1
|
Corti C, Koca B, Rahman T, Mittendorf EA, Tolaney SM. Recent Advances in Immune Checkpoint Inhibitors for Triple-Negative Breast Cancer. Immunotargets Ther 2025; 14:339-357. [PMID: 40196378 PMCID: PMC11974553 DOI: 10.2147/itt.s495751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
While immunotherapy has transformed treatment across various cancers, its impact on breast cancer is relatively limited. Recent advances have established immunotherapy as an effective approach for triple-negative breast cancer (TNBC), an aggressive subtype with limited therapeutic targets and poor prognosis. Specifically, pembrolizumab, an immune checkpoint inhibitor (ICI), is now approved for both first-line metastatic and early-stage TNBC. In metastatic TNBC, combining ICIs with chemotherapy, particularly pembrolizumab, has demonstrated survival benefits in patients with PD-L1-positive disease. However, extending these benefits to broader populations has proven challenging, highlighting the need for better patient selection and novel strategies. Emerging approaches include combining ICIs with antibody-drug conjugates, PARP inhibitors, dual ICIs, and bispecific antibodies targeting angiogenesis and immune checkpoints. These strategies aim to overcome resistance and expand immunotherapy's efficacy beyond the PD-1/PD-L1 pathway. In early-stage disease, pembrolizumab combined with chemotherapy in the neoadjuvant setting has significantly improved pathologic complete response, event-free survival, and overall survival, establishing a new standard of care. Ongoing research aims to determine the optimal timing for ICI administration, explore less toxic chemotherapy backbones, utilize biomarkers for personalized treatment, and assess whether adding complementary treatments, such as radiation therapy for high-risk cases, can improve outcomes. This review examines the successes and setbacks of ICI use in TNBC, offering a comprehensive overview of current practices and future directions. It emphasizes optimizing ICI timing, leveraging biomarkers, and integrating novel agents to refine treatment approaches for both metastatic and early-stage TNBC. As immunotherapy continues to evolve, future research must address the unmet needs of this challenging breast cancer subtype, offering hope for improved outcomes.
Collapse
Affiliation(s)
- Chiara Corti
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Beyza Koca
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tasnim Rahman
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Breast Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
2
|
Corti C, Binboğa Kurt B, Koca B, Rahman T, Conforti F, Pala L, Bianchini G, Criscitiello C, Curigliano G, Garrido-Castro AC, Kabraji SK, Waks AG, Mittendorf EA, Tolaney SM. Estrogen Signaling in Early-Stage Breast Cancer: Impact on Neoadjuvant Chemotherapy and Immunotherapy. Cancer Treat Rev 2025; 132:102852. [PMID: 39571402 DOI: 10.1016/j.ctrv.2024.102852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 01/01/2025]
Abstract
Neoadjuvant chemoimmunotherapy (NACIT) has been shown to improve pathologic complete response (pCR) rates and survival outcomes in stage II-III triple-negative breast cancer (TNBC). Promising pCR rate improvements have also been documented for selected patients with estrogen receptor-positive (ER+) human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC). However, one size does not fit all and predicting which patients will benefit from NACIT remains challenging. Accurate predictions would be useful to minimize immune-related toxicity, which can be severe, irreversible, and potentially impact fertility and quality of life, and to identify patients in need of alternative treatments. This review aims to capitalize on the existing translational and clinical evidence on predictors of treatment response in patients with early-stage BC treated with neoadjuvant chemotherapy (NACT) and NACIT. It summarizes evidence suggesting that NACT/NACIT effectiveness may correlate with pre-treatment tumor characteristics, including mutational profiles, ER expression and signaling, immune cell presence and spatial organization, specific gene signatures, and the levels of proliferating versus quiescent cancer cells. However, the predominantly qualitative and descriptive nature of many studies highlights the challenges in integrating various potential response determinants into a validated, comprehensive, and multimodal predictive model. The potential of novel multi-modal approaches, such as those based on artificial intelligence, to overcome current challenges remains unclear, as these tools are not free from bias and shortcut learning. Despite these limitations, the rapid evolution of these technologies, coupled with further efforts in basic and translational research, holds promise for improving treatment outcome predictions in early HER2- BC.
