chen X, Sharma VP, Entenberg D, Condeelis JS, and Oktay MH
- Subjects
- Humans, Mice, Animals, Female, Cell Line, Tumor, Signal Transduction, Macrophages metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, NF-kappa B genetics, NF-kappa B metabolism, Breast Neoplasms pathology
- Abstract
Metastasis is a multistep process that leads to the formation of clinically detectable tumor foci at distant organs and frequently to patient demise. Only a subpopulation of breast cancer cells within the primary tumor can disseminate systemically and cause metastasis. To disseminate, cancer cells must express MenaINV, an isoform of the actin regulatory protein Mena, encoded by the ENAH gene, that endows tumor cells with transendothelial migration activity, allowing them to enter and exit the blood circulation. We have previously demonstrated that MenaINV mRNA and protein expression is induced in cancer cells by macrophage contact. In this study, we discovered the precise mechanism by which macrophages induce MenaINV expression in tumor cells. We examined the promoter of the human and mouse ENAH gene and discovered a conserved NF-κB transcription factor binding site. Using live imaging of an NF-κB activity reporter and staining of fixed tissues from mouse and human breast cancer, we further determined that for maximal induction of MenaINV in cancer cells, NF-κB needs to cooperate with the Notch1 signaling pathway. Mechanistically, Notch1 signaling does not directly increase MenaINV expression, but it enhances and sustains NF-κB signaling through retention of p65, an NF-κB transcription factor, in the nucleus of tumor cells, leading to increased MenaINV expression. In mice, these signals are augmented following chemotherapy treatment and abrogated upon macrophage depletion. Targeting Notch1 signaling in vivo decreased NF-κB signaling activation and MenaINV expression in the primary tumor and decreased metastasis. Altogether, these data uncover mechanistic targets for blocking MenaINV induction that should be explored clinically to decrease cancer cell dissemination and improve survival of patients with metastatic disease., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. Intravital imaging to study cancer progression and metastasis.
- Author
-
Entenberg D, Oktay MH, and Condeelis JS
- Subjects
- Animals, Humans, Intravital Microscopy methods, Tumor Microenvironment, Mammals, Neoplasms diagnostic imaging, Neoplasms pathology
- Abstract
Navigation through the bulk tumour, entry into the blood vasculature, survival in the circulation, exit at distant sites and resumption of proliferation are all steps necessary for tumour cells to successfully metastasize. The ability of tumour cells to complete these steps is highly dependent on the timing and sequence of the interactions that these cells have with the tumour microenvironment (TME), including stromal cells, the extracellular matrix and soluble factors. The TME thus plays a major role in determining the overall metastatic phenotype of tumours. The complexity and cause-and-effect dynamics of the TME cannot currently be recapitulated in vitro or inferred from studies of fixed tissue, and are best studied in vivo, in real time and at single-cell resolution. Intravital imaging (IVI) offers these capabilities, and recent years have been a time of immense growth and innovation in the field. Here we review some of the recent advances in IVI of mammalian models of cancer and describe how IVI is being used to understand cancer progression and metastasis, and to develop novel treatments and therapies. We describe new techniques that allow access to a range of tissue and cancer types, novel fluorescent reporters and biosensors that allow fate mapping and the probing of functional and phenotypic states, and the clinical applications that have arisen from applying these techniques, reporters and biosensors to study cancer. We finish by presenting some of the challenges that remain in the field, how to address them and future perspectives., (© 2022. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
21. Assessment of MRI to estimate metastatic dissemination risk and prometastatic effects of chemotherapy.
- Author
-
Karagiannis GS, Bianchi A, Sanchez LR, Ambadipudi K, Cui MH, Anampa JM, Asiry S, Wang Y, Harney AS, Pastoriza JM, Lin Y, Chen X, Jones JG, Entenberg D, Haddad D, Hodges LJ, Duong TQ, Sparano JA, Oktay MH, Branch CA, and Condeelis JS
- Abstract
Metastatic dissemination in breast cancer is regulated by specialized intravasation sites called "tumor microenvironment of metastasis" (TMEM) doorways, composed of a tumor cell expressing the actin-regulatory protein Mena, a perivascular macrophage, and an endothelial cell, all in stable physical contact. High TMEM doorway number is associated with an increased risk of distant metastasis in human breast cancer and mouse models of breast carcinoma. Here, we developed a novel magnetic resonance imaging (MRI) methodology, called TMEM Activity-MRI, to detect TMEM-associated vascular openings that serve as the portal of entry for cancer cell intravasation and metastatic dissemination. We demonstrate that TMEM Activity-MRI correlates with primary tumor TMEM doorway counts in both breast cancer patients and mouse models, including MMTV-PyMT and patient-derived xenograft models. In addition, TMEM Activity-MRI is reduced in mouse models upon treatment with rebastinib, a specific and potent TMEM doorway inhibitor. TMEM Activity-MRI is an assay that specifically measures TMEM-associated vascular opening (TAVO) events in the tumor microenvironment, and as such, can be utilized in mechanistic studies investigating molecular pathways of cancer cell dissemination and metastasis. Finally, we demonstrate that TMEM Activity-MRI increases upon treatment with paclitaxel in mouse models, consistent with prior observations that chemotherapy enhances TMEM doorway assembly and activity in human breast cancer. Our findings suggest that TMEM Activity-MRI is a promising precision medicine tool for localized breast cancer that could be used as a non-invasive test to determine metastatic risk and serve as an intermediate pharmacodynamic biomarker to monitor therapeutic response to agents that block TMEM doorway-mediated dissemination., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Racial disparity in distant recurrence-free survival in patients with localized breast cancer: A pooled analysis of National Surgical Adjuvant Breast and Bowel Project trials.
- Author
-
Kim G, Pastoriza JM, Qin J, Lin J, Karagiannis GS, Condeelis JS, Yothers G, Anderson S, Julian T, Entenberg D, Rohan TE, Xue X, Sparano JA, and Oktay MH
- Subjects
- Chemotherapy, Adjuvant, Female, Humans, Neoadjuvant Therapy, Neoplasm Recurrence, Local drug therapy, Receptors, Estrogen analysis, Breast Neoplasms drug therapy, Breast Neoplasms surgery
- Abstract
Background: Black race is associated with worse outcome in patients with breast cancer. The distant relapse-free survival (DRFS) between Black and White women with localized breast cancer who participated in National Cancer Institute-sponsored clinical trial was evaluated., Methods: Pooled data were analyzed from 8 National Surgical Adjuvant Breast and Bowel Project (NSABP) trials including 9702 women with localized breast cancer treated with adjuvant chemotherapy (AC, n = 7485) or neoadjuvant chemotherapy (NAC, n = 2217), who self-reported as Black (n = 1070) or White (n = 8632) race. The association between race and DRFS was analyzed using log-rank tests and multivariate Cox regression., Results: After adjustment for covariates including age, tumor size, nodal status, body mass index and taxane use, and treatment (AC vs NAC), Black race was associated with an inferior DRFS in estrogen receptor-positive (ER+; hazard ratio [HR], 1.24; 95% CI, 1.05-1.46; P = .01), but not in ER- disease (HR, 0.97; 95% CI, 0.83-1.14; P = .73), and significant interaction between race and ER status was observed (P = .03). There was no racial disparity in DRFS among patients with pathologic complete response (pCR) (log-rank P = .8). For patients without pCR, Black race was associated with worse DRFS in ER+ (HR, 1.67; 95% CI, 1.14-2.45; P = .01), but not in ER- disease (HR, 0.91; 95% CI, 0.65-1.28; P = .59)., Conclusions: Black race was associated with significantly inferior DRFS in ER+ localized breast cancer treated with AC or NAC, but not in ER- disease. In the NAC group, racial disparity was also observed in patients with residual ER+ breast cancer at surgery, but not in those who had pCR., Lay Summary: Black women with breast cancer have worse outcomes compared with White women. We investigated if this held true in the context of clinical trials that provide controlled treatment setting. Black women with cancer expressing estrogen receptors (ERs) had worse outcome than White women. If breast cancers did not express ERs, there was no racial disparity in outcome. We also observed racial disparity in women who received chemotherapy before their cancer was removed, but only if they had cancer expressing ERs and residual disease on completion of treatment. If the cancer disappeared with presurgical chemotherapy, there was no racial disparity., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
