1
|
Mao YQ, Han SF, Zhang SL, Zhang ZY, Kong CY, Chen HL, Li ZM, Cai PR, Han B, Wang LS. An approach using Caenorhabditis elegans screening novel targets to suppress tumour cell proliferation. Cell Prolif 2020; 53:e12832. [PMID: 32452127 PMCID: PMC7309951 DOI: 10.1111/cpr.12832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Tumour cell proliferation requires high metabolism to meet the bioenergetics and biosynthetic needs. Dauer in Caenorhabditis elegans is characterized by lower metabolism, and we established an approach with C elegans to find potential tumour therapy targets. MATERIALS AND METHODS RNAi screening was used to find dauer-related genes, and these genes were further analysed in glp-1(-) mutants for tumour-suppressing testing. The identified tumour-related genes were verified in clinical tumour tissues. RESULTS The lifespan of glp-1(-) mutants was found to be extended by classical dauer formation signalling. Then, 61 of 287 kinase-coding genes in Caenorhabditis elegans were identified as dauer-related genes, of which 27 were found to be homologous to human oncogenes. Furthermore, 12 dauer-related genes were randomly selected for tumour-suppressing test, and six genes significantly extended the lifespan of glp-1(-) mutants. Of these six genes, F47D12.9, W02B12.12 and gcy-21 were newly linked to dauer formation. These three new dauer-related genes significantly suppressed tumour cell proliferation and thus extended the lifespan of glp-1(-) mutants in a longevity- or dauer-independent manner. The mRNA expression profiles indicated that these dauer-related genes trigged similar low metabolism pattern in glp-1(-) mutants. Notably, the expression of homolog gene DCAF4L2/F47D12.9, TSSK6/W02B12.12 and NPR1/gcy-21 was found to be higher in glioma compared with adjacent normal tissue. In addition, the high expression of TSSK6/W02B12.12 and NPR1/gcy-21 correlated with a worse survival in glioma patients. CONCLUSIONS Dauer gene screening in combination with tumour-suppressing test in glp-1(-) mutants provided a useful approach to find potential targets for tumour therapy via suppressing tumour cell proliferation and rewiring tumour cell metabolism.
Collapse
Affiliation(s)
- Yu-Qin Mao
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - San-Feng Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Shi-Long Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zheng-Yan Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Chao-Yue Kong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Hui-Ling Chen
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhan-Ming Li
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Pei-Ran Cai
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Bing Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Li-Shun Wang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Garcia V, Bonhoeffer S, Fu F. Cancer-induced immunosuppression can enable effectiveness of immunotherapy through bistability generation: A mathematical and computational examination. J Theor Biol 2020; 492:110185. [PMID: 32035826 PMCID: PMC7079339 DOI: 10.1016/j.jtbi.2020.110185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
Abstract
The presence of an immunological barrier in cancer- immune system interaction (CISI) is consistent with the bistability patterns in that system. In CISI models, bistability patterns are consistent with immunosuppressive effects dominating immunoproliferative effects. Bistability could be harnessed to devise effective combination immunotherapy approaches.
Cancer immunotherapies rely on how interactions between cancer and immune system cells are constituted. The more essential to the emergence of the dynamical behavior of cancer growth these interactions are, the more effectively they may be used as mechanisms for interventions. Mathematical modeling can help unearth such connections, and help explain how they shape the dynamics of cancer growth. Here, we explored whether there exist simple, consistent properties of cancer-immune system interaction (CISI) models that might be harnessed to devise effective immunotherapy approaches. We did this for a family of three related models of increasing complexity. To this end, we developed a base model of CISI, which captures some essential features of the more complex models built on it. We find that the base model and its derivates can plausibly reproduce biological behavior that is consistent with the notion of an immunological barrier. This behavior is also in accord with situations in which the suppressive effects exerted by cancer cells on immune cells dominate their proliferative effects. Under these circumstances, the model family may display a pattern of bistability, where two distinct, stable states (a cancer-free, and a full-grown cancer state) are possible. Increasing the effectiveness of immune-caused cancer cell killing may remove the basis for bistability, and abruptly tip the dynamics of the system into a cancer-free state. Additionally, in combination with the administration of immune effector cells, modifications in cancer cell killing may be harnessed for immunotherapy without the need for resolving the bistability. We use these ideas to test immunotherapeutic interventions in silico in a stochastic version of the base model. This bistability-reliant approach to cancer interventions might offer advantages over those that comprise gradual declines in cancer cell numbers.