Collapse
Affiliation(s)
- Chiara Corti
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Busem Binboğa Kurt
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Beyza Koca
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Tasnim Rahman
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Fabio Conforti
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Laura Pala
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, IRCCS, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Ana C Garrido-Castro
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sheheryar K Kabraji
- Department of Medicine, Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adrienne G Waks
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Maleki EH, Bahrami AR, Matin MM. Cancer cell cycle heterogeneity as a critical determinant of therapeutic resistance. Genes Dis 2024; 11:189-204. [PMID: 37588236 PMCID: PMC10425754 DOI: 10.1016/j.gendis.2022.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
Intra-tumor heterogeneity is now arguably one of the most-studied topics in tumor biology, as it represents a major obstacle to effective cancer treatment. Since tumor cells are highly diverse at genetic, epigenetic, and phenotypic levels, intra-tumor heterogeneity can be assumed as an important contributing factor to the nullification of chemotherapeutic effects, and recurrence of the tumor. Based on the role of heterogeneous subpopulations of cancer cells with varying cell-cycle dynamics and behavior during cancer progression and treatment; herein, we aim to establish a comprehensive definition for adaptation of neoplastic cells against therapy. We discuss two parallel and yet distinct subpopulations of tumor cells that play pivotal roles in reducing the effects of chemotherapy: "resistant" and "tolerant" populations. Furthermore, this review also highlights the impact of the quiescent phase of the cell cycle as a survival mechanism for cancer cells. Beyond understanding the mechanisms underlying the quiescence, it provides an insightful perspective on cancer stem cells (CSCs) and their dual and intertwined functions based on their cell cycle state in response to treatment. Moreover, CSCs, epithelial-mesenchymal transformed cells, circulating tumor cells (CTCs), and disseminated tumor cells (DTCs), which are mostly in a quiescent state of the cell cycle are proved to have multiple biological links and can be implicated in our viewpoint of cell cycle heterogeneity in tumors. Overall, increasing our knowledge of cell cycle heterogeneity is a key to identifying new therapeutic solutions, and this emerging concept may provide us with new opportunities to prevent the dreadful cancer recurrence.
Collapse
Affiliation(s)
- Ebrahim H. Maleki
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 31-007 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, 917751376 Mashhad, Iran
| |
Collapse
|
4
|
Yang F, Fan Z, Zhang L, He Y, Hu R, Xiang J, Fu S, Wang G, Wang J, Tao X, Zhang P. Preparation and anti-triple-negative breast cancer cell effect of a nanoparticle for the codelivery of paclitaxel and gemcitabine. DISCOVER NANO 2023; 18:119. [PMID: 37735318 PMCID: PMC10513990 DOI: 10.1186/s11671-023-03899-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Amphiphilic polymers (HA-ANI) were prepared by grafting hyaluronic acid (HA) and 6-(2-nitroimidazole)hexylamine (ANI) and then self-assemble in water to form nanoparticles (NPs) that could be loaded with paclitaxel (PTX) and gemcitabine (GEM) by dialysis. Infrared spectroscopy and 1H-NMR indicated the successful synthesis of HA-ANI. Three different ratios of NPs were prepared by adjusting the ratios of hydrophilic and hydrophobic materials, and the particle size decreased as the ratio of hydrophilic materials increased. When HA:ANI = 2.0:1, the nanoparticles had the smallest size distribution, good stability and near spherical shape and had high drug loading and encapsulation rates. In vitro release experiments revealed that NADPH could accelerate the drug release from NPs. Cellular uptake rate reached 86.50% at 6 h. The toxic effect of dual drug-loaded nanoparticles (P/G NPs) on MDA-MB-231 cells at 48 h was stronger than that of the free drug. The AO/EB double-staining assay revealed that a large number of late apoptotic cells appeared in the P/G NPs group, and the degree of cell damage was significantly stronger than that of the free drug group. In the cell migration assay, the 24 h-cell migration rate of the P/G NPs group was 5.99%, which was much lower than that of the free group (13.87% and 17.00%). In conclusion, MDA-MB-231 cells could effectively take up P/G NPs, while the introduction of the nano-codelivery system could significantly enhance the toxicity of the drug to MDA-MB-231 cells as well as the migration inhibition effect.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Zehui Fan
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Lixia Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yanjuan He
- Department of Pediatrics, The Fourth Hospital of Changsha, 70 Lushan Road, Changsha, 410006, Hunan, China
| | - Run Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jinkun Xiang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shiyang Fu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Guowei Wang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jianlong Wang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiaojun Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Pan Zhang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
5
|
Kluska M, Piastowska-Ciesielska AW, Tokarz P. Cell Cycle Status Influences Resistance to Apoptosis Induced by Oxidative Stress in Human Breast Cancer Cells, Which Is Accompanied by Modulation of Autophagy. Curr Issues Mol Biol 2023; 45:6325-6338. [PMID: 37623218 PMCID: PMC10453102 DOI: 10.3390/cimb45080399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer cells are characterised by uncontrolled cell proliferation; however, some of them can temporarily arrest their cell cycle at the G0 or G1 phase, which could contribute to tumour heterogeneity and drug resistance. The cell cycle status plays a critical role in chemosensitivity; however, the influence of G0- and G1-arrest has not been elucidated. To study the cell cycle arrest-mediated resistance, we used MCF-7 cells and generated three populations of cells: (1) cells arrested in the G0-like phase, (2) cells that resumed the cell cycle after the G0-like phase and (3) cells arrested in early G1 with a history of G0-like arrest. We observed that both the G0-like- and the G1-arrested cells acquired resistance to apoptosis induced by oxidative stress, accompanied by a decreased intracellular reactive oxygen species and DNA damage. This effect was associated with increased autophagy, likely facilitating their survival at DNA damage insult. The cell cycle reinitiation restored a sensitivity to oxidative stress typical for cells with a non-modulated cell cycle, with a concomitant decrease in autophagy. Our results support the need for further research on the resistance of G0- and G1-arrested cancer cells to DNA-damaging agents and present autophagy as a candidate for targeting in anticancer treatment.