23. SWIP-a stabilized window for intravital imaging of the murine pancreas.
- Author
-
Du W, Adkisson C, Ye X, Duran CL, Chellakkan Selvanesan B, Gravekamp C, Oktay MH, McAuliffe JC, Condeelis JS, Panarelli NC, Norgard RJ, Sela Y, Stanger BZ, and Entenberg D
- Subjects
- Acute Disease, Animals, Intravital Microscopy, Mice, Pancreas diagnostic imaging, Pancreas pathology, Carcinoma, Pancreatic Ductal diagnostic imaging, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms pathology, Pancreatitis chemically induced, Pancreatitis diagnostic imaging, Pancreatitis pathology
- Abstract
Pancreatitis and pancreatic ductal adenocarcinoma (PDAC) are grave illnesses with high levels of morbidity and mortality. Intravital imaging (IVI) is a powerful technique for visualizing physiological processes in both health and disease. However, the application of IVI to the murine pancreas presents significant challenges, as it is a deep, compliant, visceral organ that is difficult to access, easily damaged and susceptible to motion artefacts. Existing imaging windows for stabilizing the pancreas during IVI have unfortunately shown poor stability for time-lapsed imaging on the minutes to hours scale, or are unable to accommodate both the healthy and tumour-bearing pancreata. To address these issues, we developed an improved stabilized window for intravital imaging of the pancreas (SWIP), which can be applied to not only the healthy pancreas but also to solid tumours like PDAC. Here, we validate the SWIP and use it to visualize a variety of processes for the first time, including (1) single-cell dynamics within the healthy pancreas, (2) transformation from healthy pancreas to acute pancreatitis induced by cerulein, and (3) the physiology of PDAC in both autochthonous and orthotopically injected models. SWIP can not only improve the imaging stability but also expand the application of IVI in both benign and malignant pancreas diseases.
- Published
- 2022
- Full Text
- View/download PDF
24. Combining TMEM Doorway Score and Mena Calc Score Improves the Prediction of Distant Recurrence Risk in HR+/HER2- Breast Cancer Patients.
- Author
-
Ye X, Oktay MH, Xue X, Rohan TE, Ginter PS, D'Alfonso T, Kornaga EN, Morris DG, Entenberg D, and Condeelis JS
- Abstract
Purpose: to develop several digital pathology-based machine vision algorithms for combining TMEM and Mena
Calc scores and determine if a combination of these biomarkers improves the ability to predict development of distant metastasis over and above that of either biomarker alone., Methods: This retrospective study included a subset of 130 patients (65 patients with no recurrence and 65 patients with a recurrence at 5 years) from the Calgary Tamoxifen cohort of breast cancer patients. Patients had confirmed invasive breast cancer and received adjuvant tamoxifen therapy. Of the 130 patients, 86 cases were suitable for analysis in this study. Sequential sections of formalin-fixed paraffin-embedded patient samples were stained for TMEM doorways (immunohistochemistry triple staining) and MenaCalc (immunofluorescence staining). Stained sections were imaged, aligned, and then scored for TMEM doorways and MenaCalc . Different ways of combining TMEM doorway and MenaCalc scores were evaluated and compared to identify the best performing combined marker by using the restricted mean survival time (RMST) difference method., Results: the best performing combined marker gave an RMST difference of 5.27 years (95% CI: 1.71-8.37), compared to 3.56 years (95% CI: 0.95-6.1) for the associated standalone TMEM doorway analysis and 2.94 years (95% CI: 0.25-5.87) for the associated standalone MenaCalc analysis., Conclusions: combining TMEM doorway and MenaCalc scores as a new biomarker improves prognostication over that observed with TMEM doorway or MenaCalc Score alone in this cohort of 86 patients.- Published
- 2022
- Full Text
- View/download PDF
25. Primary tumor associated macrophages activate programs of invasion and dormancy in disseminating tumor cells.
- Author
-
Borriello L, Coste A, Traub B, Sharma VP, Karagiannis GS, Lin Y, Wang Y, Ye X, Duran CL, Chen X, Friedman M, Sosa MS, Sun D, Dalla E, Singh DK, Oktay MH, Aguirre-Ghiso JA, Condeelis JS, and Entenberg D
- Subjects
- Animals, Breast Neoplasms genetics, Cell Line, Tumor, Female, Humans, Mice, Mice, Inbred C57BL, Neoplasms, Experimental, Neoplastic Stem Cells, Phenotype, Tumor Microenvironment physiology, Tumor-Associated Macrophages physiology
- Abstract
Metastases are initiated by disseminated tumor cells (DTCs) that colonize distant organs. Growing evidence suggests that the microenvironment of the primary tumor primes DTCs for dormant or proliferative fates. However, the manner in which this occurs remains poorly understood. Here, using the Window for High-Resolution Intravital Imaging of the Lung (WHRIL), we study the live lung longitudinally and follow the fate of individual DTCs that spontaneously disseminate from orthotopic breast tumors. We find that spontaneously DTCs have increased levels of retention, increased speed of extravasation, and greater survival after extravasation, compared to experimentally metastasized tumor cells. Detailed analysis reveals that a subset of macrophages within the primary tumor induces a pro-dissemination and pro-dormancy DTC phenotype. Our work provides insight into how specific primary tumor microenvironments prime a subpopulation of cells for expression of proteins associated with dissemination and dormancy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. MYC Levels Regulate Metastatic Heterogeneity in Pancreatic Adenocarcinoma.