Collapse
Affiliation(s)
- Victor Garcia
- Institute of Applied Simulation, Zurich University of Applied Sciences, Einsiedlerstrasse 31a, 8820 Wädenswil, Switzerland; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland; Institute for Social and Preventive Medicine, University of Bern, Finkenhubelweg 11, 3012 Bern, Switzerland; Department of Biology, Stanford University, 371 Serra Mall, Stanford CA 94305, USA.
| | | | - Feng Fu
- Department of Mathematics, Dartmouth College, 27 N. Main Street, 6188 Kemeny Hall, Hanover, NH 03755-3551, USA; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland
| |
Collapse
|
3
|
Flüh C, Mafael V, Adamski V, Synowitz M, Held-Feindt J. Dormancy and NKG2D system in brain metastases: Analysis of immunogenicity. Int J Mol Med 2019; 45:298-314. [PMID: 31894267 PMCID: PMC6984787 DOI: 10.3892/ijmm.2019.4449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Patients with breast cancer (BC) and lung cancer (LC) are prone to developing brain metastases, which are associated with devastating prognoses. Dormant tumor cells, a population of non-apoptotic quiescent cells and immunological escape mechanisms, including the Natural Killer Group 2 member D (NKG2D) receptor-ligand system, represent potential mechanisms of tumor recurrence. To date, the immunological characteristics of dormant tumor cells concerning the NKG2D system in cerebral malignancies are mostly unknown. In the present study, an extensive characterization of dormant and NKG2D ligand (NKG2DL)+ cells in cerebral metastases was performed. The expression profiles and localization patterns of various NKG2DL and several dormancy markers were analyzed in solid human brain metastases from patients with BC and LC using immunostaining and reverse transcription-quantitative polymerase chain reaction analyses. Statistical analysis was performed using Student's t-test and Bravais-Pearson correlation analysis. Not only 'peripheral', but also 'central' dormancy markers, which had been previously described in primary brain tumors, were identified in all cerebral metastases at detectable levels at protein and mRNA levels. Notably, the majority of NKG2DL+ cells were also positive for 'central' dormancy markers, but not 'peripheral' dormancy markers in both patient groups. This cell population may represent a promising future therapeutic target.
Collapse
Affiliation(s)
- Charlotte Flüh
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Victor Mafael
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Vivian Adamski
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| |
Collapse
|
4
|
Abstract
Anticancer immunotherapies involving the use of immune-checkpoint inhibitors or adoptive cellular transfer have emerged as new therapeutic pillars within oncology. These treatments function by overcoming or relieving tumour-induced immunosuppression, thereby enabling immune-mediated tumour clearance. While often more effective and better tolerated than traditional and targeted therapies, many patients have innate or acquired resistance to immunotherapies. Cancer immunoediting is the process whereby the immune system can both constrain and promote tumour development, which proceeds through three phases termed elimination, equilibrium and escape. Throughout these phases, tumour immunogenicity is edited, and immunosuppressive mechanisms that enable disease progression are acquired. The mechanisms of resistance to immunotherapy seem to broadly overlap with those used by cancers as they undergo immunoediting to evade detection by the immune system. In this Review, we discuss how a deeper understanding of the mechanisms underlying the cancer immunoediting process can provide insight into the development of resistance to immunotherapies and the strategies that can be used to overcome such resistance.
Collapse
|
5
|
Malinzi J, Amima I. Mathematical analysis of a tumour-immune interaction model: A moving boundary problem. Math Biosci 2018; 308:8-19. [PMID: 30537482 DOI: 10.1016/j.mbs.2018.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/06/2018] [Accepted: 12/07/2018] [Indexed: 01/21/2023]
Abstract
A spatio-temporal mathematical model, in the form of a moving boundary problem, to explain cancer dormancy is developed. Analysis of the model is carried out for both temporal and spatio-temporal cases. Stability analysis and numerical simulations of the temporal model replicate experimental observations of immune-induced tumour dormancy. Travelling wave solutions of the spatio-temporal model are determined using the hyperbolic tangent method and minimum wave speeds of invasion are calculated. Travelling wave analysis depicts that cell invasion dynamics are mainly driven by their motion and growth rates. A stability analysis of the spatio-temporal model shows a possibility of dynamical stabilization of the tumour-free steady state. Simulation results reveal that the tumour swells to a dormant level.
Collapse
Affiliation(s)
- Joseph Malinzi
- Department of Mathematics and Applied Mathematics, University of Pretoria, Private Bag X 20, Hatfield, Pretoria 0028, South Africa.
| | - Innocenter Amima
- Department of Mathematical Sciences, Stellenbosch University, Private Bag X1 Matieland, 7602, South Africa.
