Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1906 2023-05-05 18:49:34 |
2 format correct + 10 word(s) 1916 2023-05-06 04:55:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Monaco, M.L.; Idris, O.A.; Essani, K. Basic Biology of Triple-Negative Breast Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/43892 (accessed on 02 July 2024).
Monaco ML, Idris OA, Essani K. Basic Biology of Triple-Negative Breast Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/43892. Accessed July 02, 2024.
Monaco, Michael L., Omer A. Idris, Karim Essani. "Basic Biology of Triple-Negative Breast Cancer" Encyclopedia, https://encyclopedia.pub/entry/43892 (accessed July 02, 2024).
Monaco, M.L., Idris, O.A., & Essani, K. (2023, May 05). Basic Biology of Triple-Negative Breast Cancer. In Encyclopedia. https://encyclopedia.pub/entry/43892
Monaco, Michael L., et al. "Basic Biology of Triple-Negative Breast Cancer." Encyclopedia. Web. 05 May, 2023.
Basic Biology of Triple-Negative Breast Cancer
Edit

Triple-negative breast cancer (TNBC) is an aggressive as well as the most dangerous form of breast cancer. Due to the lack of biomarkers that can be targeted by specific molecular therapeutics, treatment is usually limited to chemotherapy and surgery where applicable. 

triple-negative breast cancer metastasis cancer biology standard of care pathogenesis of cancer

1. Triple-Negative Breast Cancer (TNBC)

Breast cancer (BC) is the most diagnosed cancer in the United States for women, and is also the number two killer among cancers in women [1][2]. The biggest challenge with BC treatment lies in the heterogeneity of the tumors. There are three different receptors that define different BC subtypes: estrogen receptors (ERs, inside cells), progesterone receptors (PRs, inside cells), and human epidermal growth factor receptor 2 (HER-2, also named CD340, a cell surface receptor) [3][4]. Of all of the new cases of BC each year, approximately 10–20% are negative for all 3 receptors and are categorized as triple-negative breast cancer (TNBC) [5][6]; however, within TNBC itself, there are several different subtypes based on differential gene expression. These types are as follows: basal-like (BL1 and BL2), mesenchymal (M), mesenchymal-stem-cell-like (MSL), immunomodulatory (IM), and luminal androgen receptor (LAR) [7][8][9][10].
The BL1 and BL2 subtypes differ in that BL1 has elevated gene expression for DNA damage response, proliferation signaling, and cell cycle checkpoint loss, whereas BL2 has elevated growth factor (GF) signaling, glycolysis/gluconeogenesis signaling, and myoepithelial surface receptors. This suggests that cells of the BL1/2 subtypes may be of basal or myoepithelial origin. The M subtype has elevated expression of cell motility pathways, receptor interaction with the extracellular matrix (ECM), and cell differentiation pathways. The MSL subtype shares expression with the M subtype, but it also uniquely expresses pathways related to GF signaling (inositol phosphate metabolism, platelet-derived growth factor (PDGF), epidermal growth factor receptor (EGFR), calcium (Ca2+), G-protein-coupled receptor (GPCR), and extracellular-related kinase (ERK 1 and 2)), epithelial–mesenchymal transition (EMT), and Wnt/β-catenin. The MSL subtype also has a reduced expression of genes for cell proliferation. The IM subtype has an elevated expression of pathways involving immune cell signaling, cytokine signaling, antigen processing/presentation, and immune signal transduction. Finally, the LAR subtype has a heavily elevated expression of pathways related to hormone regulation, particularly with androgen receptor signaling (which can be approximately nine times as much as that of other TNBC subtypes) [8]. These extremely diverse genetic regulation conditions define tumor heterogeneity within TNBC, which helps to explain why TNBC is extremely difficult to treat in comparison with other BCs [8][11].