- Author
-
Maddipati R, Norgard RJ, Baslan T, Rathi KS, Zhang A, Saeid A, Higashihara T, Wu F, Kumar A, Annamalai V, Bhattacharya S, Raman P, Adkisson CA, Pitarresi JR, Wengyn MD, Yamazoe T, Li J, Balli D, LaRiviere MJ, Ngo TC, Folkert IW, Millstein ID, Bermeo J, Carpenter EL, McAuliffe JC, Oktay MH, Brekken RA, Lowe SW, Iacobuzio-Donahue CA, Notta F, and Stanger BZ
- Subjects
- Adenocarcinoma secondary, Animals, Carcinoma, Pancreatic Ductal secondary, Disease Models, Animal, Humans, Mice, Pancreatic Neoplasms pathology, Adenocarcinoma genetics, Carcinoma, Pancreatic Ductal genetics, Gene Expression Regulation, Neoplastic, Genes, myc, Neoplasm Metastasis, Pancreatic Neoplasms genetics
- Abstract
The degree of metastatic disease varies widely among patients with cancer and affects clinical outcomes. However, the biological and functional differences that drive the extent of metastasis are poorly understood. We analyzed primary tumors and paired metastases using a multifluorescent lineage-labeled mouse model of pancreatic ductal adenocarcinoma (PDAC)-a tumor type in which most patients present with metastases. Genomic and transcriptomic analysis revealed an association between metastatic burden and gene amplification or transcriptional upregulation of MYC and its downstream targets. Functional experiments showed that MYC promotes metastasis by recruiting tumor-associated macrophages, leading to greater bloodstream intravasation. Consistent with these findings, metastatic progression in human PDAC was associated with activation of MYC signaling pathways and enrichment for MYC amplifications specifically in metastatic patients. Collectively, these results implicate MYC activity as a major determinant of metastatic burden in advanced PDAC. SIGNIFICANCE: Here, we investigate metastatic variation seen clinically in patients with PDAC and murine PDAC tumors and identify MYC as a major driver of this heterogeneity. This article is highlighted in the In This Issue feature, p. 275 ., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
27. Multiphoton intravital microscopy of rodents.
- Author
-
Scheele CLGJ, Herrmann D, Yamashita E, Celso CL, Jenne CN, Oktay MH, Entenberg D, Friedl P, Weigert R, Meijboom FLB, Ishii M, Timpson P, and van Rheenen J
- Abstract
Tissues are heterogeneous with respect to cellular and non-cellular components and in the dynamic interactions between these elements. To study the behaviour and fate of individual cells in these complex tissues, intravital microscopy (IVM) techniques such as multiphoton microscopy have been developed to visualize intact and live tissues at cellular and subcellular resolution. IVM experiments have revealed unique insights into the dynamic interplay between different cell types and their local environment, and how this drives morphogenesis and homeostasis of tissues, inflammation and immune responses, and the development of various diseases. This Primer introduces researchers to IVM technologies, with a focus on multiphoton microscopy of rodents, and discusses challenges, solutions and practical tips on how to perform IVM. To illustrate the unique potential of IVM, several examples of results are highlighted. Finally, we discuss data reproducibility and how to handle big imaging data sets., Competing Interests: Competing interests P.T. receives reagents from Kadmon, InxMed (also consultant), Redx Pharma, Équilibre Biopharmaceuticals and Amplia Therapeutics. Under a licensing agreement between Amplia Therapeutics and Garvan Institute of Medical Research, D.H. and P.T. (consultant) are entitled to milestone payments. All other authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
28. Live tumor imaging shows macrophage induction and TMEM-mediated enrichment of cancer stem cells during metastatic dissemination.
- Author
-
Sharma VP, Tang B, Wang Y, Duran CL, Karagiannis GS, Xue EA, Entenberg D, Borriello L, Coste A, Eddy RJ, Kim G, Ye X, Jones JG, Grunblatt E, Agi N, Roy S, Bandyopadhyaya G, Adler E, Surve CR, Esposito D, Goswami S, Segall JE, Guo W, Condeelis JS, Wakefield LM, and Oktay MH
- Subjects
- Animals, Breast Neoplasms diagnostic imaging, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Female, Humans, Intravital Microscopy, Mice, Mice, SCID, Neoplasm Metastasis, Neoplastic Cells, Circulating immunology, Neoplastic Stem Cells immunology, Receptors, Notch genetics, Receptors, Notch immunology, Signal Transduction, Tumor Microenvironment immunology, Breast Neoplasms immunology, Macrophages immunology, Neoplastic Stem Cells cytology
- Abstract
Cancer stem cells (CSCs) play an important role during metastasis, but the dynamic behavior and induction mechanisms of CSCs are not well understood. Here, we employ high-resolution intravital microscopy using a CSC biosensor to directly observe CSCs in live mice with mammary tumors. CSCs display the slow-migratory, invadopod-rich phenotype that is the hallmark of disseminating tumor cells. CSCs are enriched near macrophages, particularly near macrophage-containing intravasation sites called Tumor Microenvironment of Metastasis (TMEM) doorways. Substantial enrichment of CSCs occurs on association with TMEM doorways, contributing to the finding that CSCs represent >60% of circulating tumor cells. Mechanistically, stemness is induced in non-stem cancer cells upon their direct contact with macrophages via Notch-Jagged signaling. In breast cancers from patients, the density of TMEM doorways correlates with the proportion of cancer cells expressing stem cell markers, indicating that in human breast cancer TMEM doorways are not only cancer cell intravasation portals but also CSC programming sites., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
29. Targeting Tie2 in the Tumor Microenvironment: From Angiogenesis to Dissemination.
- Author
-
Duran CL, Borriello L, Karagiannis GS, Entenberg D, Oktay MH, and Condeelis JS
- Abstract
The Tie2 receptor tyrosine kinase is expressed in vascular endothelial cells, tumor-associated macrophages, and tumor cells and has been a major focus of research in therapies targeting the tumor microenvironment. The most extensively studied Tie2 ligands are Angiopoietin 1 and 2 (Ang1, Ang2). Ang1 plays a critical role in vessel maturation, endothelial cell migration, and survival. Ang2, depending on the context, may function to disrupt connections between the endothelial cells and perivascular cells, promoting vascular regression. However, in the presence of VEGF-A, Ang2 instead promotes angiogenesis. Tie2-expressing macrophages play a critical role in both tumor angiogenesis and the dissemination of tumor cells from the primary tumor to secondary sites. Therefore, Ang-Tie2 signaling functions as an angiogenic switch during tumor progression and metastasis. Here we review the recent advances and complexities of targeting Tie2 signaling in the tumor microenvironment as a possible anti-angiogenic, and anti-metastatic, therapy and describe its use in combination with chemotherapy.
- Published
- 2021
- Full Text
- View/download PDF
30. Molecular markers of risk of subsequent invasive breast cancer in women with ductal carcinoma in situ: protocol for a population-based cohort study.