| |
Collapse
|
6
|
Wang H, Stoecklein NH, Lin PP, Gires O. Circulating and disseminated tumor cells: diagnostic tools and therapeutic targets in motion. Oncotarget 2018; 8:1884-1912. [PMID: 27683128 PMCID: PMC5352105 DOI: 10.18632/oncotarget.12242] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022] Open
Abstract
Enumeration of circulating tumor cells (CTCs) in peripheral blood with the gold standard CellSearchTM has proven prognostic value for tumor recurrence and progression of metastatic disease. Therefore, the further molecular characterization of isolated CTCs might have clinical relevance as liquid biopsy for therapeutic decision-making and to monitor disease progression. The direct analysis of systemic cancer appears particularly important in view of the known disparity in expression of therapeutic targets as well as epithelial-to-mesenchymal transition (EMT)-based heterogeneity between primary and systemic tumor cells, which all substantially complicate monitoring and therapeutic targeting at present. Since CTCs are the potential precursor cells of metastasis, their in-depth molecular profiling should also provide a useful resource for target discovery. The present review will discuss the use of systemically spread cancer cells as liquid biopsy and focus on potential target antigens.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, Medical Faculty, University Hospital of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy of Head and Neck Tumors, Helmholtz, Germany
| |
Collapse
|
7
|
Lee E, Decker AM, Cackowski FC, Kana LA, Yumoto K, Jung Y, Wang J, Buttitta L, Morgan TM, Taichman RS. Growth Arrest-Specific 6 (GAS6) Promotes Prostate Cancer Survival by G 1 Arrest/S Phase Delay and Inhibition of Apoptosis During Chemotherapy in Bone Marrow. J Cell Biochem 2016; 117:2815-2824. [PMID: 27153245 PMCID: PMC5223280 DOI: 10.1002/jcb.25582] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) is known to develop resistance to chemotherapy. Growth arrest-specific 6 (GAS6), plays a role in tumor progression by regulating growth in many cancers. Here, we explored how GAS6 regulates the cell cycle and apoptosis of PCa cells in response to chemotherapy. We found that GAS6 is sufficient to significantly increase the fraction of cells in G1 and the duration of phase in PCa cells. Importantly, the effect of GAS6 on G1 is potentiated during docetaxel chemotherapy. GAS6 altered the levels of several key cell cycle regulators, including the downregulation of Cyclin B1 (G2 /M phase), CDC25A, Cyclin E1, and CDK2 (S phase entry), while the upregulation of cell cycle inhibitors p27 and p21, Cyclin D1, and CDK4. Importantly, these changes became further accentuated during docetaxel treatment in the presence of GAS6. Moreover, GAS6 alters the apoptotic response of PCa cells during docetaxel chemotherapy. Docetaxel induced PCa cell apoptosis is efficiently suppressed in PCa cell culture in the presence of GAS6 or GAS6 secreted from co-cultured osteoblasts. Similarly, the GAS6-expressing bone environment protects PCa cells from apoptosis within primary tumors in vivo studies. Docetaxel induced significant levels of Caspase-3 and PARP cleavage in PCa cells, while GAS6 protected PCa cells from docetaxel-induced apoptotic signaling. Together, these data suggest that GAS6, expressed by osteoblasts in the bone marrow, plays a significant role in the regulation of PCa cell survival during chemotherapy, which will have important implications for targeting metastatic disease. J. Cell. Biochem. 117: 2815-2824, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eunsohl Lee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Ann M Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Frank C Cackowski
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, 48109, Michigan
| | - Lulia A Kana
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Kenji Yumoto
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Jingcheng Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Laura Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 48109, Michigan
| | - Todd M Morgan
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, 48109, Michigan
| | - Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan.
| |
Collapse
|
8
|
Evans EB, Lin SY. New insights into tumor dormancy: Targeting DNA repair pathways. World J Clin Oncol 2015; 6:80-88. [PMID: 26468441 PMCID: PMC4600194 DOI: 10.5306/wjco.v6.i5.80] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/02/2015] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, major strides have advanced the techniques for early detection and treatment of cancer. However, metastatic tumor growth still accounts for the majority of cancer-related deaths worldwide. In fact, breast cancers are notorious for relapsing years or decades after the initial clinical treatment, and this relapse can vary according to the type of breast cancer. In estrogen receptor-positive breast cancers, late tumor relapses frequently occur whereas relapses in estrogen receptor-negative cancers or triple negative tumors arise early resulting in a higher mortality risk. One of the main causes of metastasis is tumor dormancy in which cancer cells remain concealed, asymptomatic, and untraceable over a prolonged period of time. Under certain conditions, dormant cells can re-enter into the cell cycle and resume proliferation leading to recurrence. However, the molecular and cellular regulators underlying this transition remain poorly understood. To date, three mechanisms have been identified to trigger tumor dormancy including cellular, angiogenic, and immunologic dormancies. In addition, recent studies have suggested that DNA repair mechanisms may contribute to the survival of dormant cancer cells. In this article, we summarize the recent experimental and clinical evidence governing cancer dormancy. In addition, we will discuss the role of DNA repair mechanisms in promoting the survival of dormant cells. This information provides mechanistic insight to explain why recurrence occurs, and strategies that may enhance therapeutic approaches to prevent disease recurrence.
Collapse
|
9
|
Macnamara C, Eftimie R. Memory versus effector immune responses in oncolytic virotherapies. J Theor Biol 2015; 377:1-9. [DOI: 10.1016/j.jtbi.2015.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/27/2015] [Accepted: 04/01/2015] [Indexed: 12/01/2022]
|
10
|
Wu YY, V. Nguyen A, Wu XX, Loh M, Vu M, Zou Y, Liu Q, Guo P, Wang Y, Montgomery LL, Orlofsky A, Rand JH, Lin EY. Antiphospholipid Antibodies Promote Tissue Factor–Dependent Angiogenic Switch and Tumor Progression. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3359-75. [DOI: 10.1016/j.ajpath.2014.07.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 12/30/2022]
|
11
|
A cellular automaton model for tumor dormancy: emergence of a proliferative switch. PLoS One 2014; 9:e109934. [PMID: 25329892 PMCID: PMC4199683 DOI: 10.1371/journal.pone.0109934] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/12/2014] [Indexed: 01/06/2023] Open
Abstract
Malignant cancers that lead to fatal outcomes for patients may remain dormant for very long periods of time. Although individual mechanisms such as cellular dormancy, angiogenic dormancy and immunosurveillance have been proposed, a comprehensive understanding of cancer dormancy and the “switch” from a dormant to a proliferative state still needs to be strengthened from both a basic and clinical point of view. Computational modeling enables one to explore a variety of scenarios for possible but realistic microscopic dormancy mechanisms and their predicted outcomes. The aim of this paper is to devise such a predictive computational model of dormancy with an emergent “switch” behavior. Specifically, we generalize a previous cellular automaton (CA) model for proliferative growth of solid tumor that now incorporates a variety of cell-level tumor-host interactions and different mechanisms for tumor dormancy, for example the effects of the immune system. Our new CA rules induce a natural “competition” between the tumor and tumor suppression factors in the microenvironment. This competition either results in a “stalemate” for a period of time in which the tumor either eventually wins (spontaneously emerges) or is eradicated; or it leads to a situation in which the tumor is eradicated before such a “stalemate” could ever develop. We also predict that if the number of actively dividing cells within the proliferative rim of the tumor reaches a critical, yet low level, the dormant tumor has a high probability to resume rapid growth. Our findings may shed light on the fundamental understanding of cancer dormancy.