2. Development of Heterogeneity in Triple-Negative Breast Cancer

There have been a few theories to describe how heterogeneity in TNBC is established and maintained. The two main concepts are the cancer stem cell (CSC) hypothesis and the clonal evolution model (CEM). Other concepts, such as the deregulation of adult mammary stem cells (aMSCs), are described as contributing to developing TNBC in its initial stages [12][13][14][15].
The division of aMSCs gives rise to progenitors that further differentiate into basal or luminal progenitor cells. Under normal conditions, if basal or luminal progenitor cells are made, they further develop into mature basal or luminal cells; however, when the development of luminal progenitors is disrupted, cancer cells may develop. This is believed to occur via the improper regulation of Wnt/β-catenin pathways, transcription factors (TFs), such as Snail, and embryonic stem cell (ESC) markers (Sox2, Nanog, and Oct4). Combinations of genetic alterations to these pathways can make the progenitor cells de-differentiate and return to a proliferative state, which, when combined with other mutations, may result in TNBC [15].
The CSC model proposes that there is a hierarchy within cancers of tumorigenic cancer cells and their non-tumorigenic progeny. Within these subpopulations, the CSCs are hypothesized to be the driving forces behind the growth of a tumor, its progression, resistance to certain treatments, and metastasis [12][16][17][18][19]. This model does not address how to account for the massive variability in tumor heterogeneity between cancers of the same type in different individuals. Early characterizations of tumorigenic CSCs in BCs were via the expression of CD44 and not CD24 (including the opposite expression pattern); however, these characterizations have not held because many subtypes do not follow these marker patterns [12]. Moreover, the alteration of aldehyde dehydrogenase 1 (ALDH1) expression has also been independently reported to have tumorigenic potential in BCs [20].
The CEM presumes that there is one original clone that becomes cancerous, which then divides and, as carcinogenesis progresses, the cell as well as its subsequent progeny accumulate mutations in each cycle of clonal expansion, which results in the heterogeneity of the tumor. This model also assumes that there is no hierarchy among the cells and that, instead, natural selection determines which clones are successful and continue to divide. The cells that cannot undergo division are maintained as part of the tumor or will senesce, eventually being eliminated via necrosis [13][14]. Tumor heterogeneity may originate via some combination of the two models and several additional variables, including environmental factors, genetic background, and immune status. Hence, additional studies are needed to define the role of these contributing factors in individual cancers in order to formulate a more comprehensive model in the future.

3. Triple-Negative Breast Cancer Pathogenesis

Among the approximately 268,600 new cases of BC in the United States in 2019, about 10–20% of these were TNBC [21]. Estimates for 2022 suggested 287,850 new cases of BC in women of the United States, resulting in approximately 43,250 deaths [1]. TNBC, as a whole, is more aggressive, has more p53 mutations, has higher mitotic indices, has less defined nuclear pleomorphisms, has a higher average grade, and demonstrates higher rates of proliferation than typical BCs [22]. In addition to the above, TNBC has a high risk for relapsing in a patient following therapy, which factors into its high mortality compared to other BCs [23].
In the development and progression of carcinogenesis, epithelial–mesenchymal transition (EMT) has been repeatedly shown to be important in changing cell profile, metastasis, and even tumor recurrence [24][25][26]. Two genes, SLUG (a transcriptional repressor and regulator) and SOX10 (a TF), have been shown to have increased and preferential expression within TNBC. It has been shown that, when ALDH1 is simultaneously expressed with SLUG, there is an association with shorter disease-free survival rates and the conversion from EMT into mesenchymal–epithelial transition (MET) in metastatic sites. MET is the reverse of EMT, and it is denoted by the reacquisition of epithelial characteristics [27][28]. Of the TNBC subtypes, M and MSL have been linked to metaplastic BCs expressing some of these characteristics [29].
Another factor that contributes to the difficulty in treating TNBCs is the high degree of variance among the mutations acquired. The most common mutations are to p53 (up to 80%), followed by phosphatase and tensin homolog (PTEN) (35%) as well as inositol-polyphosphate-4-phosphatase type 2 (INPP4B, 30%) [30][31][32]. So far, none of the mutations described in TNBCs have been determined to be the “driver” of carcinogenesis [33]. The BRCA1 and BRCA2 genes have been shown to contribute to carcinogenesis particularly in BCs with a familial linkage (15–20%) or in those who are already carriers of one mutation in breast tissue [34][35][36]. Other risk factors are numerous, and lifestyle habits, such as diet and obesity, can increase the likelihood of developing TNBC [37]. During metastasis, TNBC has been documented to go to the lungs, liver, and brain more often than bone, which is the typical site for other BCs [38]. In a separate study, it was observed that, of 116 patients diagnosed with TNBC between 2000 and 2006 at the Dana–Farber Cancer Institute, 46% of the patients were diagnosed with metastases to parts of the central nervous system (CNS), and the median survival rate following prognosis was 4.6 months [39].