- Author
-
Rohan TE, Ginsberg M, Wang Y, Couch FJ, Feigelson HS, Greenlee RT, Honda S, Stark A, Chitale D, Wang T, Xue X, Oktay MH, Sparano JA, and Loudig O
- Subjects
- Cohort Studies, Female, Humans, Biomarkers, Tumor, Breast Neoplasms epidemiology, Breast Neoplasms genetics, Carcinoma, Ductal, Breast, Carcinoma, Intraductal, Noninfiltrating epidemiology, Carcinoma, Intraductal, Noninfiltrating genetics, MicroRNAs
- Abstract
Introduction: Ductal carcinoma in situ (DCIS) of the breast is a non-obligate precursor of invasive breast cancer (IBC). Many DCIS patients are either undertreated or overtreated. The overarching goal of the study described here is to facilitate detection of patients with DCIS at risk of IBC development. Here, we propose to use risk factor data and formalin-fixed paraffin-embedded (FFPE) DCIS tissue from a large, ethnically diverse, population-based cohort of 8175 women with a first diagnosis of DCIS and followed for subsequent IBC to: identify/validate miRNA expression changes in DCIS tissue associated with risk of subsequent IBC; evaluate ipsilateral IBC risk in association with two previously identified marker sets (triple immunopositivity for p16, COX-2, Ki67; Oncotype DX Breast DCIS score); examine the association of risk factor data with IBC risk., Methods and Analysis: We are conducting a series of case-control studies nested within the cohort. Cases are women with DCIS who developed subsequent IBC; controls (2/case) are matched to cases on calendar year of and age at DCIS diagnosis. We project 485 cases/970 controls in the aim focused on risk factors. We estimate obtaining FFPE tissue for 320 cases/640 controls for the aim focused on miRNAs; of these, 173 cases/346 controls will be included in the aim focused on p16, COX-2 and Ki67 immunopositivity, and of the latter, 156 case-control pairs will be included in the aim focused on the Oncotype DX Breast DCIS score®. Multivariate conditional logistic regression will be used for statistical analyses., Ethics and Dissemination: Ethics approval was obtained from the Institutional Review Boards of Albert Einstein College of Medicine (IRB 2014-3611), Kaiser Permanente Colorado, Kaiser Permanente Hawaii, Henry Ford Health System, Mayo Clinic, Marshfield Clinic Research Institute and Hackensack Meridian Health, and from Lifespan Research Protection Office. The study results will be presented at meetings and published in peer-reviewed journals., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
31. A Permanent Window for Investigating Cancer Metastasis to the Lung.
- Author
-
Borriello L, Traub B, Coste A, Oktay MH, and Entenberg D
- Subjects
- Humans, Lung diagnostic imaging, Magnetic Resonance Imaging, Positron-Emission Tomography, Tomography, X-Ray Computed, Lung Neoplasms diagnostic imaging, Neoplasms
- Abstract
Metastasis, accounting for ~90% of cancer-related mortality, involves the systemic spread of cancer cells from primary tumors to secondary sites such as the bone, brain, and lung. Although extensively studied, the mechanistic details of this process remain poorly understood. While common imaging modalities, including computed tomography (CT), positron emission tomography (PET), and magnetic resonance imaging (MRI), offer varying degrees of gross visualization, each lacks the temporal and spatial resolution necessary to detect the dynamics of individual tumor cells. To address this, numerous techniques have been described for intravital imaging of common metastatic sites. Of these sites, the lung has proven especially challenging to access for intravital imaging owing to its delicacy and critical role in sustaining life. Although several approaches have previously been described for single-cell intravital imaging of the intact lung, all involve highly invasive and terminal procedures, limiting the maximum possible imaging duration to 6-12 h. Described here is an improved technique for the permanent implantation of a minimally invasive thoracic optical Window for High-Resolution Imaging of the Lung (WHRIL). Combined with an adapted approach to microcartography, the innovative optical window facilitates serial intravital imaging of the intact lung at single-cell resolution across multiple imaging sessions and spanning multiple weeks. Given the unprecedented duration of time over which imaging data can be gathered, the WHRIL can facilitate the accelerated discovery of the dynamic mechanisms underlying metastatic progression and numerous additional biologic processes within the lung.
- Published
- 2021
- Full Text
- View/download PDF
32. SUN-MKL1 Crosstalk Regulates Nuclear Deformation and Fast Motility of Breast Carcinoma Cells in Fibrillar ECM Microenvironment.
- Author
-
Sharma VP, Williams J, Leung E, Sanders J, Eddy R, Castracane J, Oktay MH, Entenberg D, and Condeelis JS
- Subjects
- Animals, Cell Line, Tumor, Cell Nucleus pathology, Extracellular Matrix pathology, Female, Mammary Neoplasms, Animal pathology, Mice, SCID, Rats, Mice, Cell Movement, Cell Nucleus metabolism, Extracellular Matrix metabolism, Mammary Neoplasms, Animal metabolism, Microtubule-Associated Proteins metabolism, Neoplasm Proteins metabolism, Transcription Factors metabolism, Tumor Microenvironment
- Abstract
Aligned collagen fibers provide topography for the rapid migration of single tumor cells (streaming migration) to invade the surrounding stroma, move within tumor nests towards blood vessels to intravasate and form distant metastases. Mechanisms of tumor cell motility have been studied extensively in the 2D context, but the mechanistic understanding of rapid single tumor cell motility in the in vivo context is still lacking. Here, we show that streaming tumor cells in vivo use collagen fibers with diameters below 3 µm. Employing 1D migration assays with matching in vivo fiber dimensions, we found a dependence of tumor cell motility on 1D substrate width, with cells moving the fastest and the most persistently on the narrowest 1D fibers (700 nm-2.5 µm). Interestingly, we also observed nuclear deformation in the absence of restricting extracellular matrix pores during high speed carcinoma cell migration in 1D, similar to the nuclear deformation observed in tumor cells in vivo. Further, we found that actomyosin machinery is aligned along the 1D axis and actomyosin contractility synchronously regulates cell motility and nuclear deformation. To further investigate the link between cell speed and nuclear deformation, we focused on the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex proteins and SRF-MKL1 signaling, key regulators of mechanotransduction, actomyosin contractility and actin-based cell motility. Analysis of The Cancer Genome Atlas dataset showed a dramatic decrease in the LINC complex proteins SUN1 and SUN2 in primary tumor compared to the normal tissue. Disruption of LINC complex by SUN1 + 2 KD led to multi-lobular elongated nuclei, increased tumor cell motility and concomitant increase in F-actin, without affecting Lamin proteins. Mechanistically, we found that MKL1, an effector of changes in cellular G-actin to F-actin ratio, is required for increased 1D motility seen in SUN1 + 2 KD cells. Thus, we demonstrate a previously unrecognized crosstalk between SUN proteins and MKL1 transcription factor in modulating nuclear shape and carcinoma cell motility in an in vivo relevant 1D microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
33. Breast Cancer Cell Re-Dissemination from Lung Metastases-A Mechanism for Enhancing Metastatic Burden.
- Author
-
Borriello L, Condeelis J, Entenberg D, and Oktay MH
- Abstract
Although metastatic disease is the primary cause of mortality in cancer patients, the mechanisms leading to overwhelming metastatic burden are still incompletely understood. Metastases are the endpoint of a series of multi-step events involving cancer cell intravasation, dissemination to distant organs, and outgrowth to metastatic colonies. Here we show, for the first-time, that breast cancer cells do not solely disseminate to distant organs from primary tumors and metastatic nodules in the lymph nodes, but also do so from lung metastases. Thus, our findings indicate that metastatic dissemination could continue even after the removal of the primary tumor. Provided that the re-disseminated cancer cells initiate growth upon arrival to distant sites, cancer cell re-dissemination from metastatic foci could be one of the crucial mechanisms leading to overt metastases and patient demise. Therefore, the development of new therapeutic strategies to block cancer cell re-dissemination would be crucial to improving survival of patients with metastatic disease.