Collapse
|
12
|
TBK1 regulates prostate cancer dormancy through mTOR inhibition. Neoplasia 2014; 15:1064-74. [PMID: 24027431 DOI: 10.1593/neo.13402] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 12/25/2022] Open
Abstract
The mechanisms that regulate hematopoietic stem cell (HSC) dormancy and self-renewal are well established and are largely dependent on signals emanating from the HSC niche. Recently, we found that prostate cancer (PCa) cells target the HSC niche in mouse bone marrow (BM) during metastasis. Little is known, however, as to how the HSC niche may regulate dormancy in cancer cells. In this study, we investigated the effects of TANK binding kinase 1 (TBK1) on PCa dormancy in the BM niche. We found that binding with niche osteoblasts induces the expression of TBK1 in PCa cells PC3 and C4-2B. Interestingly, TBK1 interacts with mammalian target of rapamycin (mTOR) and inhibits its function. Rapamycin, an mTOR inhibitor, induces cell cycle arrest of PCa cells and enhances chemotherapeutic resistance of PCa cells. As a result, the knockdown of TBK1 decreases PCa stem-like cells and drug resistance in vitro and in vivo. Taken together, these results strongly indicate that TBK1 plays an important role in the dormancy and drug resistance of PCa.
Collapse
|
13
|
Saleem M, Agrawal T, Anees A. A study of tumour growth based on stoichiometric principles: a continuous model and its discrete analogue. JOURNAL OF BIOLOGICAL DYNAMICS 2014; 8:117-34. [PMID: 24963981 PMCID: PMC4220851 DOI: 10.1080/17513758.2014.913718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In this paper, we consider a continuous mathematically tractable model and its discrete analogue for the tumour growth. The model formulation is based on stoichiometric principles considering tumour-immune cell interactions in potassium (K (+))-limited environment. Our both continuous and discrete models illustrate 'cancer immunoediting' as a dynamic process having all three phases namely elimination, equilibrium and escape. The stoichiometric principles introduced into the model allow us to study its dynamics with the variation in the total potassium in the surrounding of the tumour region. It is found that an increase in the total potassium may help the patient fight the disease for a longer period of time. This result seems to be in line with the protective role of the potassium against the risk of pancreatic cancer as has been reported by Bravi et al. [Dietary intake of selected micronutrients and risk of pancreatic cancer: An Italian case-control study, Ann. Oncol. 22 (2011), pp. 202-206].
Collapse
Affiliation(s)
- M Saleem
- a Department of Applied Mathematics , Z.H. College of Engineering and Technology, A.M.U ., Aligarh 202002 , India
| | | | | |
Collapse
|
14
|
Wang SH, Lin SY. Tumor dormancy: potential therapeutic target in tumor recurrence and metastasis prevention. Exp Hematol Oncol 2013; 2:29. [PMID: 24502434 PMCID: PMC4176492 DOI: 10.1186/2162-3619-2-29] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/12/2013] [Indexed: 12/14/2022] Open
Abstract
In past decades, cancer patient survival has been improved with earlier detection and advancements in therapy. However, many patients who exhibit no clinical symptoms after frontline therapy subsequently suffer, often many years later, aggressive tumor recurrence. Cancer recurrence represents a critical clinical challenge in effectively treating malignancies and for patients’ quality of life. Tumor cell dormancy may help to explain treatment resistance and recurrence or metastatic reactivation. Understanding the dormant stage of tumor cells may help in discovering ways to maintain the dormant state or permanently eliminate dormant residual disseminated tumor cells. Over the past decade, numerous studies indicate that various mechanisms of tumor dormancy exist, including cellular dormancy (quiescence), angiogenic dormancy, and immunologic dormancy. In this short review, we summarize recent experimental and clinical evidence for these three mechanisms and other possible tumor microenvironment mechanisms that may influence tumor dormancy.
