1
|
Bhin J, Yemelyanenko J, Chao X, Klarenbeek S, Opdam M, Malka Y, Hoekman L, Kruger D, Bleijerveld O, Brambillasca CS, Sprengers J, Siteur B, Annunziato S, van Haren MJ, Martin NI, van de Ven M, Peters D, Agami R, Linn SC, Boven E, Altelaar M, Jonkers J, Zingg D, Wessels LF. MYC is a clinically significant driver of mTOR inhibitor resistance in breast cancer. J Exp Med 2023; 220:e20211743. [PMID: 37642941 PMCID: PMC10465700 DOI: 10.1084/jem.20211743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Targeting the PI3K-AKT-mTOR pathway is a promising therapeutic strategy for breast cancer treatment. However, low response rates and development of resistance to PI3K-AKT-mTOR inhibitors remain major clinical challenges. Here, we show that MYC activation drives resistance to mTOR inhibitors (mTORi) in breast cancer. Multiomic profiling of mouse invasive lobular carcinoma (ILC) tumors revealed recurrent Myc amplifications in tumors that acquired resistance to the mTORi AZD8055. MYC activation was associated with biological processes linked to mTORi response and counteracted mTORi-induced translation inhibition by promoting translation of ribosomal proteins. In vitro and in vivo induction of MYC conferred mTORi resistance in mouse and human breast cancer models. Conversely, AZD8055-resistant ILC cells depended on MYC, as demonstrated by the synergistic effects of mTORi and MYCi combination treatment. Notably, MYC status was significantly associated with poor response to everolimus therapy in metastatic breast cancer patients. Thus, MYC is a clinically relevant driver of mTORi resistance that may stratify breast cancer patients for mTOR-targeted therapies.
Collapse
Affiliation(s)
- Jinhyuk Bhin
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Department of Biomedical System Informatics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Julia Yemelyanenko
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Xue Chao
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yuval Malka
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Liesbeth Hoekman
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dinja Kruger
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Onno Bleijerveld
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Chiara S. Brambillasca
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Justin Sprengers
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bjørn Siteur
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Stefano Annunziato
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Matthijs J. van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Reuven Agami
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Sabine C. Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Epie Boven
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maarten Altelaar
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Daniel Zingg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Lodewyk F.A. Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
2
|
Histology-based survival outcomes in hormone receptor-positive metastatic breast cancer treated with targeted therapies. NPJ Breast Cancer 2022; 8:131. [PMID: 36539444 PMCID: PMC9768132 DOI: 10.1038/s41523-022-00499-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
The addition of targeted therapies (TT) to endocrine therapy (ET) has improved the outcomes of patients with HR-positive, HER2-negative metastatic breast cancer (mBC). However, it is unknown whether patients with invasive lobular carcinoma (ILC) or mixed invasive ductal and lobular carcinoma (mixed) histologies experience the same magnitude of benefit from this therapy as those with invasive ductal carcinoma (IDC). We aim to determine whether patients with IDC, ILC, and mixed HR+/HER2- mBC derive similar benefit from the addition of cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6is), mammalian target of rapamycin inhibitor (mTORi), and phosphoinositide 3-kinase inhibitor (PI3Ki) to ET in HR+/HER2- mBC. We conducted an observational, population-based investigation using data from the MD Anderson prospectively collected database. We conducted a histology-based analysis of progression-free survival (PFS) and overall survival (OS) durations in 3784 patients with HR+/HER2- mBC who were treated with TT plus ET between January 1, 2010, and December 31, 2021. Out of the 3784 patients, 2975 were included in the final analysis. Of these, 2249 received CDK4/6is (81% IDC, 15% ILC, and 4% mixed), 1027 received everolimus (82% IDC, 14% ILC, and 4% mixed) and 49 received alpelisib (81% IDC and 19% ILC). The addition of targeted therapy to ET did not result in statistically significant differences in PFS or OS duration among patients with IDC, ILC, and mixed HR+/HER2- mBC. We concluded that for patients with HR+/HER2- mBC, the addition of TT to ET leads to a similar magnitude of benefit, irrespective of histology.