- Published
- 2021
- Full Text
- View/download PDF
34. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy.
- Author
-
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, and Karagiannis GS
- Subjects
- Animals, Biomarkers, Cell Movement, Disease Management, Disease Progression, Disease Susceptibility, Epithelial-Mesenchymal Transition, Humans, Immunotherapy, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Neoplasms therapy, Tumor Escape immunology, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Tumor-Associated Macrophages pathology, Neoplasm Metastasis immunology, Neoplasms etiology, Neoplasms pathology, Tumor Microenvironment immunology
- Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Asiry, Kim, Filippou, Sanchez, Entenberg, Marks, Oktay and Karagiannis.)
- Published
- 2021
- Full Text
- View/download PDF
35. Hematogenous Dissemination of Breast Cancer Cells From Lymph Nodes Is Mediated by Tumor MicroEnvironment of Metastasis Doorways.
- Author
-
Coste A, Karagiannis GS, Wang Y, Xue EA, Lin Y, Skobe M, Jones JG, Oktay MH, Condeelis JS, and Entenberg D
- Abstract
In primary breast tumors, cancer cells hematogenously disseminate through doorways in the vasculature composed of three-cell complexes (known as Tumor MicroEnvironment of Metastasis) comprising a perivascular macrophage, a tumor cell overexpressing the actin-regulatory protein Mammalian Enabled (Mena), and an endothelial cell, all in direct physical contact. It has been previously shown that once tumor cells establish lymph node metastases in patients, TMEM doorways form in the metastatic tumor cell nests. However, it has not been established if such lymph node-TMEM doorways actively transit tumor cells into the peripheral circulation and on to tertiary sites. To address this question in this short report, we used a mouse model of lymph node metastasis to demonstrate that TMEM doorways: (1) exist in tumor-positive lymph nodes of mice, (2) are restricted to the blood vascular endothelium, (3) serve as a mechanism for further dissemination to peripheral sites such as to the lungs, and (4) their activity can be abrogated by a pharmaceutical intervention. Our data suggest that cancer cell dissemination via TMEM doorways is a common mechanism of breast cancer cell dissemination to distant sites and thus the pharmacological targeting of TMEM may be necessary, even after resection of the primary tumor, to suppress cancer cell dissemination., (Copyright © 2020 Coste, Karagiannis, Wang, Xue, Lin, Skobe, Jones, Oktay, Condeelis and Entenberg.)
- Published
- 2020
- Full Text
- View/download PDF
36. The role of the tumor microenvironment in tumor cell intravasation and dissemination.
- Author
-
Borriello L, Karagiannis GS, Duran CL, Coste A, Oktay MH, Entenberg D, and Condeelis JS
- Subjects
- Humans, Neoplasm Invasiveness, Neoplasm Metastasis, Tumor Microenvironment physiology
- Abstract
Metastasis, a process that requires tumor cell dissemination followed by tumor growth, is the primary cause of death in cancer patients. An essential step of tumor cell dissemination is intravasation, a process by which tumor cells cross the blood vessel endothelium and disseminate to distant sites. Studying this process is of utmost importance given that intravasation in the primary tumor, as well as the secondary and tertiary metastases, is the key step in the systemic spread of tumor cells, and that this process continues even after removal of the primary tumor. High-resolution intravital imaging of the tumor microenvironment of breast carcinoma has revealed that tumor cell intravasation exclusively occurs at doorways, termed "Tumor MicroEnvironment of Metastasis" (TMEM), composed of three different cell types: a Tie2
high /VEGFhigh perivascular macrophage, a Mena overexpressing tumor cell, and an endothelial cell, all in direct contact. In this review article, we discuss the interactions between these cell types, the subsequent signaling events which lead to tumor cell intravasation, and the role of invadopodia in supporting tumor cell invasion and dissemination. We end our review by discussing how the knowledge acquired from the use of intravital imaging is now leading to new clinical trials targeting tumor cell dissemination and preventing metastatic progression., (Copyright © 2020 Elsevier GmbH. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
37. The Contribution of Race to Breast Tumor Microenvironment Composition and Disease Progression.
- Author
-
Kim G, Pastoriza JM, Condeelis JS, Sparano JA, Filippou PS, Karagiannis GS, and Oktay MH
- Abstract
Breast cancer is the second most commonly diagnosed cancer in American women following skin cancer. Despite overall decrease in breast cancer mortality due to advances in treatment and earlier screening, black patients continue to have 40% higher risk of breast cancer related death compared to white patients. This disparity in outcome persists even when controlled for access to care and stage at presentation and has been attributed to differences in tumor subtypes or gene expression profiles. There is emerging evidence that the tumor microenvironment (TME) may contribute to the racial disparities in outcome as well. Here, we provide a comprehensive review of current literature available regarding race-dependent differences in the TME. Notably, black patients tend to have a higher density of pro-tumorigenic immune cells (e.g., M2 macrophages, regulatory T cells) and microvasculature. Although immune cells are classically thought to be anti-tumorigenic, increase in M2 macrophages and angiogenesis may lead to a paradoxical increase in metastasis by forming doorways of tumor cell intravasation called tumor microenvironment of metastasis (TMEM). Furthermore, black patients also have higher serum levels of inflammatory cytokines, which provide a positive feedback loop in creating a pro-metastatic TME. Lastly, we propose that the higher density of immune cells and angiogenesis observed in the TME of black patients may be a result of evolutionary selection for a more robust immune response in patients of African geographic ancestry. Better understanding of race-dependent differences in the TME will aid in overcoming the racial disparity in breast cancer mortality., (Copyright © 2020 Kim, Pastoriza, Condeelis, Sparano, Filippou, Karagiannis and Oktay.)
- Published
- 2020
- Full Text
- View/download PDF
38. Stem Cell Determinant SOX9 Promotes Lineage Plasticity and Progression in Basal-like Breast Cancer.
- Author
-
Christin JR, Wang C, Chung CY, Liu Y, Dravis C, Tang W, Oktay MH, Wahl GM, and Guo W
- Subjects
- Animals, Cell Lineage, Cell Plasticity physiology, Cell Proliferation physiology, Disease Progression, Female, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Up-Regulation, Breast Neoplasms metabolism, Breast Neoplasms pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, SOX9 Transcription Factor metabolism
- Abstract
Lineage plasticity is important for the development of basal-like breast cancer (BLBC), an aggressive cancer subtype. While BLBC is likely to originate from luminal progenitor cells, it acquires substantial basal cell features and contains a heterogenous collection of cells exhibiting basal, luminal, and hybrid phenotypes. Why luminal progenitors are prone to BLBC transformation and what drives luminal-to-basal reprogramming remain unclear. Here, we show that the transcription factor SOX9 acts as a determinant for estrogen-receptor-negative (ER
- ) luminal stem/progenitor cells (LSPCs). SOX9 controls LSPC activity in part by activating both canonical and non-canonical nuclear factor κB (NF-κB) signaling. Inactivation of TP53 and RB via expression of SV40 TAg in a BLBC mouse tumor model leads to upregulation of SOX9, which drives luminal-to-basal reprogramming in vivo. Furthermore, SOX9 deletion inhibits the progression of ductal carcinoma in situ (DCIS)-like lesions to invasive carcinoma. These data show that ER- LSPC determinant SOX9 acts as a lineage plasticity driver for BLBC progression., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