Collapse
Affiliation(s)
| | - Shiaw-Yih Lin
- Department of Systems Biology, Unit 950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,, Houston, TX 77054, USA.
| |
Collapse
|
15
|
Hurst RE, Hauser PJ, Kyker KD, Heinlen JE, Hodde JP, Hiles MC, Kosanke SD, Dozmorov M, Ihnat MA. Suppression and activation of the malignant phenotype by extracellular matrix in xenograft models of bladder cancer: a model for tumor cell "dormancy". PLoS One 2013; 8:e64181. [PMID: 23717563 PMCID: PMC3663841 DOI: 10.1371/journal.pone.0064181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 04/12/2013] [Indexed: 01/08/2023] Open
Abstract
A major problem in cancer research is the lack of a tractable model for delayed metastasis. Herein we show that cancer cells suppressed by SISgel, a gel-forming normal ECM material derived from Small Intestine Submucosa (SIS), in flank xenografts show properties of suppression and re-activation that are very similar to normal delayed metastasis and suggest these suppressed cells can serve as a novel model for developing therapeutics to target micrometastases or suppressed cancer cells. Co-injection with SISgel suppressed the malignant phenotype of highly invasive J82 bladder cancer cells and highly metastatic JB-V bladder cancer cells in nude mouse flank xenografts. Cells could remain viable up to 120 days without forming tumors and appeared much more highly differentiated and less atypical than tumors from cells co-injected with Matrigel. In 40% of SISgel xenografts, growth resumed in the malignant phenotype after a period of suppression or dormancy for at least 30 days and was more likely with implantation of 3 million or more cells. Ordinary Type I collagen did not suppress malignant growth, and tumors developed about as well with collagen as with Matrigel. A clear signal in gene expression over different cell lines was not seen by transcriptome microarray analysis, but in contrast, Reverse Phase Protein Analysis of 250 proteins across 4 cell lines identified Integrin Linked Kinase (ILK) signaling that was functionally confirmed by an ILK inhibitor. We suggest that cancer cells suppressed on SISgel could serve as a model for dormancy and re-awakening to allow for the identification of therapeutic targets for treating micrometastases.
Collapse
Affiliation(s)
- Robert E Hurst
- Department of Urology, College of Medicine, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Cugini C, Klepac-Ceraj V, Rackaityte E, Riggs JE, Davey ME. Porphyromonas gingivalis: keeping the pathos out of the biont. J Oral Microbiol 2013; 5:19804. [PMID: 23565326 PMCID: PMC3617648 DOI: 10.3402/jom.v5i0.19804] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 01/21/2023] Open
Abstract
The primary goal of the human microbiome initiative has been to increase our understanding of the structure and function of our indigenous microbiota and their effects on human health and predisposition to disease. Because of its clinical importance and accessibility for in vivo study, the oral biofilm is one of the best-understood microbial communities associated with the human body. Studies have shown that there is a succession of select microbial interactions that directs the maturation of a defined community structure, generating the formation of dental plaque. Although the initiating factors that lead to disease development are not clearly defined, in many individuals there is a fundamental shift from a health-associated biofilm community to one that is pathogenic in nature and a central player in the pathogenic potential of this community is the presence of Porphyromonas gingivalis. This anaerobic bacterium is a natural member of the oral microbiome, yet it can become highly destructive (termed pathobiont) and proliferate to high cell numbers in periodontal lesions, which is attributed to its arsenal of specialized virulence factors. Hence, this organism is regarded as a primary etiologic agent of periodontal disease progression. In this review, we summarize some of the latest information regarding what is known about its role in periodontitis, including pathogenic potential as well as ecological and nutritional parameters that may shift this commensal to a virulent state. We also discuss parallels between the development of pathogenic biofilms and the human cellular communities that lead to cancer, specifically we frame our viewpoint in the context of 'wounds that fail to heal'.
Collapse
Affiliation(s)
- Carla Cugini
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA ; Department of Oral Medicine Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
17
|
Tumor dormancy: long-term survival in a hostile environment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:181-200. [PMID: 23143980 DOI: 10.1007/978-1-4614-1445-2_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor dormancy occurs when cancer cells are present but the tumor does not grow. Following treatment, patients may enter complete remission in which persistent cells represent the minimal residual disease (MRD). Experimental models and clinical data suggest that the absolute quantity of this MRD is extremely low. Very few cancer cells can persist for years or decades under these hostile conditions that include continuous exposure to maintenance treatment, autologous anti-tumor immune response, and a nonpermissive microenvironment. Dormant tumor cells may survive despite these destruction factors if they adapt and develop strategies to escape from cell death. Escape may result in a state of equilibrium between MRD and the patient. Equilibrium between the immune response and tumor cells can result in long-term tumor dormancy; however, after variable lengths of time, tumor dormancy ends, and the disease progresses. Experimental models have shown that dormant tumor cells may over-express B7-H1 and B7.1 and inhibit cytotoxic T-cell mediated lysis. This resistance could be therapeutically targeted using drugs like MEK inhibitors that modulate pathways involved in B7-H1 expression. Dormant tumor cells may also develop nonspecific resistance mechanisms to cell death, such as deregulation of JAK/STAT and mTORC2/AKT pathways or autocrine and paracrine production of cytokines. This deregulation leads to cross-resistance between the immune response and cytotoxic drugs, indicating that the long-term selection that occurs in vivo during tumor dormancy may ultimately result in resistant relapse. Long-term selection of cancer cells in vitro using tyrosine kinase inhibitors selects cells that harbor the same resistance mechanisms as dormant tumor cells. Elucidating the mechanisms underlying the equilibrium that allows for the persistence of dormant tumor cells presents a novel strategy for targeted drug treatment in the context of maintenance therapy.
