1
|
Jiang S, Wang X, Ma Z. Efficacy of combined immunotherapy and targeted therapy in overcoming barriers to postoperative recurrence in squamous subtype anaplastic thyroid carcinoma with abscess: a case report and literature review. Front Oncol 2025; 15:1477954. [PMID: 40177243 PMCID: PMC11961886 DOI: 10.3389/fonc.2025.1477954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Molecularly targeted therapies and immunotherapy are increasingly being employed in the treatment of aggressive, recurrent thyroid cancer. Evidence from several studies indicates that a significant proportion of tumor patients derive limited benefit from immunotherapy as a monotherapy, with vascular abnormalities in solid tumors contributing to immune evasion. Numerous studies, both domestic and international, have assessed the efficacy of combining immune checkpoint inhibitors with antiangiogenic agents across various tumor types. These studies suggest that such combination therapies are effective in controlling disease progression and extending survival, among other outcomes. Nevertheless, further research is warranted to substantiate these findings and optimize treatment protocols. Methods This study aims to describe a patient diagnosed with anaplastic thyroid carcinoma (ATC) combined with primary squamous cell carcinoma of the thyroid (PSCCT) and concurrent thyroid abscess. The patient experienced local recurrence and metastasis following surgical intervention, radiotherapy, and chemotherapy, and was found to be PD-1 negative. Disease progression was effectively controlled through combination therapy with anlotinib and tislelizumab. Additionally, a comprehensive review of the relevant literature was conducted. Results The patient exhibited disease recurrence 8 months postoperatively, notwithstanding the administration of adjuvant radiotherapy and chemotherapy. The local recurrent mass demonstrated minimal reduction following 4 cycles of targeted therapy with anlotinib. However, subsequent treatment with a combination of anlotinib and tislelizumab resulted in a substantial reduction of the neck mass and enlarged cervical lymph nodes after 12 cycles. The patient tolerated the combination therapy well, experiencing no significant adverse effects aside from pronounced fatigue. Thus, the combination therapy with anlotinib and tislelizumab proved effective in controlling the disease. Conclusion The management of postoperative recurrence of ATC-PSCCT presents significant challenges, as recurrent tumors typically demonstrate increased aggressiveness and resistance to pharmacological interventions, necessitating multimodal therapeutic approaches. Tislelizumab, an immune checkpoint inhibitor, may facilitate immune-mediated tumor clearance through the activation of various immune cells, including natural killer cells and macrophages. Despite the patient's PD-1 negativity, the combination of anlotinib and tislelizumab may exert synergistic effects through distinct mechanisms, thereby potentially enhancing therapeutic efficacy. The integration of a multi-targeted tyrosine kinase inhibitor within this combination therapy regimen warrants further investigation.
Collapse
Affiliation(s)
- Shuyun Jiang
- Department of Clinical Medicine, Qinghai University, Xining, Qinghai, China
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Xiaowu Wang
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Zhijun Ma
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| |
Collapse
|
2
|
Ma C, Li Y, Zhu H, Li Z, Liu Y. Clinical applications of circulating tumor cell detection: challenges and strategies. Clin Chem Lab Med 2024:cclm-2024-0959. [PMID: 39610299 DOI: 10.1515/cclm-2024-0959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Circulating tumor cells (CTCs) are pivotal in the distant metastasis of tumors, serving as one of the primary materials for liquid biopsy. They hold significant clinical importance in assessing prognosis, predicting efficacy, evaluating therapeutic outcomes, and studying recurrence, metastasis, and resistance mechanisms in cancer patients. Nevertheless, the rareness and heterogeneity of CTC and the complexity of metastasis make the clinical application of CTC detection confront many challenges, which may need to be settled by some practical strategies. This article will review the content mentioned above.