Collapse
Affiliation(s)
- Magdalena Kluska
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | | | - Paulina Tokarz
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
6
|
Ding L, Kun W, Xu W, Chen S, Cai Z. Comparative analysis of clinicopathological characteristics of central necrotizing breast cancer and basal cell-like breast cancer. Front Oncol 2023; 13:915949. [PMID: 37114130 PMCID: PMC10127251 DOI: 10.3389/fonc.2023.915949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
PurposeThis study aims to compare the clinicopathological and immunohistochemical characteristics of centrally necrotizing carcinoma of the breast (CNC) and basal-like breast cancer (BLBC), as well as to analyze the characteristics of the molecular typing of the CNC.MethodsThe clinicopathological features of 69 cases of CNC and 48 cases of BLBC were observed and compared. EnVision immunohistochemical staining was performed to detect the expressions of hypoxia-inducible factor 1α (HIF-1α), breast cancer susceptibility gene 1 (BRCA1), and vascular endothelial growth factor (VEGF) in CNC and BLBC.ResultsThe age of the 69 patients ranged from 32 to 80 years, with an average of 54.55 years. Gross examination showed that most tumors were well-defined single central nodules with a diameter of 1.2~5.0 cm. Microscopically, there is a large necrotic or acellular area in the center of the tumor, mainly composed of tumor coagulative necrosis with varying degrees of fibrosis or hyaline degeneration. A small amount of cancer tissue remained in the form of a ribbon or small nest around the necrotic focus. Among 69 cases of CNC, the proportion of basal cell type (56.5%) was significantly higher than that of lumen type A (18.84%), lumen type B (13.04%), HER2 overexpression (5.8%), and nonexpression (5.8%). A total of 31 cases were followed up for 8~50 months, with an average of 33.94 months. There have been nine cases of disease progression. When compared to BLBC, there were no significant differences in BRCA1 and VEGF protein expression in response to CNC (p > 0.05), but there were significant differences in protein expression in HIF-1α (p < 0.05).ConclusionThe molecular typing of CNC showed that over half of those were BLBC. No statistically significant difference in the expression of BRCA1 was observed between CNC and BLBC; thus, we predict that targeted therapy for BRCA1 in BLBC may also have considerable effects in CNC patients. The expression of HIF-1α is significantly different in CNC and BLBC, and perhaps HIF-1α can be used as a new entry point to distinguish between the two. There is a significant correlation between the expression of VEGF and HIF-1α in BLBC, and there was no significant correlation between the expression levels of the two proteins in CNC.
Collapse
Affiliation(s)
- Li Ding
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Wang Kun
- Department of Pathology, Mengcheng Hospital of Traditional Chinese Medicine, Bozhou, Anhui, China
| | - Wenjing Xu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shaohua Chen
- Department of Pathology, Bengbu Medical College and The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhaogen Cai
- Department of Pathology, Bengbu Medical College and The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- *Correspondence: Zhaogen Cai,
| |
Collapse
|
7
|
Lindell E, Zhong L, Zhang X. Quiescent Cancer Cells-A Potential Therapeutic Target to Overcome Tumor Resistance and Relapse. Int J Mol Sci 2023; 24:ijms24043762. [PMID: 36835173 PMCID: PMC9959385 DOI: 10.3390/ijms24043762] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Quiescent cancer cells (QCCs) are nonproliferating cells arrested in the G0 phase, characterized by ki67low and p27high. QCCs avoid most chemotherapies, and some treatments could further lead to a higher proportion of QCCs in tumors. QCCs are also associated with cancer recurrence since they can re-enter a proliferative state when conditions are favorable. As QCCs lead to drug resistance and tumor recurrence, there is a great need to understand the characteristics of QCCs, decipher the mechanisms that regulate the proliferative-quiescent transition in cancer cells, and develop new strategies to eliminate QCCs residing in solid tumors. In this review, we discussed the mechanisms of QCC-induced drug resistance and tumor recurrence. We also discussed therapeutic strategies to overcome resistance and relapse by targeting QCCs, including (i) identifying reactive quiescent cancer cells and removing them via cell-cycle-dependent anticancer reagents; (ii) modulating the quiescence-to-proliferation switch; and (iii) eliminating QCCs by targeting their unique features. It is believed that the simultaneous co-targeting of proliferating and quiescent cancer cells may ultimately lead to the development of more effective therapeutic strategies for the treatment of solid tumors.
Collapse
|
8
|
Alhasan BA, Gordeev SA, Knyazeva AR, Aleksandrova KV, Margulis BA, Guzhova IV, Suvorova II. The mTOR Pathway in Pluripotent Stem Cells: Lessons for Understanding Cancer Cell Dormancy. MEMBRANES 2021; 11:858. [PMID: 34832087 PMCID: PMC8620939 DOI: 10.3390/membranes11110858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022]
Abstract
Currently, the success of targeted anticancer therapies largely depends on the correct understanding of the dormant state of cancer cells, since it is increasingly regarded to fuel tumor recurrence. The concept of cancer cell dormancy is often considered as an adaptive response of cancer cells to stress, and, therefore, is limited. It is possible that the cancer dormant state is not a privilege of cancer cells but the same reproductive survival strategy as diapause used by embryonic stem cells (ESCs). Recent advances reveal that high autophagy and mTOR pathway reduction are key mechanisms contributing to dormancy and diapause. ESCs, sharing their main features with cancer stem cells, have a delicate balance between the mTOR pathway and autophagy activity permissive for diapause induction. In this review, we discuss the functioning of the mTOR signaling and autophagy in ESCs in detail that allows us to deepen our understanding of the biology of cancer cell dormancy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Irina I. Suvorova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (B.A.A.); (S.A.G.); (A.R.K.); (K.V.A.); (B.A.M.); (I.V.G.)