Collapse
|
3
|
Van Baelen K, Geukens T, Maetens M, Tjan-Heijnen V, Lord CJ, Linn S, Bidard FC, Richard F, Yang WW, Steele RE, Pettitt SJ, Van Ongeval C, De Schepper M, Isnaldi E, Nevelsteen I, Smeets A, Punie K, Voorwerk L, Wildiers H, Floris G, Vincent-Salomon A, Derksen PWB, Neven P, Senkus E, Sawyer E, Kok M, Desmedt C. Current and future diagnostic and treatment strategies for patients with invasive lobular breast cancer. Ann Oncol 2022; 33:769-785. [PMID: 35605746 DOI: 10.1016/j.annonc.2022.05.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Invasive lobular breast cancer (ILC) is the second most common type of breast cancer after invasive breast cancer of no special type (NST), representing up to 15% of all breast cancers. DESIGN Latest data on ILC are presented, focusing on diagnosis, molecular make-up according to the European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) guidelines, treatment in the early and metastatic setting and ILC-focused clinical trials. RESULTS At the imaging level, magnetic resonance imaging-based and novel positron emission tomography/computed tomography-based techniques can overcome the limitations of currently used imaging techniques for diagnosing ILC. At the pathology level, E-cadherin immunohistochemistry could help improving inter-pathologist agreement. The majority of patients with ILC do not seem to benefit as much from (neo-)adjuvant chemotherapy as patients with NST, although chemotherapy might be required in a subset of high-risk patients. No differences in treatment efficacy are seen for anti-human epidermal growth factor receptor 2 (HER2) therapies in the adjuvant setting and cyclin-dependent kinases 4 and 6 inhibitors in the metastatic setting. The clinical utility of the commercially available prognostic gene expression-based tests is unclear for patients with ILC. Several ESCAT alterations differ in frequency between ILC and NST. Germline BRCA1 and PALB2 alterations are less frequent in patients with ILC, while germline CDH1 (gene coding for E-cadherin) alterations are more frequent in patients with ILC. Somatic HER2 mutations are more frequent in ILC, especially in metastases (15% ILC versus 5% NST). A high tumour mutational burden, relevant for immune checkpoint inhibition, is more frequent in ILC metastases (16%) than in NST metastases (5%). Tumours with somatic inactivating CDH1 mutations may be vulnerable for treatment with ROS1 inhibitors, a concept currently investigated in early and metastatic ILC. CONCLUSION ILC is a unique malignancy based on its pathological and biological features leading to differences in diagnosis as well as in treatment response, resistance and targets as compared to NST.
Collapse
Affiliation(s)
- K Van Baelen
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; Departments of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - T Geukens
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - M Maetens
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | - V Tjan-Heijnen
- Medical Oncology Department, Maastricht University Medical Center (MUMC), School of GROW, Maastricht, The Netherlands
| | - C J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - S Linn
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands; Departments of Medical Oncology, Amsterdam, The Netherlands; Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - F-C Bidard
- Department of Medical Oncology, Institut Curie, UVSQ/Paris-Saclav University, Paris, France
| | - F Richard
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | - W W Yang
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - R E Steele
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - S J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - C Van Ongeval
- Departments of Radiology, UZ Leuven, Leuven, Belgium
| | - M De Schepper
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; Pathology, UZ Leuven, Leuven, Belgium
| | - E Isnaldi
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - A Smeets
- Surgical Oncology, UZ Leuven, Leuven, Belgium
| | - K Punie
- General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - L Voorwerk
- Departments of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H Wildiers
- General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - G Floris
- Pathology, UZ Leuven, Leuven, Belgium
| | | | - P W B Derksen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Neven
- Departments of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - E Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - E Sawyer
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Cancer Centre, King's College London, London, UK
| | - M Kok
- Departments of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C Desmedt
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Bestard O, Crespo E. Disarming the Old Foe. Restoring T-Cell Immune Function with mTor-Inhibitors to Tackle Cytomegalovirus Infection. J Am Soc Nephrol 2022; 33:6-8. [PMID: 34969818 PMCID: PMC8763183 DOI: 10.1681/asn.2021111471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Oriol Bestard
- Department of Nephrology and Kidney Transplantation, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Vall d’Hebron Barcelona Campus Hospital, Barcelona Autonomous University, Barcelona, Spain
| | - Elena Crespo
- Department of Nephrology and Kidney Transplantation, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Vall d’Hebron Barcelona Campus Hospital, Barcelona Autonomous University, Barcelona, Spain
| |
Collapse
|
5
|
Sflomos G, Schipper K, Koorman T, Fitzpatrick A, Oesterreich S, Lee AV, Jonkers J, Brunton VG, Christgen M, Isacke C, Derksen PWB, Brisken C. Atlas of Lobular Breast Cancer Models: Challenges and Strategic Directions. Cancers (Basel) 2021; 13:5396. [PMID: 34771558 PMCID: PMC8582475 DOI: 10.3390/cancers13215396] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Invasive lobular carcinoma (ILC) accounts for up to 15% of all breast cancer (BC) cases and responds well to endocrine treatment when estrogen receptor α-positive (ER+) yet differs in many biological aspects from other ER+ BC subtypes. Up to 30% of patients with ILC will develop late-onset metastatic disease up to ten years after initial tumor diagnosis and may experience failure of systemic therapy. Unfortunately, preclinical models to study ILC progression and predict the efficacy of novel therapeutics are scarce. Here, we review the current advances in ILC modeling, including cell lines and organotypic models, genetically engineered mouse models, and patient-derived xenografts. We also underscore four critical challenges that can be addressed using ILC models: drug resistance, lobular tumor microenvironment, tumor dormancy, and metastasis. Finally, we highlight the advantages of shared experimental ILC resources and provide essential considerations from the perspective of the European Lobular Breast Cancer Consortium (ELBCC), which is devoted to better understanding and translating the molecular cues that underpin ILC to clinical diagnosis and intervention. This review will guide investigators who are considering the implementation of ILC models in their research programs.