Collapse
|
18
|
Almog N. Genes and regulatory pathways involved in persistence of dormant micro-tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:3-17. [PMID: 23143972 DOI: 10.1007/978-1-4614-1445-2_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Micro-tumors can remain dormant for prolonged periods of time before they switch and enter the rapid growth phase. This initial stage in tumor progression is clearly understudied. In spite of high prevalence, significant clinical implications and increased interest by the research community, tumor dormancy is still poorly understood. The topic of tumor dormancy also suffers from a lack of definition and an agreed upon terminology to describe it. Additionally, the number of reproducible experimental models available for studying indolence of human micro-tumors is quite limited. Here, we describe the development of a general class of in vivo models of indolent human tumors and how these models can be used to elucidate molecular and cellular mechanisms involved in the regulation of dormancy. The models consist of human tumor cell lines that form microscopic cancerous lesions in mice. Although these lesions contain viable and fully malignant cancer cells, the tumors do not expand in size but remain occult for prolonged periods until they eventually spontaneously switch and become fast-growing tumors. Consistent with Judah Folkman's vision that tumors will remain occult and microscopic until they acquire the ability to recruit new and functional blood vessels, the dormancy period of the micro-tumors is associated with impaired angiogenic capacity. Such models can be used for dissecting the host and the tumor-derived regulatory mechanisms of tumor dormancy. Understanding the process by which dormant tumors can overcome growth constraints and emerge from dormancy, resuming size expansion, may provide insights into novel strategies to prolong the dormancy state or to block tumor formation in the early stages, before they are physically detected or become symptomatic.
Collapse
Affiliation(s)
- Nava Almog
- Tufts University School of Medicine, Boston, MA 02135, USA.
| |
Collapse
|
19
|
Satchi-Fainaro R, Ferber S, Segal E, Ma L, Dixit N, Ijaz A, Hlatky L, Abdollahi A, Almog N. Prospective identification of glioblastoma cells generating dormant tumors. PLoS One 2012; 7:e44395. [PMID: 22970208 PMCID: PMC3435314 DOI: 10.1371/journal.pone.0044395] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 08/03/2012] [Indexed: 01/03/2023] Open
Abstract
Although dormant tumors are highly prevalent within the human population, the underlying mechanisms are still mostly unknown. We have previously identified the consensus gene expression pattern of dormant tumors. Here, we show that this gene expression signature could be used for the isolation and identification of clones which generate dormant tumors. We established single cell-derived clones from the aggressive tumor-generating U-87 MG human glioblastoma cell line. Based only on the expression pattern of genes which were previously shown to be associated with tumor dormancy, we identified clones which generate dormant tumors. We show that very high expression levels of thrombospondin and high expression levels of angiomotin and insulin-like growth factor binding protein 5 (IGFBP5), together with low levels of endothelial specific marker (ESM) 1 and epithelial growth factor receptor (EGFR) characterize the clone which generates dormant U-87 MG derived glioblastomas. These tumors remained indolent both in subcutaneous and orthotopic intracranial sites, in spite of a high prevalence of proliferating cells. We further show that tumor cells which form U-87 MG derived dormant tumors have an impaired angiogenesis potential both in vitro and in vivo and have a slower invasion capacity. This work demonstrates that fast-growing tumors contain tumor cells that when isolated will form dormant tumors and serves as a proof-of-concept for the use of transcriptome profiles in the identification of such cells. Isolating the tumor cells that form dormant tumors will facilitate understanding of the underlying mechanisms of dormant micro-metastases, late recurrence, and changes in rate of tumor progression.
Collapse
Affiliation(s)
- Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shiran Ferber
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ehud Segal
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Lili Ma
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Niharika Dixit
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ambreen Ijaz
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Lynn Hlatky
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Amir Abdollahi
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Department of Radiation Oncology, German Cancer Research Center and University of Heidelberg Medical School, Heidelberg, Germany
| | - Nava Almog
- Center of Cancer Systems Biology, Steward Research & Specialty Projects Corp., St. Elizabeth’s Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
20
|
Tumor Dormancy Resulting from Subcutaneous Injection to SCID Mice with Cultured Nasopharyngeal Carcinoma Cells Is Mediated via IFN-γ Induction of a Highly Differentiated Phenotype. Cancer Biother Radiopharm 2011; 26:417-26. [DOI: 10.1089/cbr.2011.0958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
21
|
Li C, Cao S, Liu Z, Ye X, Chen L, Meng S. RNAi-mediated downregulation of uPAR synergizes with targeting of HER2 through the ERK pathway in breast cancer cells. Int J Cancer 2010; 127:1507-16. [PMID: 20063318 DOI: 10.1002/ijc.25159] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Overexpression of urokinase plasminogen activator receptor (uPAR) or HER2 (erbB-2) in breast cancer is associated with a poor prognosis. We previously reported that gene amplification and overexpression of HER2 and uPAR occur in 70% of HER2-amplified tumor cells from blood or tissue of patients with breast cancer. In this study, we first examined whether depletion of HER2 and uPAR synergized in suppression of the growth of breast cancer cells that overexpress both HER2 and uPAR (SKBR3 and ZR 751). The results showed that depletion of either HER2 or uPAR by RNA interference suppressed cell growth and induced cell apoptosis, but that these effects were significantly enhanced in cells depleted of both HER2 and uPAR. Mechanistic analysis demonstrated that silencing of HER2 and uPAR caused suppression of MAPK signal pathways, resulting in decrease of ERK activity and prompting a high p38/ERK activity ratio. The level of the phosphorylated form of ERK was decreased in cells depleted of HER2, uPAR or both, and the effect in cells depleted of both is the most evident. Moreover, downregulation of uPAR synergized with trastuzumab to suppress the growth and induce apoptosis of SKBR3 and ZR 751 cells. uPAR RNAi significantly enhanced the effect of trastuzumab on inhibition of MAPK signal pathways. In conclusion, targeting HER2 and uPAR has a synergistic inhibitory effect on breast cancer cells. Our results provide evidence that simultaneous downregulation of HER2 and uPAR may offer an effective tool for breast cancer therapy.