4. Triple-Negative Breast Cancer Ecology

Neoplastic cells must compete for resources with nearby healthy cells; however, these neoplastic clones also often compete amongst themselves. This is because different mutations acquired in these neoplasms impact replication and resource acquisition rates. It has been shown that neoplastic TNBC cell clones injected into opposite flanks of mice and rats can inhibit the growth of one another or that one set of clones can completely inhibit the other [40][41]. Neoplastic cells have also been described as parasites working towards establishing independence [14]. It has been suggested that the stimulation of fibroblasts and macrophages towards angiogenesis and the release of growth factors could aid TNBC clones in establishing tumors. The release of elastases and matrix metalloproteinases (MMPs) by macrophages for ECM degradation has also been suggested to be a contributing factor in establishing tumor beds and metastasis [42][43]. Additionally, it has been suggested that neoplastic TNBC clones participate in a commensal interaction with nearby non-metastatic clones where diffusible factors are absorbed, allowing them to take up a metastatic phenotype without needing to accumulate genetic mutations first [40][44][45][46].
Furthermore, data acquired from studying cocultures of different BC cell lines in vitro show that certain interactions between cells can impact growth, both positively and negatively. MDA-MB-231 cells coincubated with supernatants of MCF-7 cells increased proliferation over time, whereas coincubation with BT-474 cells or simply other MDA-MB-231 cells hampered growth [47]. Potential explanations for these observations were hypothesized as distant cell interactions where cell culture medium differences created competitively favorable environmental conditions or that ER/PR cells can utilize auto and paracrine self-regulation pathways to synthesize their own GFs more frequently than BC cells that express ER and/or PR [48]. Therefore, these cells could independently sustain their growth without an autocrine loop. Neoplasms also have important interactions with the TME in supporting the growth of a developing tumor. These interactions are complex as differing TME conditions have varied impacts on carcinogenesis; normal epithelial cells can become carcinomas [49] and teratocarcinomas can have their phenotype suppressed [50] or even potentially reverted [51] based on conditions present in the TME.

5. Current Standard Therapies for Triple-Negative Breast Cancer

Due to the extreme heterogeneity of TNBC, chemotherapy with anthracyclines or taxanes remains the standard for the treatment of TNBC. There are a few mechanistic targets for therapy such as DNA repair, cell proliferation, and p53 [52]. Patients with TNBC have been documented to receive significant benefit from and even respond better to chemotherapy than other BC types do, despite the more aggressive phenotypes [53][54][55]. Ultimately, if a patient’s TNBC becomes metastatic, particularly during a recurrence, survival falls to 0%; even combinatorial therapies that trade increased toxicity for potentially elevated responses result in no benefit to survival rates [56][57].
However, in March 2019, the US FDA granted accelerated approval for an immunotherapy regimen for patients with BC. The treatment includes the use of atezolizumab (Tecentriq®) combined with nanoparticle albumin-bound paclitaxel (Abraxane®, nab-paclitaxel) in patients with metastatic TNBC where ≥1% of the tumor area is positive for immune cells expressing programmed cell death-ligand 1 (PD-L1) [58][59]. Atezolizumab is a humanized IgG1 monoclonal antibody that binds directly to PD-L1 and blocks a receptor’s interaction with PD-1 as well as the costimulatory protein B7-1 [60]. Nab-paclitaxel is a microtubule inhibitor that, when bound to nanoparticle albumin, increases solubility, minimizes hypersensitivity reactions, has better transport across endothelial tissue, greater tissue penetration, and a slower elimination time as compared to unbound paclitaxel [61][62][63][64][65][66]. Results from a double-blind, randomized, and placebo phase III clinical trial lead to this regimen’s accelerated approval [60]. When the combination of atezolizumab and nab-paclitaxel was compared with the placebo plus nab-paclitaxel for the primary treatment group, patients had an average increased progression-free survival (PFS) of 1.7 months and a 20% reduction in risk for disease progression and/or death. In the PD-L1-positive subgroup, the PFS increased further to 2.5 months as well as further decreased the risk for disease progression and/or death to 38% [60]. Even though this new therapy shows promise to increase the lifespan of patients slightly, this may merely delay the inevitable in most cases; therefore, new targeted therapies are still needed for TNBC.