| |
Collapse
|
9
|
Safaric Tepes P, Pal D, Lindsted T, Ibarra I, Lujambio A, Jimenez Sabinina V, Senturk S, Miller M, Korimerla N, Huang J, Glassman L, Lee P, Zeltsman D, Hyman K, Esposito M, Hannon GJ, Sordella R. An epigenetic switch regulates the ontogeny of AXL-positive/EGFR-TKi-resistant cells by modulating miR-335 expression. eLife 2021; 10:e66109. [PMID: 34254585 PMCID: PMC8285107 DOI: 10.7554/elife.66109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/10/2021] [Indexed: 01/15/2023] Open
Abstract
Despite current advancements in research and therapeutics, lung cancer remains the leading cause of cancer-related mortality worldwide. This is mainly due to the resistance that patients develop against chemotherapeutic agents over the course of treatment. In the context of non-small cell lung cancers (NSCLC) harboring EGFR-oncogenic mutations, augmented levels of AXL and GAS6 have been found to drive resistance to EGFR tyrosine kinase inhibitors such as Erlotinib and Osimertinib in certain tumors with mesenchymal-like features. By studying the ontogeny of AXL-positive cells, we have identified a novel non-genetic mechanism of drug resistance based on cell-state transition. We demonstrate that AXL-positive cells are already present as a subpopulation of cancer cells in Erlotinib-naïve tumors and tumor-derived cell lines and that the expression of AXL is regulated through a stochastic mechanism centered on the epigenetic regulation of miR-335. The existence of a cell-intrinsic program through which AXL-positive/Erlotinib-resistant cells emerge infers the need of treating tumors harboring EGFR-oncogenic mutations upfront with combinatorial treatments targeting both AXL-negative and AXL-positive cancer cells.
Collapse
Affiliation(s)
- Polona Safaric Tepes
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Faculty of Pharmacy University of LjubljanaLjubljanaSlovenia
| | - Debjani Pal
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Graduate Program in Molecular and Cellular Biology, Stony Brook UniversityStony Brook, New YorkUnited States
| | - Trine Lindsted
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Ingrid Ibarra
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Amaia Lujambio
- Icahn School of Medicine at Mount Sinai, Hess Center for Science and MedicineNew YorkUnited States
| | | | - Serif Senturk
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Madison Miller
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Navya Korimerla
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Graduate Program in Biomedical Engineering, Stony Brook UniversityNew YorkUnited States
| | - Jiahao Huang
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Lawrence Glassman
- Northwell Health Long Island, Jewish Medical CenterNew YorkUnited States
| | - Paul Lee
- Northwell Health Long Island, Jewish Medical CenterNew YorkUnited States
| | - David Zeltsman
- Northwell Health Long Island, Jewish Medical CenterNew YorkUnited States
| | - Kevin Hyman
- Northwell Health Long Island, Jewish Medical CenterNew YorkUnited States
| | - Michael Esposito
- Northwell Health Long Island, Jewish Medical CenterNew YorkUnited States
| | - Gregory J Hannon
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Cancer Research UK – Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - Raffaella Sordella
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Watson School of Biological Sciences, Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| |
Collapse
|
10
|
Regan JL, Schumacher D, Staudte S, Steffen A, Lesche R, Toedling J, Jourdan T, Haybaeck J, Mumberg D, Henderson D, Győrffy B, Regenbrecht CRA, Keilholz U, Schäfer R, Lange M. RNA sequencing of long-term label-retaining colon cancer stem cells identifies novel regulators of quiescence. iScience 2021; 24:102618. [PMID: 34142064 PMCID: PMC8185225 DOI: 10.1016/j.isci.2021.102618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/23/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Recent data suggest that therapy-resistant quiescent cancer stem cells (qCSCs) are the source of relapse in colon cancer. Here, using colon cancer patient-derived organoids and xenografts, we identify rare long-term label-retaining qCSCs that can re-enter the cell cycle to generate new tumors. RNA sequencing analyses demonstrated that these cells display the molecular hallmarks of quiescent tissue stem cells, including expression of p53 signaling genes, and are enriched for transcripts common to damage-induced quiescent revival stem cells of the regenerating intestine. In addition, we identify negative regulators of cell cycle, downstream of p53, that we show are indicators of poor prognosis and may be targeted for qCSC abolition in both p53 wild-type and mutant tumors. These data support the temporal inhibition of downstream targets of p53 signaling, in combination with standard-of-care treatments, for the elimination of qCSCs and prevention of relapse in colon cancer. Colon tumors contain therapy-resistant quiescent cancer stem cells (qCSCs) qCSC gene expression mirrors that of quiescent stem cells of the regenerating gut qCSCs are enriched for p53 signaling genes qCSC elimination may be achieved by inhibiting downstream targets of p53 signaling
Collapse
Affiliation(s)
- Joseph L Regan
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Dirk Schumacher
- Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany
| | - Stephanie Staudte
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany.,Department of Radiation Oncology and Radiotherapy, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Andreas Steffen