Collapse
Affiliation(s)
- Chunhui Ma
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yang Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Hai Zhu
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Zhiyong Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yi Liu
- 26460 The Fifth Medical Center of Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
3
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [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: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Ciccioli M, Kim K, Khazan N, Khoury JD, Cooke MJ, Miller MC, O'Shannessy DJ, Pailhes-Jimenez AS, Moore RG. Identification of circulating tumor cells captured by the FDA-cleared Parsortix ® PC1 system from the peripheral blood of metastatic breast cancer patients using immunofluorescence and cytopathological evaluations. J Exp Clin Cancer Res 2024; 43:240. [PMID: 39169412 PMCID: PMC11337573 DOI: 10.1186/s13046-024-03149-x] [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: 05/03/2024] [Accepted: 08/03/2024] [Indexed: 08/23/2024] Open
Abstract
Circulating Tumor Cells (CTCs) may serve as a non-invasive source of tumor material to investigate an individual's disease in real-time. The Parsortix® PC1 System, the first FDA-cleared medical device for the capture and harvest of CTCs from peripheral blood of metastatic breast cancer (MBC) patients for use in subsequent user-validated downstream analyses, enables the epitope-independent capture of CTCs with diverse phenotypes based on cell size and deformability. The aim of this study was to determine the proportion of MBC patients and self-declared female healthy volunteers (HVs) that had CTCs identified using immunofluorescence (IF) or Wright-Giemsa (WG) staining. Peripheral blood from 76 HVs and 76 MBC patients was processed on Parsortix® PC1 Systems. Harvested cells were cytospun onto a charged slide and immunofluorescently stained for identification of CTCs expressing epithelial markers. The IF slides were subsequently WG-stained and analyzed for CTC identification based on morphological features of malignant cells. All testing was performed by operators blinded to the clinical status of each subject. CTCs were identified on the IF slides in 45.3% (≥ 1) / 24.0% (≥ 5) of the MBC patients (range = 0 - 125, mean = 7) and in 6.9% (≥ 1) / 2.8% (≥ 5) of the HVs (range = 0 - 28, mean = 1). Among the MBC patients with ≥ 1 CTC, 70.6% had only CK + /EpCAM- CTCs, with none having EpCAM + /CK- CTCs. CTC clusters were identified in 56.0% of the CTC-positive patients. On the WG-stained slides, CTCs were identified in 42.9% (≥ 1) / 21.4% (≥ 5) of the MBC patients (range = 0 - 41, mean = 4) and 4.3% (≥ 1) / 2.9% (≥ 5) of the HVs (range = 0 - 14, mean = 0). This study demonstrated the ability of the Parsortix® PC1 System to capture and harvest CTCs from a significantly larger proportion of MBC patients compared to HVs when coupled with both IF and WG cytomorphological assessment. The presence of epithelial cells in subjects without diagnosed disease has been previously described, with their significance being unclear. Interestingly, a high proportion of the identified CTCs did not express EpCAM, highlighting the limitations of using EpCAM-based approaches.
Collapse
Affiliation(s)
| | - Kyukwang Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Negar Khazan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Joseph D Khoury
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | - Richard G Moore
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
5
|
Kouhmareh K, Martin E, Finlay D, Bhadada A, Hernandez-Vargas H, Downey F, Allen JK, Teriete P. Capture of circulating metastatic cancer cell clusters from lung cancer patients can reveal unique genomic profiles and potential anti-metastatic molecular targets: A proof-of-concept study. PLoS One 2024; 19:e0306450. [PMID: 39083508 PMCID: PMC11290651 DOI: 10.1371/journal.pone.0306450] [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: 02/28/2024] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
Metastasis remains the leading cause of cancer deaths worldwide and lung cancer, known for its highly metastatic progression, remains among the most lethal of malignancies. Lung cancer metastasis can selectively spread to multiple different organs, however the genetic and molecular drivers for this process are still poorly understood. Understanding the heterogeneous genomic profile of lung cancer metastases is considered key in identifying therapeutic targets that prevent its spread. Research has identified the key source for metastasis being clusters of cells rather than individual cancer cells. These clusters, known as metastatic cancer cell clusters (MCCCs) have been shown to be 100-fold more tumorigenic than individual cancer cells. Unfortunately, access to these primary drivers of metastases remains difficult and has limited our understanding of their molecular and genomic profiles. Strong evidence in the literature suggests that differentially regulated biological pathways in MCCCs can provide new therapeutic drug targets to help combat cancer metastases. In order to expand research into MCCCs and their role in metastasis, we demonstrate a novel, proof of principle technology, to capture MCCCs directly from patients' whole blood. Our platform can be readily tuned for different solid tumor types by combining a biomimicry-based margination effect coupled with immunoaffinity to isolate MCCCs. Adopting a selective capture approach based on overexpressed CD44 in MCCCs provides a methodology that preferentially isolates them from whole blood. Furthermore, we demonstrate a high capture efficiency of more than 90% when spiking MCCC-like model cell clusters into whole blood. Characterization of the captured MCCCs from lung cancer patients by immunofluorescence staining and genomic analyses, suggests highly differential morphologies and genomic profiles. This study lays the foundation to identify potential drug targets thus unlocking a new area of anti-metastatic therapeutics.