Collapse
Affiliation(s)
- George Sflomos
- ISREC—Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Koen Schipper
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (K.S.); (A.F.); (C.I.)
| | - Thijs Koorman
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (T.K.); (P.W.B.D.)
| | - Amanda Fitzpatrick
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (K.S.); (A.F.); (C.I.)
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.O.); (A.V.L.)
- Magee Women’s Cancer Research Institute, Pittsburgh, PA 15213, USA
- Cancer Biology Program, Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Adrian V. Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.O.); (A.V.L.)
- Magee Women’s Cancer Research Institute, Pittsburgh, PA 15213, USA
- Cancer Biology Program, Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Oncode Institute, 1066 CX Amsterdam, The Netherlands
| | - Valerie G. Brunton
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK;
| | - Matthias Christgen
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
| | - Clare Isacke
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (K.S.); (A.F.); (C.I.)
| | - Patrick W. B. Derksen
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (T.K.); (P.W.B.D.)
| | - Cathrin Brisken
- ISREC—Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (K.S.); (A.F.); (C.I.)
| |
Collapse
|
6
|
Invasive Lobular Carcinoma of the Breast: Ongoing Trials, Challenges, and Future Directions. CURRENT BREAST CANCER REPORTS 2021. [DOI: 10.1007/s12609-021-00412-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Purpose of Review
Invasive lobular carcinoma (ILC) is increasingly recognized as a distinct subtype of breast cancer with unique management challenges. We reviewed currently available clinical trials for patients with ILC.
Recent Findings
We describe the rationale for and study design of clinical trials for patients with both early stage and metastatic ILC. Molecular alterations specific to or enriched in ILC may serve as treatment targets.
Summary
ILC has specific features that may be treatment targets. Clinical trials for ILC are available and being developed.
Collapse
|
7
|
el Hage A, Dormond O. Combining mTOR Inhibitors and T Cell-Based Immunotherapies in Cancer Treatment. Cancers (Basel) 2021; 13:1359. [PMID: 33802831 PMCID: PMC8002586 DOI: 10.3390/cancers13061359] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
mTOR regulates several processes that control tumor development, including cancer cell growth, angiogenesis and the immune response to tumor. Accordingly, mTOR inhibitors have been thoroughly explored in cancer therapy but have failed to provide long-lasting anticancer benefits. Several resistance mechanisms that counteract the antitumor effect of mTOR inhibitors have been identified and have highlighted the need to use mTOR inhibitors in combination therapies. In this context, emerging evidence has demonstrated that mTOR inhibitors, despite their immunosuppressive properties, provide anticancer benefits to immunotherapies. In fact, mTOR inhibitors also display immunostimulatory effects, in particular by promoting memory CD8+ T cell generation. Hence, mTOR inhibitors represent a therapeutic opportunity to promote antitumor CD8 responses and to boost the efficacy of different modalities of cancer immunotherapy. In this context, strategies to reduce the immunosuppressive activity of mTOR inhibitors and therefore to shift the immune response toward antitumor immunity will be useful. In this review, we present the different classes of mTOR inhibitors and discuss their effect on immune cells by focusing mainly on CD8+ T cells. We further provide an overview of the different preclinical studies that investigated the anticancer effects of mTOR inhibitors combined to immunotherapies.