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Ralf Lesche
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| | - Joern Toedling
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| | - Thibaud Jourdan
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria.,Diagnostic & Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Dominik Mumberg
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - David Henderson
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary.,TTK Cancer Biomarker Research Group, Institute of Enzymology, 1117 Budapest, Hungary
| | - Christian R A Regenbrecht
- Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,CELLphenomics GmbH, 13125 Berlin, Germany.,Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Reinhold Schäfer
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany
| | - Martin Lange
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| |
Collapse
|
11
|
Resistance to Neoadjuvant Treatment in Breast Cancer: Clinicopathological and Molecular Predictors. Cancers (Basel) 2020; 12:cancers12082012. [PMID: 32708049 PMCID: PMC7463925 DOI: 10.3390/cancers12082012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 01/30/2023] Open
Abstract
Neoadjuvant Chemotherapy (NAC) in Breast Cancer (BC) has proved useful for the reduction in tumor burden prior to surgery, allowing for a more extensive breast preservation and the eradication of subjacent micrometastases. However, the impact on prognosis is highly dependent on the establishment of Pathological Complete Response (pCR), in particular for Triple Negative (TN) and Hormonal Receptor negative/Human Epidermal growth factor Receptor 2 positive (HR-/HER2+) subtypes. Several pCR predictors, such as PAM50, Integrative Cluster (IntClust), mutations in PI3KCA, or the Trastuzumab Risk model (TRAR), are useful molecular tools for estimating response to treatment and are prognostic. Major evolution events during BC NAC that feature the Residual Disease (RD) are the loss of HR and HER2, which are prognostic of bad outcome, and stemness and immune depletion-related gene expression aberrations. This dynamic nature of the determinants of response to BC NAC, together with the extensive heterogeneity of BC, raises the need to discern the individual and subtype-specific determinants of resistance. Moreover, refining the current approaches for a comprehensive monitoring of tumor evolution during treatment, RD, and eventual recurrences is essential for identifying new actionable alterations and the integral best management of the disease.
Collapse
|
12
|
Francescangeli F, Contavalli P, De Angelis ML, Careccia S, Signore M, Haas TL, Salaris F, Baiocchi M, Boe A, Giuliani A, Tcheremenskaia O, Pagliuca A, Guardiola O, Minchiotti G, Colace L, Ciardi A, D'Andrea V, La Torre F, Medema J, De Maria R, Zeuner A. A pre-existing population of ZEB2 + quiescent cells with stemness and mesenchymal features dictate chemoresistance in colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:2. [PMID: 31910865 PMCID: PMC6947904 DOI: 10.1186/s13046-019-1505-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/12/2019] [Indexed: 12/28/2022]
Abstract
Background Quiescent/slow cycling cells have been identified in several tumors and correlated with therapy resistance. However, the features of chemoresistant populations and the molecular factors linking quiescence to chemoresistance are largely unknown. Methods A population of chemoresistant quiescent/slow cycling cells was isolated through PKH26 staining (which allows to separate cells on the basis of their proliferation rate) from colorectal cancer (CRC) xenografts and subjected to global gene expression and pathway activation analyses. Factors expressed by the quiescent/slow cycling population were analyzed through lentiviral overexpression approaches for their ability to induce a dormant chemoresistant state both in vitro and in mouse xenografts. The correlation between quiescence-associated factors, CRC consensus molecular subtype and cancer prognosis was analyzed in large patient datasets. Results Untreated colorectal tumors contain a population of quiescent/slow cycling cells with stem cell features (quiescent cancer stem cells, QCSCs) characterized by a predetermined mesenchymal-like chemoresistant phenotype. QCSCs expressed increased levels of ZEB2, a transcription factor involved in stem cell plasticity and epithelial-mesenchymal transition (EMT), and of antiapototic factors pCRAF and pASK1. ZEB2 overexpression upregulated pCRAF/pASK1 levels resulting in increased chemoresistance, enrichment of cells with stemness/EMT traits and proliferative slowdown of tumor xenografts. In parallel, chemotherapy treatment of tumor xenografts induced the prevalence of QCSCs with a stemness/EMT phenotype and activation of the ZEB2/pCRAF/pASK1 axis, resulting in a chemotherapy-unresponsive state. In CRC patients, increased ZEB2 levels correlated with worse relapse-free survival and were strongly associated to the consensus molecular subtype 4 (CMS4) characterized by dismal prognosis, decreased proliferative rates and upregulation of EMT genes. Conclusions These results show that chemotherapy-naive tumors contain a cell population characterized by a coordinated program of chemoresistance, quiescence, stemness and EMT. Such population becomes prevalent upon drug treatment and is responsible for chemotherapy resistance, thus representing a key target for more effective therapeutic approaches.