Collapse
Affiliation(s)
| | - Erika Martin
- PhenoVista Biosciences, San Diego, CA, United States of America
| | - Darren Finlay
- National Cancer Institute Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States of America
| | | | | | | | | | - Peter Teriete
- TumorGen Inc., San Diego, CA, United States of America
- IDEAYA Biosciences, South San Francisco, CA, United States of America
| |
Collapse
|
6
|
Sayed ZS, Khattap MG, Madkour MA, Yasen NS, Elbary HA, Elsayed RA, Abdelkawy DA, Wadan AHS, Omar I, Nafady MH. Circulating tumor cells clusters and their role in Breast cancer metastasis; a review of literature. Discov Oncol 2024; 15:94. [PMID: 38557916 PMCID: PMC10984915 DOI: 10.1007/s12672-024-00949-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Breast cancer is a significant and deadly threat to women globally. Moreover, Breast cancer metastasis is a complicated process involving multiple biological stages, which is considered a substantial cause of death, where cancer cells spread from the original tumor to other organs in the body-representing the primary mortality factor. Circulating tumor cells (CTCs) are cancer cells detached from the primary or metastatic tumor and enter the bloodstream, allowing them to establish new metastatic sites. CTCs can travel alone or in groups called CTC clusters. Studies have shown that CTC clusters have more potential for metastasis and a poorer prognosis than individual CTCs in breast cancer patients. However, our understanding of CTC clusters' formation, structure, function, and detection is still limited. This review summarizes the current knowledge of CTC clusters' biological properties, isolation, and prognostic significance in breast cancer. It also highlights the challenges and future directions for research and clinical application of CTC clusters.
Collapse
Affiliation(s)
- Zeinab S Sayed
- Faculty of Applied Medical Science, Misr University for Science and Technology, 26Th of July Corridor, 6Th of October, Giza Governorate, Postal Code: 77, Egypt
| | - Mohamed G Khattap
- Technology of Radiology and Medical Imaging Program, Faculty of Applied Health Sciences Technology, Galala University, Suez, 435611, Egypt
| | | | - Noha S Yasen
- Radiology and Imaging Technology Department, Faculty of Applied Health Science Technology, Delta University for Science and Technology, Gamasa, Al Mansurah, Egypt
| | - Hanan A Elbary
- Faculty of Applied Medical Science, Misr University for Science and Technology, 26Th of July Corridor, 6Th of October, Giza Governorate, Postal Code: 77, Egypt
| | - Reem A Elsayed
- Faculty of Applied Medical Science, Misr University for Science and Technology, 26Th of July Corridor, 6Th of October, Giza Governorate, Postal Code: 77, Egypt
| | - Dalia A Abdelkawy
- Faculty of Applied Medical Science, Misr University for Science and Technology, 26Th of July Corridor, 6Th of October, Giza Governorate, Postal Code: 77, Egypt
| | | | - Islam Omar
- Faculty of Pharmacy, South Valley University, Qena, Egypt
| | - Mohamed H Nafady
- Radiation Sciences Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, 6th of october, Egypt.