39. Intravital Imaging Techniques for Biomedical and Clinical Research.
- Author
-
Coste A, Oktay MH, Condeelis JS, and Entenberg D
- Subjects
- Animals, Humans, Intravital Microscopy, Microscopy, Fluorescence, Multiphoton
- Abstract
Intravital imaging, the direct visualization of cells and tissues within a living animal, is a technique that has been employed for the better part of a century. The advent of confocal and multiphoton microscopy has dramatically improved the power of intravital imaging, making it possible to obtain optical sections of tissues non-destructively. This review discusses the various techniques used for intravital imaging, describes how intravital imaging provides information about cellular and tissue dynamics not possible to be garnered by other techniques, and details several ways in which intravital imaging is making a direct impact on the clinical care of patients. © 2019 International Society for Advancement of Cytometry., (© 2019 International Society for Advancement of Cytometry.)
- Published
- 2020
- Full Text
- View/download PDF
40. Validation of an Automated Quantitative Digital Pathology Approach for Scoring TMEM, a Prognostic Biomarker for Metastasis.
- Author
-
Entenberg D, Oktay MH, D'Alfonso T, Ginter PS, Robinson BD, Xue X, Rohan TE, Sparano JA, Jones JG, and Condeelis JS
- Abstract
Metastasis causes ~90% of breast cancer mortality. However, standard prognostic tests based mostly on proliferation genes do not measure metastatic potential. Tumor MicroEnvironment of Metastasis (TMEM), an immunohistochemical biomarker for doorways on blood vessels that support tumor cell dissemination is prognostic for metastatic outcome in breast cancer patients. Studies quantifying TMEM doorways have involved manual scoring by pathologists utilizing static digital microscopy: a labor-intensive process unsuitable for use in clinical practice. We report here a validation study evaluating a new quantitative digital pathology (QDP) tool (TMEM-DP) for identification and quantification of TMEM doorways that closely mimics pathologists' workflow and reduces pathologists' variability to levels suitable for use in a clinical setting. Blinded to outcome, QDP was applied to a nested case-control study consisting of 259 matched case-control pairs. Sixty subjects of these were manually scored by five pathologists, digitally recorded using whole slide imaging (WSI), and then used for algorithm development and optimization. Validation was performed on the remainder of the cohort. TMEM-DP shows excellent reproducibility and concordance and reduces pathologist time from ~60 min to ~5 min per case. Concordance between manual scoring and TMEM-DP was found to be >0.79. These results show that TMEM-DP is capable of accurately identifying and scoring TMEM doorways (also known as MetaSite score) equivalent to pathologists., Competing Interests: J.S.C., J.G.J., M.H.O., and T.E.R. had equity interest (including patents) in MetaStat, Inc. J.A.S. had equity interest in MetaStat, Inc. J.S.C., J.G.J., T.E.R., D.E., and J.A.S. served as consultant/advisory board members for MetaStat, Inc. The rest of the authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
41. A small-molecule allosteric inhibitor of BAX protects against doxorubicin-induced cardiomyopathy.
- Author
-
Amgalan D, Garner TP, Pekson R, Jia XF, Yanamandala M, Paulino V, Liang FG, Corbalan JJ, Lee J, Chen Y, Karagiannis GS, Sanchez LR, Liang H, Narayanagari SR, Mitchell K, Lopez A, Margulets V, Scarlata M, Santulli G, Asnani A, Peterson RT, Hazan RB, Condeelis JS, Oktay MH, Steidl U, Kirshenbaum LA, Gavathiotis E, and Kitsis RN
- Subjects
- Animals, Apoptosis physiology, Doxorubicin adverse effects, Mice, Necrosis, bcl-2-Associated X Protein, Cardiomyopathies chemically induced, Zebrafish metabolism
- Abstract
Doxorubicin remains an essential component of many cancer regimens, but its use is limited by lethal cardiomyopathy, which has been difficult to target, owing to pleiotropic mechanisms leading to apoptotic and necrotic cardiac cell death. Here we show that BAX is rate-limiting in doxorubicin-induced cardiomyopathy and identify a small-molecule BAX inhibitor that blocks both apoptosis and necrosis to prevent this syndrome. By allosterically inhibiting BAX conformational activation, this compound blocks BAX translocation to mitochondria, thereby abrogating both forms of cell death. When co-administered with doxorubicin, this BAX inhibitor prevents cardiomyopathy in zebrafish and mice. Notably, cardioprotection does not compromise the efficacy of doxorubicin in reducing leukemia or breast cancer burden in vivo, primarily due to increased priming of mitochondrial death mechanisms and higher BAX levels in cancer cells. This study identifies BAX as an actionable target for doxorubicin-induced cardiomyopathy and provides a prototype small-molecule therapeutic., Competing Interests: Competing interests R.N.K., E.G., D.A., T.P.G. and L.A.K. are inventors on a patent application PCT/US2018/021644 submitted by Albert Einstein College of Medicine that covers compounds, compositions and methods for BAX inhibition for the treatment of diseases and disorders.
- Published
- 2020
- Full Text
- View/download PDF
42. Tumor Microenvironment of Metastasis (TMEM) Doorways Are Restricted to the Blood Vessel Endothelium in Both Primary Breast Cancers and Their Lymph Node Metastases.
- Author
-
Ginter PS, Karagiannis GS, Entenberg D, Lin Y, Condeelis J, Jones JG, and Oktay MH
- Abstract
Cancer cells metastasize from primary tumors to regional lymph nodes and distant sites via the lymphatic and blood vascular systems, respectively. Our prior work has demonstrated that in primary breast tumors, cancer cells utilize a three-cell complex (known as tumor microenvironment of metastasis, or TMEM) composed of a perivascular macrophage, a tumor cell expressing high levels of the actin-regulatory protein mammalian enabled (Mena), and an endothelial cell as functional "doorways" for hematogenous dissemination. Here, we studied a well-annotated case-control cohort of human invasive ductal carcinoma of the breast and metastatic lymph nodes from a separate breast cancer cohort. We demonstrate that in primary breast tumors, blood vessels are always present within tumor cell nests (TCNs) and tumor-associated stroma (TAS), while lymphatic vessels are only occasionally present in TCN and TAS. Furthermore, TMEM doorways not only exist in primary tumors as previously reported but also in lymph node metastases. In addition, we show that TMEM intravasation doorways are restricted to the blood vascular endothelium in both primary tumors and lymph node metastases, suggesting that breast cancer dissemination to distant sites from both primary tumors and metastatic foci in lymph nodes occurs hematogenously at TMEM doorways. TMEMs are very rarely detected at lymphatic vessels and do not confer clinical prognostic significance, indicating they are not participants in TMEM-associated hematogenous dissemination. These findings are consistent with recent observations that hematogenous dissemination from lymph nodes occurs via blood vessels., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