Collapse
Affiliation(s)
- Federica Francescangeli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paola Contavalli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Silvia Careccia
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Michele Signore
- RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Tobias Longin Haas
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Federico Salaris
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marta Baiocchi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Boe
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanita, Viale Regina Elena 299, 00161, Rome, Italy
| | - Olga Tcheremenskaia
- Environment and Health Department, Istituto Superiore di Sanita, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alfredo Pagliuca
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Ombretta Guardiola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR,Via Pietro Castellino 111, 80131, Naples, Italy
| | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR,Via Pietro Castellino 111, 80131, Naples, Italy
| | - Lidia Colace
- Department of Surgical Sciences, Policlinico Umberto I/Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Antonio Ciardi
- Department of Surgery "Pietro Valdoni", Policlinico Umberto I/Sapienza University of Rome, via Lancisi 2, 00161, Rome, Italy
| | - Vito D'Andrea
- Department of Surgical Sciences, Policlinico Umberto I/Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Filippo La Torre
- Surgical Sciences and Emergency Department, Policlinico Umberto I/Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - JanPaul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Ruggero De Maria
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy. .,Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy.
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
13
|
Abstract
The transition between proliferating and quiescent states must be carefully regulated to ensure that cells divide to create the cells an organism needs only at the appropriate time and place. Cyclin-dependent kinases (CDKs) are critical for both transitioning cells from one cell cycle state to the next, and for regulating whether cells are proliferating or quiescent. CDKs are regulated by association with cognate cyclins, activating and inhibitory phosphorylation events, and proteins that bind to them and inhibit their activity. The substrates of these kinases, including the retinoblastoma protein, enforce the changes in cell cycle status. Single cell analysis has clarified that competition among factors that activate and inhibit CDK activity leads to the cell's decision to enter the cell cycle, a decision the cell makes before S phase. Signaling pathways that control the activity of CDKs regulate the transition between quiescence and proliferation in stem cells, including stem cells that generate muscle and neurons. © 2020 American Physiological Society. Compr Physiol 10:317-344, 2020.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA.,Department of Biological Chemistry, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
14
|
De Angelis ML, Francescangeli F, La Torre F, Zeuner A. Stem Cell Plasticity and Dormancy in the Development of Cancer Therapy Resistance. Front Oncol 2019; 9:626. [PMID: 31355143 PMCID: PMC6636659 DOI: 10.3389/fonc.2019.00626] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer treatment with either standard chemotherapy or targeted agents often results in the emergence of drug-refractory cell populations, ultimately leading to therapy failure. The biological features of drug resistant cells are largely overlapping with those of cancer stem cells and include heterogeneity, plasticity, self-renewal ability, and tumor-initiating capacity. Moreover, drug resistance is usually characterized by a suppression of proliferation that can manifest as quiescence, dormancy, senescence, or proliferative slowdown. Alterations in key cellular pathways such as autophagy, unfolded protein response or redox signaling, as well as metabolic adaptations also contribute to the establishment of drug resistance, thus representing attractive therapeutic targets. Moreover, a complex interplay of drug resistant cells with the micro/macroenvironment and with the immune system plays a key role in dictating and maintaining the resistant phenotype. Recent studies have challenged traditional views of cancer drug resistance providing innovative perspectives, establishing new connections between drug resistant cells and their environment and indicating unexpected therapeutic strategies. In this review we discuss recent advancements in understanding the mechanisms underlying drug resistance and we report novel targeting agents able to overcome the drug resistant status, with particular focus on strategies directed against dormant cells. Research on drug resistant cancer cells will take us one step forward toward the development of novel treatment approaches and the improvement of relapse-free survival in solid and hematological cancer patients.
Collapse
Affiliation(s)
- Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Filippo La Torre
- Department of Surgical Sciences Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
15
|
Stem Cells Inhibition by Bevacizumab in Combination with Neoadjuvant Chemotherapy for Breast Cancer. J Clin Med 2019; 8:jcm8050612. [PMID: 31064127 PMCID: PMC6572380 DOI: 10.3390/jcm8050612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Preclinical works have suggested cytotoxic chemotherapies may increase the number of cancer stem cells (CSC) whereas angiogenesis inhibition may decrease CSC proliferation. We developed a proof of concept clinical trial to explore bevacizumab activity on breast CSC. Breast cancer patients requiring preoperative chemotherapy were included in this open-label, randomized, prospective, multicenter phase II trial. All received FEC-docetaxel combination, and patients randomized in the experimental arm received concomitant bevacizumab. The primary endpoint was to describe ALDH1 (Aldehyde dehydrogenase 1) positive tumor cells rate before treatment and after the fourth cycle. Secondary objectives included safety, pathological complete response (pCR) rate, disease-free survival (DFS), relapse-free survival (RFS), and overall survival (OS). Seventy-five patients were included. ALDH1+ cells rate increase was below the predefined 5% threshold in both arms for the 32 patients with two time points available. Grade 3 or 4 adverse events rates were similar in both arms. A non-significant increase in pCR was observed in the bevacizumab arm (42.6% vs. 18.2%, p = 0.06), but survival was not improved (OS: p = 0.89; DFS: p = 0.45; and RFS: p = 0.68). The increase of ALDH1+ tumor cells rate after bevacizumab-based chemotherapy was less than 5%. However, as similar results were observed with chemotherapy alone, bevacizumab impact on breast CSC cells cannot be confirmed.