| |
Collapse
|
7
|
Bacon ER, Ihle K, Guo W, Egelston CA, Simons DL, Wei C, Tumyan L, Schmolze D, Lee PP, Waisman JR. Tumor heterogeneity and clinically invisible micrometastases in metastatic breast cancer-a call for enhanced surveillance strategies. NPJ Precis Oncol 2024; 8:81. [PMID: 38553598 PMCID: PMC10980766 DOI: 10.1038/s41698-024-00572-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024] Open
Abstract
The biology of metastatic breast cancer (MBC) is understudied, primarily due to the difficulty of procuring multiple samples from patients with oligometastatic breast cancer. We developed a rapid postmortem tissue procurement program that allows the collection and analysis of numerous metastatic lesions, subclinical locations, and potential pre-metastatic niches that fall within this scope. We conducted a rapid postmortem tissue collection study on 9 patients with MBC. Patients and their families consented to donate tissues immediately after death in an IRB-approved study. Various disease subtypes, progression histories, organ involvement, and final causes of death are reported. In patients with hormone receptor-positive (HR+) disease, estrogen receptor (ER), progesterone receptor (PR), HER2, and Ki-67 expression were heterogeneous across metastatic lesions within individual patients. Disease phenotype at the end of life trended toward complete loss of HR expression. Nearly all (n = 7) patients exhibited extensive tumor involvement of additional organs that had not been previously diagnosed clinically and were not retrospectively visible on recent imaging. Of these seven individuals, three included organs uncommonly associated with MBC: kidney, spleen, pancreas, and ovary. Finally, we identified clinically undetectable micrometastases in several organs uncommonly involved in MBC. Our findings raise several clinically relevant questions regarding the mechanisms of metastatic progression. Insights from this study argue for better surveillance strategies for monitoring MBC. We highlight the need to capture more accurate biomarker information in the context of heterogeneous disease and urge the consideration of treatment strategies that combine multiple targeted therapies.
Collapse
Affiliation(s)
- Eliza R Bacon
- The Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA.
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA.
| | - Kena Ihle
- The Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Weihua Guo
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - Christina Wei
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Lusine Tumyan
- Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - James R Waisman
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
8
|
Li H, Huang H, Tan H, Jia Q, Song W, Zhang Q, Zhou B, Bai J. Key processes in tumor metastasis and therapeutic strategies with nanocarriers: a review. Mol Biol Rep 2024; 51:197. [PMID: 38270746 DOI: 10.1007/s11033-023-08910-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/26/2024]
Abstract
Cancer metastasis is the leading cause of cancer-related death. Metastasis occurs at all stages of tumor development, with unexplored changes occurring at the primary site and distant colonization sites. The growing understanding of the metastatic process of tumor cells has contributed to the emergence of better treatment options and strategies. This review summarizes a range of features related to tumor cell metastasis and nanobased drug delivery systems for inhibiting tumor metastasis. The mechanisms of tumor metastasis in the ideal order of metastatic progression were summarized. We focus on the prominent role of nanocarriers in the treatment of tumor metastasis, summarizing the latest applications of nanocarriers in combination with drugs to target important components and processes of tumor metastasis and providing ideas for more effective nanodrug delivery systems.