References

  1. Giaquinto, A.N.; Sung, H.; Miller, K.D.; Kramer, J.L.; Newman, L.A.; Minihan, A.; Jemal, A.; Siegel, R.L. Breast Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 524–541.
  2. American Cancer Society. Cancer Facts and Figures 2022; American Cancer Society: Atlanta, GA, USA, 2022.
  3. Polyak, K. Review series introduction Heterogeneity in breast cancer. J. Clin. Investig. 2011, 121, 2011–2013.
  4. Neophytou, C.; Boutsikos, P.; Papageorgis, P. Molecular Mechanisms and Emerging Therapeutic Targets of Triple-Negative Breast Cancer Metastasis. Front. Oncol. 2018, 8, 31.
  5. Slamon, D.J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; et al. Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2. N. Engl. J. Med. 2001, 344, 783–792.
  6. Anders, C.K.; Carey, L. A Biology, Metastatic Patterns and Treatment of Patients with Triple-Negtive Breast Cancer. Clin. Breast Cancer 2010, 9, S73–S81.
  7. Lehmann, B.D.; Jovanović, B.; Chen, X.; Estrada, M.V.; Johnson, K.N.; Shyr, Y.; Moses, H.L.; Sanders, M.E.; Pietenpol, J.A. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368.
  8. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. 2011, 121, 2750.
  9. Chen, X.; Li, J.; Gray, W.H.; Lehmann, B.D.; Bauer, J.A.; Shyr, Y.; Pietenpol, J.A. TNBC type: A subtyping tool for triple-negative breast cancer. Cancer Inform. 2012, 11, 147–156.
  10. Hatzis, C. A Genomic Predictor of Response and Survival Following Taxane-Anthracycline Chemotherapy for Invasive Breast Cancer. JAMA 2011, 305, 1873.
  11. Bertucci, F.; Finetti, P.; Cervera, N.; Charafe-Jauffret, E.; Mamessier, E.; Adélaïde, J.; Debono, S.; Houvenaeghel, G.; Maraninchi, D.; Viens, P.; et al. Gene expression profiling shows medullary breast cancer is a subgroup of basal breast cancers. Cancer Res. 2006, 66, 4636–4644.
  12. Corbin, M.; Morrison, S. Tumor heterogeneity and cancer cell plasticity. Nature 2013, 501, 328–337.
  13. Marusyk, A.; Polyak, K. Tumor heterogeneity: Causes and consequences. Biochim. Biophys. Acta 2011, 1805, 105–117.
  14. Merlo, L.M.F.; Pepper, J.W.; Reid, B.J.; Maley, C.C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 2006, 6, 924–935.
  15. Rangel, M.C.; Bertolette, D.; Castro, N.P.; Klauzinska, M.; Cuttitta, F.; Salomon, D.S. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Res. Treat. 2016, 156, 211–226.
  16. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760.
  17. Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Jo, M.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of Reactive Oxygen Species Levels and Radioresistance in Cancer Stem Cells. Nature 2009, 458, 780–783.
  18. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell 2008, 133, 704–715.
  19. Balic, M.; Lin, H.; Young, L.; Hawes, D.; Giuliano, A.; McNamara, G.; Datar, R.H.; Cote, R.J. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin. Cancer Res. 2006, 12, 5615–5621.
  20. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567.
  21. American Cancer Society. Breast Cancer Facts and Figure 2019 and Figure 2020; American Cancer Society: Atlanta, GA, USA, 2019; pp. 1–44.