Collapse
Affiliation(s)
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| |
Collapse
|
8
|
Pramod N, Nigam A, Basree M, Mawalkar R, Mehra S, Shinde N, Tozbikian G, Williams N, Majumder S, Ramaswamy B. Comprehensive Review of Molecular Mechanisms and Clinical Features of Invasive Lobular Cancer. Oncologist 2021; 26:e943-e953. [PMID: 33641217 DOI: 10.1002/onco.13734] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
Invasive lobular carcinoma (ILC) accounts for 10% to 15% of breast cancers in the United States, 80% of which are estrogen receptor (ER)-positive, with an unusual metastatic pattern of spread to sites such as the serosa, meninges, and ovaries, among others. Lobular cancer presents significant challenges in detection and clinical management given its multifocality and multicentricity at presentation. Despite the unique features of ILC, it is often lumped with hormone receptor-positive invasive ductal cancers (IDC); consequently, ILC screening, treatment, and follow-up strategies are largely based on data from IDC. Despite both being treated as ER-positive breast cancer, querying the Cancer Genome Atlas database shows distinctive molecular aberrations in ILC compared with IDC, such as E-cadherin loss (66% vs. 3%), FOXA1 mutations (7% vs. 2%), and GATA3 mutations (5% vs. 20%). Moreover, compared with patients with IDC, patients with ILC are less likely to undergo breast-conserving surgery, with lower rates of complete response following therapy as these tumors are less chemosensitive. Taken together, this suggests that ILC is biologically distinct, which may influence tumorigenesis and therapeutic strategies. Long-term survival and clinical outcomes in patients with ILC are worse than in stage- and grade-matched patients with IDC; therefore, nuanced criteria are needed to better define treatment goals and protocols tailored to ILC's unique biology. This comprehensive review highlights the histologic and clinicopathologic features that distinguish ILC from IDC, with an in-depth discussion of ILC's molecular alterations and biomarkers, clinical trials and treatment strategies, and future targets for therapy. IMPLICATIONS FOR PRACTICE: The majority of invasive lobular breast cancers (ILCs) are hormone receptor (HR)-positive and low grade. Clinically, ILC is treated similar to HR-positive invasive ductal cancer (IDC). However, ILC differs distinctly from IDC in its clinicopathologic characteristics and molecular alterations. ILC also differs in response to systemic therapy, with studies showing ILC as less sensitive to chemotherapy. Patients with ILC have worse clinical outcomes with late recurrences. Despite these differences, clinical trials treat HR-positive breast cancers as a single disease, and there is an unmet need for studies addressing the unique challenges faced by patients diagnosed with ILC.
Collapse
Affiliation(s)
- Nikhil Pramod
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Akanksha Nigam
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Mustafa Basree
- University of Pikeville Kentucky College of Osteopathic Medicine, Pikeville, Kentucky, USA
| | - Resham Mawalkar
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Saba Mehra
- University of Toledo - Health Science Campus, Toledo, Ohio, USA
| | - Neelam Shinde
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Gary Tozbikian
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Nicole Williams
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Sarmila Majumder
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Bhuvaneswari Ramaswamy
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
9
|
Li N, Li Z, Li X, Chen B, Sun H, Zhao K. Identification of an immune-related long noncoding RNA signature that predicts prognosis in breast cancer patients. Biomark Med 2021; 15:167-180. [PMID: 33496624 DOI: 10.2217/bmm-2020-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Aim: The purpose of this study was to identify an immune-related long noncoding RNA (lncRNA) signature that predicts the prognosis of breast cancer. Materials & methods: The expression profiles of breast cancer were downloaded from The Cancer Genome Atlas. Cox regression analysis was used to identify an immune-related lncRNA signature. Results: The five immune-related lncRNAs could be used to construct a breast cancer survival prognosis model. The receiver operating characteristic curve evaluation found that the accuracy of the model for predicting the 1-, 3- and 5-year prognosis of breast cancer was 0.688, 0.708 and 0.686. Conclusion: This signature may have an important clinical significance for improving predictive results and guiding the treatment of breast cancer patients.
Collapse
Affiliation(s)
- Na Li
- Breast surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161000, PR China
| | - Zubin Li
- Breast surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161000, PR China
| | - Xin Li
- Breast surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161000, PR China
| | - Bingjie Chen
- Nursing department, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161000, PR China
| | - Huibo Sun
- Breast surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161000, PR China
| | - Kun Zhao
- Department of pathology, The Qiqihar Medical College
| |
Collapse
|
10
|
McCart Reed AE, Kalinowski L, Simpson PT, Lakhani SR. Invasive lobular carcinoma of the breast: the increasing importance of this special subtype. Breast Cancer Res 2021; 23:6. [PMID: 33413533 PMCID: PMC7792208 DOI: 10.1186/s13058-020-01384-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is the most common of the breast cancer special types, accounting for up to 15% of all breast cancer cases. ILCs are noted for their lack of E-cadherin function, which underpins their characteristic discohesive growth pattern, with cells arranged in single file and dispersed throughout the stroma. Typically, tumours are luminal in molecular subtype, being oestrogen and progesterone receptor positive, and HER2 negative. Since last reviewing the lobular literature (McCart Reed et al., Breast Cancer Res 17:12, 2015), there has been a considerable increase in research output focused on this tumour type, including studies into the pathology and management of disease, a high-resolution definition of the genomic landscape of tumours as well as the evolution of several potential therapeutic avenues. There abounds a huge amount of new data, which we will review herein.
Collapse
Affiliation(s)
- Amy E McCart Reed
- UQ Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia.
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia.
| | - Lauren Kalinowski
- UQ Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
- Department of Histopathology, Sullivan Nicolaides Pathology, Bowen Hills, Brisbane, Australia
| | - Peter T Simpson
- UQ Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
- Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, Brisbane, Australia
| |
Collapse
|