43. Chemotherapy-Induced Metastasis: Molecular Mechanisms, Clinical Manifestations, Therapeutic Interventions.
- Author
-
Karagiannis GS, Condeelis JS, and Oktay MH
- Subjects
- Animals, Cell Movement, Humans, Immunity, Innate drug effects, Neoplasm Metastasis, Neoplasms drug therapy, Neoplasms immunology, Tumor Microenvironment drug effects, Antineoplastic Agents adverse effects, Immunity, Innate immunology, Neoadjuvant Therapy adverse effects, Neoplasms pathology, Tumor Microenvironment immunology
- Abstract
Chemotherapy offers long-term clinical benefits to many patients with advanced cancer. However, recent evidence has linked the cytotoxic effects of chemotherapy with the de novo elicitation of a prometastatic tumor microenvironment. This "modified" tumor microenvironment is triggered by a chemotherapy-driven cytokine storm or through direct effects of certain chemotherapeutics on stromal and/or immune cells, the most critical being tumor-associated macrophages. These chemotherapy-educated cells act as facilitators in tumor-host cell interactions promoting the establishment of distant metastasis. Certain clinical studies now offer substantial evidence that prometastatic changes are indeed identified in the tumor microenvironment of certain patient subpopulations, especially those that do not present with any pathologic response after neoadjuvant chemotherapy. Deciphering the exact contextual prerequisites for chemotherapy-driven metastasis will be paramount for designing novel mechanism-based treatments for circumventing chemotherapy-induced metastasis., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
44. The emerging roles of macrophages in cancer metastasis and response to chemotherapy.
- Author
-
Sanchez LR, Borriello L, Entenberg D, Condeelis JS, Oktay MH, and Karagiannis GS
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Extracellular Matrix, Humans, Immunomodulation, Macrophages pathology, Neoplasm Metastasis, Neoplasm Staging, Neoplasms drug therapy, Neoplasms pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neovascularization, Pathologic, Treatment Outcome, Tumor Microenvironment, Macrophages immunology, Macrophages metabolism, Neoplasms etiology, Neoplasms metabolism
- Abstract
Macrophages represent a heterogeneous group of cells, capable of carrying out distinct functions in a variety of organs and tissues. Even within individual tissues, their functions can vary with location. Tumor-associated macrophages (TAMs) specialize into three major subtypes that carry out multiple tasks simultaneously. This is especially true in the context of metastasis, where TAMs establish most of the cellular and molecular prerequisites for successful cancer cell dissemination and seeding to the secondary site. Perivascular TAMs operate in the perivascular niche, where they promote tumor angiogenesis and aid in the assembly of intravasation sites called tumor microenvironment of metastasis (TMEM). Streaming TAMs co-migrate with tumor cells (irrespective of the perivascular niche) and promote matrix remodeling, tumor cell invasiveness, and an immunosuppressive local microenvironment. Premetastatic TAMs are recruited to the premetastatic niche, where they can assist in tumor cell extravasation, seeding, and metastatic colonization. The dynamic interplay between TAMs and tumor cells can also modify the ability of the latter to resist cytotoxic chemotherapy (a phenotype known as environment-mediated drug resistance) and induce chemotherapy-mediated pro-metastatic microenvironmental changes. These observations suggest that future therapeutics should be designed to target TAMs with the aim of suppressing the metastatic potential of tumors and rendering chemotherapy more efficient., (©2019 Society for Leukocyte Biology.)
- Published
- 2019
- Full Text
- View/download PDF
45. Assessing Tumor Microenvironment of Metastasis Doorway-Mediated Vascular Permeability Associated with Cancer Cell Dissemination using Intravital Imaging and Fixed Tissue Analysis.
- Author
-
Karagiannis GS, Pastoriza JM, Borriello L, Jafari R, Coste A, Condeelis JS, Oktay MH, and Entenberg D
- Subjects
- Animals, Breast Neoplasms blood supply, Cell Line, Tumor, Female, Humans, Mice, Neoplasm Metastasis, Neoplasm Recurrence, Local pathology, Breast Neoplasms pathology, Capillary Permeability, Intravital Microscopy, Tissue Fixation, Tumor Microenvironment
- Abstract
The most common cause of cancer related mortality is metastasis, a process that requires dissemination of cancer cells from the primary tumor to secondary sites. Recently, we established that cancer cell dissemination in primary breast cancer and at metastatic sites in the lung occurs only at doorways called Tumor MicroEnvironment of Metastasis (TMEM). TMEM doorway number is prognostic for distant recurrence of metastatic disease in breast cancer patients. TMEM doorways are composed of a cancer cell which over-expresses the actin regulatory protein Mena in direct contact with a perivascular, proangiogenic macrophage which expresses high levels of TIE2 and VEGF, where both of these cells are tightly bound to a blood vessel endothelial cell. Cancer cells can intravasate through TMEM doorways due to transient vascular permeability orchestrated by the joint activity of the TMEM-associated macrophage and the TMEM-associated Mena-expressing cancer cell. In this manuscript, we describe two methods for assessment of TMEM-mediated transient vascular permeability: intravital imaging and fixed tissue immunofluorescence. Although both methods have their advantages and disadvantages, combining the two may provide the most complete analyses of TMEM-mediated vascular permeability as well as microenvironmental prerequisites for TMEM function. Since the metastatic process in breast cancer, and possibly other types of cancer, involves cancer cell dissemination via TMEM doorways, it is essential to employ well established methods for the analysis of the TMEM doorway activity. The two methods described here provide a comprehensive approach to the analysis of TMEM doorway activity, either in naïve or pharmacologically treated animals, which is of paramount importance for pre-clinical trials of agents that prevent cancer cell dissemination via TMEM.