  22. Eberlein, T.J. Race, Breast Cancer Subtypes, and Survival in the Carolina Breast Cancer Study. Yearb. Surg. 2007, 2007, 304–305.
  23. Bauer, K.R.; Brown, M.; Cress, R.D.; Parise, C.A.; Caggiano, V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: A population-based study from the California Cancer Registry. Cancer 2007, 109, 1721–1728.
  24. Arias, A.M. Epithelial mesenchymal interactions in cancer and development. Cell 2001, 105, 425–431.
  25. Soady, K.; Smalley, M.J. Slugging their way to immortality: Driving mammary epithelial cells into a stem cell-like state. Breast Cancer Res. 2012, 14, 319.
  26. Guo, W.; Keckesova, Z.; Donaher, J.L.; Shibue, T.; Tischler, V.; Reinhardt, F.; Itzkovitz, S.; Noske, A.; Zürrer-Härdi, U.; Bell, G.; et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell 2012, 148, 1015–1028.
  27. Pomp, V.; Leo, C.; Mauracher, A.; Korol, D.; Guo, W.; Varga, Z. Differential expression of epithelial–mesenchymal transition and stem cell markers in intrinsic subtypes of breast cancer. Breast Cancer Res. Treat. 2015, 154, 45–55.
  28. Ito, M.; Shien, T.; Omori, M.; Mizoo, T. Evaluation of aldehyde dehydrogenase 1 and transcription factors in both primary breast cancer and axillary lymph node metastases as a prognostic factor. Breast Cancer 2015, 23, 2–5.
  29. Zhou, S.; Sun, X.; Yu, L.; Zhou, R.; Li, A.; Li, M.; Yang, W. Differential expression and clinical significance of epithelial-mesenchymal transition markers among different histological types of triple-negative breast cancer. J. Cancer 2018, 9, 604–613.
  30. Shah Sohrab, P.; Roth Andrew, G.; Rodrigo, O.; Arusha, H.G.; Zhao, Y.; Turashvili, G.; Ding, J.; Tse, K.; Haffari, G.; Bashashati, A.; et al. The clonal and mutational evolution spectrum of primary triple negative breast cancers. Nature 2013, 486, 395–399.
  31. Curtis, C.; Shah, S.P.; Chin, S.-F.; Turashvili, G.; Rueda, O.M.; Dunning, M.J.; Speed, D.; Lynch, A.G.; Samarajiwa, S.; Yuan, Y.; et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 2012, 486, 346–352.
  32. Kalimutho, M.; Parsons, K.; Mittal, D.; López, J.A.; Srihari, S.; Khanna, K.K. Targeted Therapies for Triple-Negative Breast Cancer: Combating a Stubborn Disease; Elsevier Current Trends: Amsterdam, The Netherlands, 2015; Volume 36, pp. 822–846.
  33. Munzone, E.; Gray, K.P.; Fumagalli, C.; Guerini-Rocco, E.; Láng, I.; Ruhstaller, T.; Gianni, L.; Kammler, R.; Viale, G.L.; Leo, A.D.; et al. Mutational analysis of triple-negative breast cancers within the International Breast Cancer Study Group (IBCSG) Trial 22-00. Breast Cancer Res. Treat. 2018, 170, 351–360.
  34. Tung, N.; Lin, N.U.; Kidd, J.; Allen, B.A.; Singh, N.; Wenstrup, R.J.; Hartman, A.R.; Winer, E.P.; Garber, J.E. Frequency of germline mutations in 25 cancer susceptibility genes in a sequential series of patients with breast cancer. J. Clin. Oncol. 2016, 34, 1460–1468.
  35. Turnbull, C.; Rahman, N. Genetic Predisposition to Breast Cancer: Past, Present, and Future. Annu. Rev. Genom. Hum. Genet. 2008, 9, 321–345.
  36. Kuchenbaecker, K.B.; Hopper, J.L.; Barnes, D.R.; Phillips, K.-A.; Mooij, T.M.; Roos-Blom, M.-J.; Jervis, S.; van Leeuwen, F.E.; Milne, R.L.; Andrieu, N.; et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017, 317, 2402.