Collapse
Affiliation(s)
- Hongjie Li
- School of Clinical Medicine, Weifang Medical University, 261053, Weifang, China
| | - Haiqin Huang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, 250012, Jinan, China
| | - Qitao Jia
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Weina Song
- Department of Pediatric Respiratory and Critical Care, Qilu Hospital of Shandong University Dezhou Hospital, 253000, Dezhou, China
| | - Qingdong Zhang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| |
Collapse
|
9
|
Lopez-Gonzalez L, Sanchez Cendra A, Sanchez Cendra C, Roberts Cervantes ED, Espinosa JC, Pekarek T, Fraile-Martinez O, García-Montero C, Rodriguez-Slocker AM, Jiménez-Álvarez L, Guijarro LG, Aguado-Henche S, Monserrat J, Alvarez-Mon M, Pekarek L, Ortega MA, Diaz-Pedrero R. Exploring Biomarkers in Breast Cancer: Hallmarks of Diagnosis, Treatment, and Follow-Up in Clinical Practice. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:168. [PMID: 38256428 PMCID: PMC10819101 DOI: 10.3390/medicina60010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Breast cancer is a prevalent malignancy in the present day, particularly affecting women as one of the most common forms of cancer. A significant portion of patients initially present with localized disease, for which curative treatments are pursued. Conversely, another substantial segment is diagnosed with metastatic disease, which has a worse prognosis. Recent years have witnessed a profound transformation in the prognosis for this latter group, primarily due to the discovery of various biomarkers and the emergence of targeted therapies. These biomarkers, encompassing serological, histological, and genetic indicators, have demonstrated their value across multiple aspects of breast cancer management. They play crucial roles in initial diagnosis, aiding in the detection of relapses during follow-up, guiding the application of targeted treatments, and offering valuable insights for prognostic stratification, especially for highly aggressive tumor types. Molecular markers have now become the keystone of metastatic breast cancer diagnosis, given the diverse array of chemotherapy options and treatment modalities available. These markers signify a transformative shift in the arsenal of therapeutic options against breast cancer. Their diagnostic precision enables the categorization of tumors with elevated risks of recurrence, increased aggressiveness, and heightened mortality. Furthermore, the existence of therapies tailored to target specific molecular anomalies triggers a cascade of changes in tumor behavior. Therefore, the primary objective of this article is to offer a comprehensive review of the clinical, diagnostic, prognostic, and therapeutic utility of the principal biomarkers currently in use, as well as of their clinical impact on metastatic breast cancer. In doing so, our goal is to contribute to a more profound comprehension of this complex disease and, ultimately, to enhance patient outcomes through more precise and effective treatment strategies.
Collapse
Affiliation(s)
- Laura Lopez-Gonzalez
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Alicia Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Cristina Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | | | - Javier Cassinello Espinosa
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Tatiana Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Oscar Fraile-Martinez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Cielo García-Montero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Ana María Rodriguez-Slocker
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - Soledad Aguado-Henche
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Jorge Monserrat
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Melchor Alvarez-Mon
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Leonel Pekarek
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Miguel A. Ortega
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| |
Collapse
|
10
|
Reduzzi C, Vismara M, Schamberger T, Silvestri M, Motta R, Polzer BM, Cappelletti V. Isolation and Genomic Analysis of Circulating Tumor Cell Clusters in Cancer Patients. Methods Mol Biol 2024; 2752:101-117. [PMID: 38194030 DOI: 10.1007/978-1-0716-3621-3_7] [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] [Indexed: 01/10/2024]
Abstract
The role of circulating tumor cell (CTC) clusters in the metastatic dissemination process is gaining increased attention. Besides homotypic clusters, heterotypic clusters that contain tumor cells admixed with normal cells are frequently observed in patients with solid tumors. Current methods used for cluster detection and enumeration do not allow an accurate estimation of the relative fractions of tumor cells. Here we describe a method for estimating tumor fraction of clusters including isolation and collection of single clusters, assessment of copy number alterations of single clusters by low-pass whole genome sequencing, and bioinformatic analysis of sequencing data.