Collapse
|
16
|
AKT1 low quiescent cancer cells in ductal carcinoma in situ of the breast. NPJ Breast Cancer 2019; 5:10. [PMID: 30911675 PMCID: PMC6428812 DOI: 10.1038/s41523-019-0105-y] [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: 09/08/2018] [Accepted: 01/08/2019] [Indexed: 12/02/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) of the breast precedes the development of invasive breast cancer and reflects the genomic changes and protein expression profile of invasive disease. AKT1low cancer cells (QCC) are a rare, drug-tolerant, epigenetically plastic, and quiescent cancer cell subset that we previously identified at a frequency of 0.5–1% in primary breast tumors using the marker profile: AKTlow/H3K9me2low/HES1high. Here we used quantitative immunofluorescence microscopy with computational image analysis to show that AKT1low QCCs are present in DCIS from patients with and without co-existing invasive breast cancer. These data suggest that a drug-resistant, quiescent cancer cell state is present in premalignant breast lesions prior to the development of invasive disease. These findings warrant further study of whether AKT1low QCCs contribute to invasive tumor development and recurrence, similar to their role in more advanced malignancy.
Collapse
|
17
|
Li Y, Chu J, Feng W, Yang M, Zhang Y, Zhang Y, Qin Y, Xu J, Li J, Vasilatos SN, Fu Z, Huang Y, Yin Y. EPHA5 mediates trastuzumab resistance in HER2-positive breast cancers through regulating cancer stem cell-like properties. FASEB J 2019; 33:4851-4865. [PMID: 30620624 DOI: 10.1096/fj.201701561rrrr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Trastuzumab is a successful, rationally designed therapy that provides significant clinical benefit for human epidermal growth factor receptor-2 (HER2)-positive breast cancer patients. However, about half of individuals with HER2-positive breast cancer do not respond to trastuzumab treatment because of various resistance mechanisms, including but not limited to: 1) shedding of the HER2 extracellular domain, 2) steric hindrance ( e.g., MUC4 and MUC1), 3) parallel pathway activation (this is the general mechanism cited in the quote above), 4) perturbation of downstream signaling events ( e.g., PTEN loss or PIK3CA mutation), and 5) immunologic mechanisms (such as FcR polymorphisms). EPHA5, a receptor tyrosine kinase, has been demonstrated to act as an anticancer agent in several cancer cell types. In this study, deletion of EPHA5 can significantly increase the resistance of HER2-positive breast cancer patients to trastuzumab. To investigate how EPHA5 deficiency induces trastuzumab resistance, clustered regularly interspaced short palindromic repeat technology was used to create EPHA5-deficient variants of breast cancer cells. EPHA5 deficiency effectively increases breast cancer stem cell (BCSC)-like properties, including NANOG, CD133+, E-cadherin expression, and the CD44+/CD24-/low phenotype, concomitantly enhancing mammosphere-forming ability. EPHA5 deficiency also caused significant aggrandized tumor malignancy in trastuzumab-sensitive xenografts, coinciding with the up-regulation of BCSC-related markers and intracellular Notch1 and PTEN/AKT signaling pathway activation. These findings highlight that EPHA5 is a potential prognostic marker for the activity of Notch1 and better sensitivity to trastuzumab in HER2-positive breast cancer. Moreover, patients with HER2-positive breast cancers expressing high Notch1 activation and low EPHA5 expression could be the best candidates for anti-Notch1 therapy.-Li, Y., Chu, J., Feng, W., Yang, M., Zhang, Y., Zhang, Y., Qin, Y., Xu, J., Li, J., Vasilatos, S. N., Fu, Z., Huang, Y., Yin, Y. EPHA5 mediates trastuzumab resistance in HER2-positive breast cancers through regulating cancer stem cell-like properties.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and.,Department of Breast Diseases, Jiangsu Province Hospital of Traditional Chinese Medicine (TMC)/Affiliated Hospital of Nanjing University of TCM, Nanjing, China
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wanting Feng
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengzhu Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhong Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanqiu Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ye Qin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and
| | - Juan Xu
- Nanjing Maternal and Child Health Medical Institute, Affiliated Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shauna N Vasilatos
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Maternal and Child Health Medical Institute, Affiliated Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Huang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Gillies RJ, Brown JS, Anderson ARA, Gatenby RA. Eco-evolutionary causes and consequences of temporal changes in intratumoural blood flow. Nat Rev Cancer 2018; 18:576-585. [PMID: 29891961 PMCID: PMC6441333 DOI: 10.1038/s41568-018-0030-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Temporal changes in blood flow are commonly observed in malignant tumours, but the evolutionary causes and consequences are rarely considered. We propose that stochastic temporal variations in blood flow and microenvironmental conditions arise from the eco-evolutionary dynamics of tumour angiogenesis in which cancer cells, as individual units of selection, can influence and respond only to local environmental conditions. This leads to new vessels arising from the closest available vascular structure regardless of the size or capacity of this parental vessel. These dynamics produce unstable vascular networks with unpredictable spatial and temporal variations in blood flow and microenvironmental conditions. Adaptations of evolving populations to temporally varying environments in nature include increased diversity, greater motility and invasiveness, and highly plastic phenotypes, allowing for broad metabolic adaptability and rapid shifts to high rates of proliferation and profound quiescence. These adaptive strategies, when adopted in cancer cells, promote many commonly observed phenotypic properties including those found in the stem phenotype and in epithelial-to-mesenchymal transition. Temporal variations in intratumoural blood flow, which occur through the promotion of cancer cell phenotypes that facilitate both metastatic spread and resistance to therapy, may have substantial clinical consequences.