  37. American Cancer Society. Cancer Facts & Figures 2017. 2017. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2017.html (accessed on 15 April 2018).
  38. Heitz, F.; Harter, P.; Traut, A.; Lueck, H.J.; Beutel, B.; du Bois, A. Cerebral metastases (CM) in breast cancer (BC) with focus on triple-negative tumors. J. Clin. Oncol. 2008, 26, 1010.
  39. Lin, N.U.; Claus, E.; Sohl, J.; Razzak, A.R.; Arnaout, A.; Winer, E.P. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer. Cancer 2008, 113, 2638–2645.
  40. Miller, B.E.; Miller, F.R.; Leith, J.; Heppner, G.H. Growth Interaction in Vivo between Tumor Subpopulations Derived from a Single Mouse Mammary Tumor Growth Interaction in Vivo between Tumor Subpopulations Derived from a Single Mouse Mammary Tumor1. Cancer Res. 1980, 40, 3977–3981.
  41. Caignard, A.; Martin, M.S.; Michel, M.F.; Martin, F. Interaction between two cellular subpopulations of a rat colonic carcinoma when inoculated to the syngeneic host. Int. J. Cancer 1985, 36, 273–279.
  42. Rundhaug, J.E. Matrix metalloproteinases and angiogenesis. J. Cell Mol. Med. 2007, 9, 267–285.
  43. Nagy, J.D. Competition and natural selection in a mathematical model of cancer. Bull. Math. Biol. 2004, 66, 663–687.
  44. Jouanneau, J.; Moens, G.; Bourgeois, Y.; Poupon, M.F.; Thiery, J.P. A minority of carcinoma cells producing acidic fibroblast growth factor induces a community effect for tumor progression. Proc. Natl. Acad. Sci. USA 1994, 91, 286–290.
  45. Axelrod, R.; Axelrod, D.E.; Pienta, K.J. Evolution of cooperation among tumor cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13474–13479.
  46. Heppner, G.H.; Miller, F.R. The Cellular Basis of Tumor Progression. In International Review of Cytology; Jeon, K.W., Ed.; International Review of Cytology; Academic Press: Cambridge, MA, USA, 1997; Volume 177, pp. 1–56.
  47. Bychkov, V.A.; Pevzner, A.M.; Nebova, J.A.; Ermakova, N.N.; Ibragimova, M.K.; Tsyganov, M.M.; Lyapunova, L.S.; Litviakov, N.V. In vitro modeling of tumor interclonal interactions using breast cancer cell lines. Exp. Oncol. 2021, 43, 118–124.
  48. Ethier, S.P. Growth factor synthesis and human breast cancer progression. J. Natl. Cancer Inst. 1995, 87, 964–973.
  49. Kuperwasser, C.; Chavarria, T.; Wu, M.; Magrane, G.; Gray, J.W.; Carey, L.; Richardson, A.; Weinberg, R.A. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc. Natl. Acad. Sci. USA 2004, 101, 4966–4971.
  50. Mintz, B.; Illmensee, K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc. Natl. Acad. Sci. USA 1975, 72, 3585–3589.
  51. Kenny, P.A.; Bissell, M.J. Tumor reversion: Correction of malignant behavior by microenvironmental cues. Int. J. Cancer 2003, 107, 688–695.
  52. Wahba, H.A.; El-hadaad, H.A. Current approaches in treatment of triple-negative breast cancer Treatment modalities of TNBC. Cancer Biol. Med. 2015, 12, 106–116.
  53. Albain, K.; Anderson, S.; Arriagada, R.; Barlow, W.; Bergh, J.; Bliss, J.; Wood, W. Comparisons between different polychemotherapy regimens for early breast cancer: Meta-analyses of long-term outcome among 100 000 women in 123 randomised trials. Lancet 2012, 379, 432–444.