Collapse
Affiliation(s)
- Carolina Reduzzi
- Biomarkers Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan, Italy
- Division of Hematology/Oncology, Weill Cornell Medicine, New York, United States
| | - Marta Vismara
- Biomarkers Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Thomas Schamberger
- Experimental Medicine and Therapy Research, University Regensburg, Regensburg, Germany
| | - Marco Silvestri
- Biomarkers Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Rosita Motta
- Biomarkers Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Bernhard M Polzer
- Cellular and Molecular Diagnostics Group, Division of Personalized Cancer Therapy, Fraunhofer Institute of Toxicology and Experimental Medicine ITEM-R, Regensburg, Germany
| | - Vera Cappelletti
- Biomarkers Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| |
Collapse
|
11
|
Zhang X, Hong B, Sun Z, Zhao J, Li M, Wei D, Wang Y, Zhang N. Development and validation of a circulating tumor cells-related signature focusing on biochemical recurrence and immunotherapy response in prostate cancer. Heliyon 2023; 9:e22648. [PMID: 38107322 PMCID: PMC10724679 DOI: 10.1016/j.heliyon.2023.e22648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Background Studies have shown that the circulating tumor cells (CTCs) play a key role for invasion and formation of distant metastases in prostate cancer (PCa). However, few CTCs-related genes (CRGs) have been developed for biochemical recurrence (BCR) prediction and clinical applications of PCa patients. Materials and methods Bioinformatics analysis with public PCa datasets were used to investigate the relationship between the differentially expressed CRGs and BCR. Lasso-COX regression analysis was used to constructed and validated a CRGs-based BCR prediction signature for PCa. Single-cell data were used to validate the expression levels of signature genes in different cell types and then explored the cell-cell communication relationships. Finally, the expression levels of signature genes were verified and the CRGs involved in immunotherapy response were further identified. Results Thirteen CRGs were differentially expressed and closely associated with BCR in PCa. Then we constructed and validated a BCR prediction signature for PCa patients based on 3 differentially expressed CRGs (EMID1, SPP1 and UBE2C), and the signature was an independent factor to predict BCR for PCa. Single-cell data showed the specific expression patterns of the signature genes, while the SPP1 pathway plays an important role in cell-cell communication. Further analyses suggested UBE2C was highly expressed in BCR group and high expression of UBE2C had a better response for patients who received immunotherapy. Moreover, the expression levels of UBE2C in CTCs were higher than other cells and tissues, indicated that UBE2C may affect the BCR event of PCa patients through CTCs. Conclusion Our findings demonstrated that CRGs were significantly associated with BCR and immunotherapy efficacy in PCa and CRGs may influence the BCR event through CTCs.
Collapse
Affiliation(s)
| | | | - Zhipeng Sun
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiahui Zhao
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingchuan Li
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Dechao Wei
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yongxing Wang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Zhao H, Wang L, Fang C, Li C, Zhang L. Factors influencing the diagnostic and prognostic values of circulating tumor cells in breast cancer: a meta-analysis of 8,935 patients. Front Oncol 2023; 13:1272788. [PMID: 38090481 PMCID: PMC10711619 DOI: 10.3389/fonc.2023.1272788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/03/2023] [Indexed: 06/29/2024] Open
Abstract
Background Circulating tumor cells (CTCs) could serve as a predictive biomarker in breast cancer (BC). Due to its high heterogeneity, the diagnostic and prognostic values of CTC are challenging. Methods We searched published studies from the databases of PubMed, Cochrane Library, Embase, and MEDLINE. The detection capability and hazard ratios (HRs) of CTCs were extracted as the clinical diagnosis and prognosis evaluation. Subgroup analyses were divided according to the detection methods, continents, treatment periods, therapeutic plans, and cancer stages. Results In this study, 35 publications had been retrieved with 8,935 patients enrolled. The diagnostic efficacy of CTC detection has 74% sensitivity and 98% specificity. The positive CTC detection (CTC+ ) would predict worse OS and PFS/DFS in both mid-therapy and post-therapy (HROS, 3.09; 95% CI, 2.17–4.39; HRPFS/DFS, 2.06; 95% CI, 1.72–2.47). Moreover, CTC+ indicated poor survival irrespective of the treatment phases and sampling times (HROS, 2.43; 95% CI, 1.85–3.19; HRPFS/DFS, 1.82; 95% CI, 1.66–1.99). The CTC+ was associated with poor survival regardless of the continents of patients (HROS = 2.43; 95% CI, 1.85–3.19). Conclusion Our study suggested that CTC+ was associated with a worse OS and PFS/DFS in the Asian population. The detection method, the threshold level of CTC+ , therapeutic approaches, and sampling times would not affect its diagnostic and prognostic values.