Collapse
Affiliation(s)
- Changfei Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
22
|
Berthon C, Driss V, Liu J, Kuranda K, Leleu X, Jouy N, Hetuin D, Quesnel B. In acute myeloid leukemia, B7-H1 (PD-L1) protection of blasts from cytotoxic T cells is induced by TLR ligands and interferon-gamma and can be reversed using MEK inhibitors. Cancer Immunol Immunother 2010; 59:1839-49. [PMID: 20814675 PMCID: PMC2945474 DOI: 10.1007/s00262-010-0909-y] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 08/16/2010] [Indexed: 12/15/2022]
Abstract
B7-H1 (PD-L1) is a B7-related protein that inhibits T-cell responses. B7-H1 participates in the immunoescape of cancer cells and is also involved in the long-term persistence of leukemic cells in a mouse model of leukemia. B7-H1 can be constitutively expressed by cancer cells, but is also induced by various stimuli. Therefore, we examined the constitutive and inducible expression of B7-H1 and the consequences of this expression in human acute myeloid leukemia (AML). We analyzed B7-H1 expression in a cohort of 79 patients with AML. In addition, we studied blast cells after incubation with interferon-gamma or toll-like receptors (TLR) ligands. Finally, we evaluated functionality of cytotoxic T-cell activity against blast cells. Expression of B7-H1 upon diagnosis was high in 18% of patients. Expression of TLR2, 4 and 9 was detected in one-third of AML samples. Expression of TLR2 and TLR4 ligands or IFN-γ induced by B7-H1 was found to protect AML cells from CTL-mediated lysis. Spontaneous B7-H1 expression was also found to be enhanced upon relapse in some patients. MEK inhibitors, including UO126 and AZD6244, reduced B7-H1 expression and restored CTL-mediated lysis of blast cells. In AML, B7-H1 expression by blasts represents a possible immune escape mechanism. The inducibility of B7-H1 expression by IFN-γ or TLR ligands suggests that various stimuli, either produced during the immune response against leukemia cells or released by infectious microorganisms, could protect leukemic cells from T cells. The efficacy of MEK inhibitors against B7-H1-mediated inhibition of CTLs suggests a possible cancer immunotherapy strategy using targeted drugs.
Collapse
Affiliation(s)
- Céline Berthon
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| | - Virginie Driss
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Jizhong Liu
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Klaudia Kuranda
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Xavier Leleu
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| | - Nathalie Jouy
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Dominique Hetuin
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Bruno Quesnel
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| |
Collapse
|
23
|
Lahuerta JJ, Mateos MV, Martínez-López J, Grande C, de la Rubia J, Rosiñol L, Sureda A, García-Laraña J, Díaz-Mediavilla J, Hernández-García MT, Carrera D, Besalduch J, de Arriba F, Oriol A, Escoda L, García-Frade J, Rivas-González C, Alegre A, Bladé J, San Miguel JF. Busulfan 12 mg/kg plus melphalan 140 mg/m2 versus melphalan 200 mg/m2 as conditioning regimens for autologous transplantation in newly diagnosed multiple myeloma patients included in the PETHEMA/GEM2000 study. Haematologica 2010; 95:1913-20. [PMID: 20663944 DOI: 10.3324/haematol.2010.028027] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The aim of this study was to compare the long-term safety and efficacy of oral busulfan 12 mg/kg plus melphalan 140 mg/m(2) and melphalan 200 mg/m(2) as conditioning regimens for autologous stem cell transplantation in newly diagnosed patients with multiple myeloma in the GEM2000 study. DESIGN AND METHODS The first 225 patients received oral busulfan 12 mg/kg plus melphalan 140 mg/m(2); because of a high frequency of veno-occlusive disease, the protocol was amended and a further 542 patients received melphalan 200 mg/m(2). RESULTS Engraftment and hospitalization times were similar in both groups. Oral busulfan 12 mg/kg plus melphalan 140 mg/m(2) resulted in higher transplant-related mortality (8.4% versus 3.5%; P=0.002) due to the increased frequency of veno-occlusive disease in this group. Response rates were similar in both arms. With respective median follow-ups of 72 and 47 months, the median progression-free survival was significantly longer with busulfan plus melphalan (41 versus 31 months; P=0.009), although survival was similar to that in the melphalan 200 mg/m(2) group. However, access to novel agents as salvage therapy after relapse/progression was significantly lower for patients receiving busulfan plus melphalan (43%) than for those receiving melphalan 200 mg/m(2) (58%; P=0.01). CONCLUSIONS Conditioning with oral busulfan 12 mg/kg plus melphalan 140 mg/m(2) was associated with longer progression-free survival but equivalent survival to that achieved with melphalan 200 mg/m(2) but this should be counterbalanced against the higher frequency of veno-occlusive disease-related deaths. This latter fact together with the limited access to novel salvage therapies in patients conditioned with oral busulfan 12 mg/kg plus melphalan 140 mg/m(2) may explain the absence of a survival difference. Oral busulfan was used in the present study; use of the intravenous formulation may reduce toxicity and result in greater efficacy, and warrants further investigation in myeloma patients. (Clinicaltrials.gov identifier: NCT00560053).