  54. Cortazar, P.; Zhang, L.; Untch, M.; Mehta, K.; Costantino, J.P.; Wolmark, N.; Bonnefoi, H.; Cameron, D.; Gianni, L.; Valagussa, P.; et al. Pathological complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014, 384, 164–172.
  55. Carey, L.A.; Dees, E.C.; Sawyer, L.; Gatti, L.; Moore, D.T.; Collichio, F.; Ollila, D.W.; Sartor, C.I.; Graham, M.L.; Perou, C.M. The triple negative paradox: Primary tumor chemosensitivity of breast cancer subtypes. Clin. Cancer Res. 2007, 13, 2329–2334.
  56. Bonotto, M.; Gerratana, L.; Poletto, E.; Driol, P.; Giangreco, M.; Russo, S.; Minisini, A.M.; Andreetta, C.; Mansutti, M.; Pisa, F.E.; et al. Measures of Outcome in Metastatic Breast Cancer: Insights from a Real-World Scenario. Oncologist 2014, 19, 608–615.
  57. Conlin, A.K.; Seidman, A.D. Taxanes in Breast Cancer: An Update Corresponding author. Curr. Oncol. Rep. 2007, 9, 22–30.
  58. US FDA Website. FDA Approves Atezolizumab for PD-L1 Positive Unresectable Locally Advanced or Metastatic Triple-Negative Breast Cancer. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-pd-l1-positive-unresectable-locally-advanced-or-metastatic-triple-negative (accessed on 12 December 2022).
  59. Kang, C.; Syed, Y.Y. Atezolizumab (in Combination with Nab-Paclitaxel): A Review in Advanced Triple-Negative Breast Cancer. Drugs 2020, 80, 601–607.
  60. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Dieras, V.; Hegg, R.; Im, S.A.; Shaw Wright, G.; et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 2018, 379, 2108–2121.
  61. Ibrahim, N.K.; Desai, N.; Legha, S.; Soon-Shiong, P.; Theriault, R.L.; Rivera, E.; Esmaeli, B.; Ring, S.E.; Bedikian, A.; Hortobagyi, G.N.; et al. Phase I and pharmacokinetic study of ABI-007, a Cremophor-free, protein-stabilized, nanoparticle formulation of paclitaxel. Clin. Cancer Res. 2002, 8, 1038–1044.
  62. Gradishar, W.J.; Tjulandin, S.; Davidson, N.; Shaw, H.; Desai, N.; Bhar, P.; Hawkins, M.; O’Shaughnessy, J. Phase III Trial of Nanoparticle Albumin-Bound Paclitaxel Compared with Polyethylated Castor Oil–Based Paclitaxel in Women With Breast Cancer. J. Clin. Oncol. 2005, 23, 7794–7803.
  63. Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.; Beals, B.; Dykes, D.; et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res. 2006, 12, 1317–1324.
  64. Chen, N.; Li, Y.; Ye, Y.; Palmisano, M.; Chopra, R.; Zhou, S. Pharmacokinetics and pharmacodynamics of nab-paclitaxel in patients with solid tumors: Disposition kinetics and pharmacology distinct from solvent-based paclitaxel. J. Clin. Pharmacol. 2014, 54, 1097–1107.
  65. Gardner, E.R.; Dahut, W.L.; Scripture, C.D.; Jones, J.; Aragon-Ching, J.B.; Desai, N.; Hawkins, M.J.; Sparreboom, A.; Figg, W.D. Randomized crossover pharmacokinetic study of solvent-based paclitaxel and nab-paclitaxel. Clin. Cancer Res. 2008, 14, 4200–4205.
  66. Brufsky, A. Nab-Paclitaxel for the treatment of breast cancer: An update across treatment settings. Exp. Hematol. Oncol. 2017, 6, 7.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 305
Revisions: 2 times (View History)
Update Date: 06 May 2023
1000/1000
Video Production Service