- Published
- 2019
- Full Text
- View/download PDF
46. Loss of amphiregulin reduces myoepithelial cell coverage of mammary ducts and alters breast tumor growth.
- Author
-
Mao SPH, Park M, Cabrera RM, Christin JR, Karagiannis GS, Oktay MH, Zaiss DMW, Abrams SI, Guo W, Condeelis JS, Kenny PA, and Segall JE
- Subjects
- Amphiregulin genetics, Animals, Antigens, Polyomavirus Transforming genetics, Antigens, Polyomavirus Transforming metabolism, Cell Line, Tumor, Cell Proliferation, Epithelial Cells virology, Female, Humans, Mammary Glands, Animal cytology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental virology, Mammary Tumor Virus, Mouse genetics, Mammary Tumor Virus, Mouse pathogenicity, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neoplasm Invasiveness pathology, Polyomavirus genetics, Polyomavirus immunology, Amphiregulin metabolism, Epithelial Cells pathology, Mammary Glands, Animal pathology, Mammary Neoplasms, Experimental pathology
- Abstract
Background: Amphiregulin (AREG), a ligand of the epidermal growth factor receptor, is not only essential for proper mammary ductal development, but also associated with breast cancer proliferation and growth. In the absence of AREG, mammary ductal growth is stunted and fails to expand. Furthermore, suppression of AREG expression in estrogen receptor-positive breast tumor cells inhibits in-vitro and in-vivo growth., Methods: We crossed AREG-null (AREG
-/- ) mice with the murine luminal B breast cancer model, MMTV-PyMT (PyMT), to generate spontaneous breast tumors that lack AREG (AREG-/- PyMT). We evaluated tumor growth, cytokeratin-8 (K8)-positive luminal cells, cytokeratin-14 (K14)-positive myoepithelial cells, and expression of AREG, Ki67, and PyMT. Primary myoepithelial cells from nontumor-bearing AREG+/+ mice underwent fluorescence-activated cell sorting and were adapted to culture for in-vitro coculture studies with AT-3 cells, a cell line derived from C57Bl/6 PyMT mammary tumors., Results: Intriguingly, PyMT-induced lesions progress more rapidly in AREG-/- mice than in AREG+/+ mice. Quantification of K8+ luminal and K14+ myoepithelial cells in non-PyMT AREG-/- mammary glands showed fewer K14+ cells and a thinner myoepithelial layer. Study of AT-3 cells indicated that coculture with myoepithelial cells or exposure to AREG, epidermal growth factor, or basic fibroblast growth factor can suppress PyMT expression. Late-stage AREG-/- PyMT tumors are significantly less solid in structure, with more areas of papillary and cystic growth. Papillary areas appear to be both less proliferative and less necrotic. In The Cancer Genome Atlas database, luminal-B invasive papillary carcinomas have lower AREG expression than luminal B invasive ductal carcinomas., Conclusions: Our study has revealed a previously unknown role of AREG in myoepithelial cell development and PyMT expression. AREG expression is essential for proper myoepithelial coverage of mammary ducts. Both AREG and myoepithelial cells can suppress PyMT expression. We find that lower AREG expression is associated with invasive papillary breast cancer in both the MMTV-PyMT model and human breast cancer.- Published
- 2018
- Full Text
- View/download PDF
47. Black race and distant recurrence after neoadjuvant or adjuvant chemotherapy in breast cancer.
- Author
-
Pastoriza JM, Karagiannis GS, Lin J, Lanjewar S, Entenberg D, Condeelis JS, Sparano JA, Xue X, Rohan TE, and Oktay MH
- Subjects
- Breast Neoplasms epidemiology, Chemotherapy, Adjuvant, Cohort Studies, Disease-Free Survival, Female, Humans, Kaplan-Meier Estimate, Middle Aged, Neoadjuvant Therapy, Neoplasm Recurrence, Local epidemiology, New York epidemiology, Black or African American, Black People statistics & numerical data, Breast Neoplasms drug therapy, Breast Neoplasms ethnology, Neoplasm Recurrence, Local ethnology, White People statistics & numerical data
- Abstract
Black race compared to white race is associated with more advanced stage and biologically aggressive breast cancer. Consequently, black patients are more frequently treated with neoadjuvant chemotherapy (NAC) than white patients. However, it is unclear how distant recurrence-free survival (DRFS) of black patients treated with NAC, compares to DRFS of black patients treated with adjuvant chemotherapy (AC). We evaluated the association between race, distant recurrence, and type of chemotherapy (AC or NAC) in localized or locally advanced breast cancer. We evaluated DRFS in 807 patients, including 473 black, 252 white, 56 Hispanic, and 26 women of other or mixed race. The association between AC or NAC and DRFS was examined using multivariate Cox proportional hazard models that included race, age, stage, estrogen receptor (ER) and triple negative (TN) status. When the black and white subjects were pooled for the analysis the features associated with worse DRFS included stage III disease and age < 50 years, but not ER-negative disease, TN disease, the use of NAC, or black race. However, in the analysis stratified by race NAC was associated with worse DRFS compared to AC in black (HR 2.70; 95% CI 1.73-4.22; p < 0.0001), but not in white women (HR 1.29, 95% CI 0.56-2.95; p = 0.36). Black patients treated with NAC had worse DRFS than black patients treated with AC, or white patients treated with either NAC or AC. These findings need to be validated in a large-scale observational study and the effect of NAC on the breast cancer microenvironment in black women needs to be further evaluated.
- Published
- 2018
- Full Text
- View/download PDF
48. Chemotherapy-induced metastasis: mechanisms and translational opportunities.
- Author
-
Karagiannis GS, Condeelis JS, and Oktay MH
- Subjects
- Animals, Humans, Neoplasm Metastasis, Neoplastic Cells, Circulating drug effects, Neoplastic Cells, Circulating pathology, Antineoplastic Agents adverse effects, Neoplasms drug therapy, Neoplasms pathology
- Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
- Published
- 2018
- Full Text
- View/download PDF
49. Chemotherapy-induced metastasis in breast cancer.
- Author
-
Karagiannis GS, Condeelis JS, and Oktay MH
- Published
- 2017
- Full Text
- View/download PDF
50. A metastasis biomarker (MetaSite Breast ™ Score) is associated with distant recurrence in hormone receptor-positive, HER2-negative early-stage breast cancer.
- Author
-
Sparano JA, Gray R, Oktay MH, Entenberg D, Rohan T, Xue X, Donovan M, Peterson M, Shuber A, Hamilton DA, D'Alfonso T, Goldstein LJ, Gertler F, Davidson NE, Condeelis J, and Jones J
- Abstract
Metastasis is the primary cause of death in early-stage breast cancer. We evaluated the association between a metastasis biomarker, which we call "Tumor Microenviroment of Metastasis" (TMEM), and risk of recurrence. TMEM are microanatomic structures where invasive tumor cells are in direct contact with endothelial cells and macrophages, and which serve as intravasation sites for tumor cells into the circulation. We evaluated primary tumors from 600 patients with Stage I-III breast cancer treated with adjuvant chemotherapy in trial E2197 (NCT00003519), plus endocrine therapy for hormone receptor (HR)+ disease. TMEM were identified and enumerated using an analytically validated, fully automated digital pathology/image analysis method (MetaSite Breast ™), hereafter referred to as MetaSite Score (MS). The objectives were to determine the association between MS and distant relapse free interval (DRFI) and relapse free interval (RFI). MS was not associated with tumor size or nodal status, and correlated poorly with Oncotype DX Recurrence Score ( r = 0.29) in 297 patients with HR+/HER2- disease. Proportional hazards models revealed a significant positive association between continuous MS and DRFI ( p = 0.001) and RFI ( p = 0.00006) in HR+/HER2- disease in years 0-5, and by MS tertiles for DRFI ( p = 0.04) and RFI ( p = 0.01), but not after year 5 or in triple negative or HER2+ disease. Multivariate models in HR+/HER- disease including continuous MS, clinical covariates, and categorical Recurrence Score (<18, 18-30, > 30) showed MS is an independent predictor for 5-year RFI ( p = 0.05). MetaSite Score provides prognostic information for early recurrence complementary to clinicopathologic features and Recurrence Score.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.