Collapse
Affiliation(s)
- Robert J Gillies
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Robert A Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
19
|
Alves CP, Dey-Guha I, Kabraji S, Yeh AC, Talele NP, Solé X, Chowdhury J, Mino-Kenudson M, Loda M, Sgroi D, Borresen-Dale AL, Russnes HG, Ross KN, Ramaswamy S. AKT1 low Quiescent Cancer Cells Promote Solid Tumor Growth. Mol Cancer Ther 2017; 17:254-263. [PMID: 29054988 DOI: 10.1158/1535-7163.mct-16-0868] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/21/2017] [Accepted: 10/04/2017] [Indexed: 11/16/2022]
Abstract
Human tumor growth depends on rapidly dividing cancer cells driving population expansion. Even advanced tumors, however, contain slowly proliferating cancer cells for reasons that remain unclear. Here, we selectively disrupt the ability of rapidly proliferating cancer cells to spawn AKT1low daughter cells that are rare, slowly proliferating, tumor-initiating, and chemotherapy-resistant, using β1-integrin activation and the AKT1-E17K-mutant oncoprotein as experimental tools in vivo Surprisingly, we find that selective depletion of AKT1low slow proliferators actually reduces the growth of a molecularly diverse panel of human cancer cell xenograft models without globally altering cell proliferation or survival in vivo Moreover, we find that unusual cancer patients with AKT1-E17K-mutant solid tumors also fail to produce AKT1low quiescent cancer cells and that this correlates with significantly prolonged survival after adjuvant treatment compared with other patients. These findings support a model whereby human solid tumor growth depends on not only rapidly proliferating cancer cells but also on the continuous production of AKT1low slow proliferators. Mol Cancer Ther; 17(1); 254-63. ©2017 AACR.
Collapse
Affiliation(s)
- Cleidson P Alves
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Ipsita Dey-Guha
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Sheheryar Kabraji
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Albert C Yeh
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Nilesh P Talele
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Xavier Solé
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Joeeta Chowdhury
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Mari Mino-Kenudson
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Harvard Medical School, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Dennis Sgroi
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Anne-Lise Borresen-Dale
- Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hege G Russnes
- Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kenneth N Ross
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. .,Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Harvard-Ludwig Center for Cancer Research, Boston, Massachusetts
| |
Collapse
|
20
|
Sarmiento-Castro A, Caamaño-Gutiérrez E, Sims AH, Hull NJ, James MI, Santiago-Gómez A, Eyre R, Clark C, Brown ME, Brooks MD, Wicha MS, Howell SJ, Clarke RB, Simões BM. Circulating immune complexes in coccidioidomycosis. Detection and characterization. J Clin Invest 1980; 15:307-316. [PMID: 32707076 PMCID: PMC7419713 DOI: 10.1016/j.stemcr.2020.06.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/12/2023] Open
Abstract
Estrogen-receptor-positive breast tumors are treated with anti-estrogen (AE) therapies but frequently develop resistance. Cancer stem cells (CSCs) with high aldehyde dehydrogenase activity (ALDH+ cells) are enriched following AE treatment. Here, we show that the interleukin-1β (IL-1β) signaling pathway is activated in ALDH+ cells, and data from single cells reveals that AE treatment selects for IL-1 receptor (IL1R1)-expressing ALDH+ cells. Importantly, CSC activity is reduced by an IL1R1 inhibitor in AE-resistant models. Moreover, IL1R1 expression is increased in the tumors of patients treated with AE therapy and predicts treatment failure. Single-cell gene expression analysis revealed that at least two subpopulations exist within the ALDH+ population, one proliferative and one quiescent. Following AE therapy the quiescent population is expanded, which suggests CSC dormancy as an adaptive strategy that facilitates treatment resistance. Targeting of ALDH+IL1R1+ cells merits testing as a strategy to combat AE resistance in patients with residual disease. Anti-estrogen-resistant ALDH+ cells have increased CSC activity in ER+ tumors The IL1R1-expressing ALDH+ CSC population expands after anti-estrogen treatment IL1R1 expression predicts anti-estrogen treatment failure Targeting of ALDH+IL1R1+ cells reverses anti-estrogen resistance
Collapse
Affiliation(s)
- Aida Sarmiento-Castro
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Eva Caamaño-Gutiérrez
- Technology Directorate, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer Group, University of Edinburgh Cancer Research UK Centre, Edinburgh EH4 2XR, UK
| | - Nathan J Hull
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Mark I James
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Angélica Santiago-Gómez
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Rachel Eyre
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Christopher Clark
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Martha E Brown
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael D Brooks
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max S Wicha
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sacha J Howell
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK
| | - Robert B Clarke
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK.
| | - Bruno M Simões
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, UK.
| |
Collapse
|