Collapse
|
24
|
Molecular mechanisms underlying tumor dormancy. Cancer Lett 2010; 294:139-46. [PMID: 20363069 DOI: 10.1016/j.canlet.2010.03.004] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 03/01/2010] [Accepted: 03/04/2010] [Indexed: 12/15/2022]
Abstract
Evidence suggests that dormant, microscopic tumors are not only common, but are highly prevalent in otherwise healthy individuals. Due to their small size and non-invasive nature, these dormant tumors remain asymptomatic and, in most cases, undetected. With advances in diagnostic imaging and molecular biology, it is now becoming clear that such neoplasms can remain in an asymptomatic, dormant stage for considerable periods of time without expanding in size. Although a number of processes may play a role in thwarting the expansion of microscopic tumors, one critical mechanism behind tumor dormancy is the ability of the tumor population to induce angiogenesis. Although cancer can arise through multiple pathways, it is assumed that essentially most tumors begin as microscopic, non-angiogenic neoplasms which cannot expand in size until vasculature is established. It is now becoming clear that cancer does not progress through a continuous exponential growth and mass expansion. Clinical cancer is usually manifested only in late, unavoidably symptomatic stages of the disease when tumors are sufficiently large to be readily detected. While dormancy in primary tumors is best defined as the time between the carcinogenic transformation event and the onset of inexorable progressive growth, it can also occur as minimal residual or occult disease from treated tumors or as micro-metastases. The existence of dormant tumors has important implications for the early detection and treatment of cancer. Elucidating the regulatory machinery of these processes will be instrumental in identifying novel early cancer biomarkers and could provide a rationale for the development of dormancy-promoting tumor therapies. Despite the high prevalence of microscopic, dormant tumors in humans and the significant clinical implications of their early detection, this area in cancer research has, to date, been under-investigated. In this mini review observations, models and experimental approaches to study tumor dormancy are summarized. Additionally, analogies and distinctions between the concepts of "tumor dormancy" and that of the "cellular dormancy" of tumor cells, as well as between the "exit from tumor dormancy" and the "onset of the angiogenic switch" are discussed.
Collapse
|
25
|
Eftimie R, Bramson JL, Earn DJD. Interactions between the immune system and cancer: a brief review of non-spatial mathematical models. Bull Math Biol 2010; 73:2-32. [PMID: 20225137 DOI: 10.1007/s11538-010-9526-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 02/18/2010] [Indexed: 12/14/2022]
Abstract
We briefly review spatially homogeneous mechanistic mathematical models describing the interactions between a malignant tumor and the immune system. We begin with the simplest (single equation) models for tumor growth and proceed to consider greater immunological detail (and correspondingly more equations) in steps. This approach allows us to clarify the necessity for expanding the complexity of models in order to capture the biological mechanisms we wish to understand. We conclude by discussing some unsolved problems in the mathematical modeling of cancer-immune system interactions.
Collapse
Affiliation(s)
- Raluca Eftimie
- Department of Mathematics and Statistic, McMaster University, Hamilton, ON, Canada, L8S 4K1.
| | | | | |
Collapse
|
26
|
Abstract
The role of the immune system in tumor dormancy is now well established. In several experimental models it is possible to induce tumor dormancy in immunocompetent hosts by prior immunization against tumor cells. Equilibrium between immune response and tumor cells leads to long-term tumor dormancy. This equilibrium is also observed early in tumor development and adaptive immunity may help contain tumor outgrowth. However, after variable times, tumor dormancy ends and the disease progresses. As the immune response remains active the tumor cells presumably escape dormancy by becoming resistant. Due to the extreme difficulty of isolating dormant tumor cells from patients, such mechanisms are poorly understood. However, experimental models have shown that dormant tumor cells may overexpress B7-H1 and B7.1, and inhibit CTL-mediated lysis. These cells resist apoptosis by methylating SOCS1, and by paracrine production of cytokines. The presence of immunoescape mechanisms in tumor cells from relapsing patients also suggests that the immune equilibrium which maintained dormancy has broken down. Identification of such mechanisms would offer new leads to favor the immune balance, and thus to clear minimal residual disease from patients.
Collapse
Affiliation(s)
- Bruno Quesnel
- INSERM, U837, E3, Institut de Recherche sur le Cancer de Lille, France.
| |
Collapse
|