Collapse
Affiliation(s)
- Hongfang Zhao
- Clinical Medicine College, Hebei University, Baoding, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Luxuan Wang
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Baoding, China
| | - Chuan Fang
- Clinical Medicine College, Hebei University, Baoding, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Baoding, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, China
| | - Chunhui Li
- Clinical Medicine College, Hebei University, Baoding, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Baoding, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, China
| | - Lijian Zhang
- Clinical Medicine College, Hebei University, Baoding, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Baoding, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, China
| |
Collapse
|
13
|
Kouhmareh K, Martin E, Finlay D, Bhadada A, Hernandez-Vargas H, Downey F, Allen JK, Teriete P. Capture of circulating metastatic cancer cell clusters from a lung cancer patient can reveal a unique genomic profile and potential anti-metastatic molecular targets: A proof of concept study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558270. [PMID: 37781582 PMCID: PMC10541091 DOI: 10.1101/2023.09.19.558270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Metastasis remains the leading cause of cancer deaths worldwide and lung cancer, known for its highly metastatic progression, remains among the most lethal of malignancies. The heterogeneous genomic profile of lung cancer metastases is often unknown. Since different metastatic events can selectively spread to multiple organs, strongly suggests more studies are needed to understand and target these different pathways. Unfortunately, access to the primary driver of metastases, the metastatic cancer cell clusters (MCCCs), remains difficult and limited. These metastatic clusters have been shown to be 100-fold more tumorigenic than individual cancer cells. Capturing and characterizing MCCCs is a key limiting factor in efforts to help treat and ultimately prevent cancer metastasis. Elucidating differentially regulated biological pathways in MCCCs will help uncover new therapeutic drug targets to help combat cancer metastases. We demonstrate a novel, proof of principle technology, to capture MCCCs directly from patients' whole blood. Our platform can be readily tuned for different solid tumor types by combining a biomimicry-based margination effect coupled with immunoaffinity to isolate MCCCs. Adopting a selective capture approach based on overexpressed CD44 in MCCCs provides a methodology that preferentially isolates them from whole blood. Furthermore, we demonstrate a high capture efficiency of more than 90% when spiking MCCC-like model cell clusters into whole blood. Characterization of the captured MCCCs from lung cancer patients by immunofluorescence staining and genomic analyses, suggests highly differential morphologies and genomic profiles., This study lays the foundation to identify potential drug targets thus unlocking a new area of anti-metastatic therapeutics.
Collapse
Affiliation(s)
- Kourosh Kouhmareh
- PhenoVista Biosciences, 6195 Cornerstone Ct E STE 114, San Diego, CA 92121
| | - Erika Martin
- PhenoVista Biosciences, 6195 Cornerstone Ct E STE 114, San Diego, CA 92121
| | - Darren Finlay
- NCI Cancer Center Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Anukriti Bhadada
- TumorGen Inc., 6197 Cornerstone Ct E STE #101, San Diego, CA 92121
| | | | - Francisco Downey
- TumorGen Inc., 6197 Cornerstone Ct E STE #101, San Diego, CA 92121
| | - Jeffrey K Allen
- TumorGen Inc., 6197 Cornerstone Ct E STE #101, San Diego, CA 92121
| | - Peter Teriete
- IDEAYA Biosciences, 7000 Shoreline Ct STE #350, South San Francisco, CA 94080
| |
Collapse
|
14
|
Chen S, Jiang W, Du Y, Yang M, Pan Y, Li H, Cui M. Single-cell analysis technologies for cancer research: from tumor-specific single cell discovery to cancer therapy. Front Genet 2023; 14:1276959. [PMID: 37900181 PMCID: PMC10602688 DOI: 10.3389/fgene.2023.1276959] [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: 08/13/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Single-cell sequencing (SCS) technology is changing our understanding of cellular components, functions, and interactions across organisms, because of its inherent advantage of avoiding noise resulting from genotypic and phenotypic heterogeneity across numerous samples. By directly and individually measuring multiple molecular characteristics of thousands to millions of single cells, SCS technology can characterize multiple cell types and uncover the mechanisms of gene regulatory networks, the dynamics of transcription, and the functional state of proteomic profiling. In this context, we conducted systematic research on SCS techniques, including the fundamental concepts, procedural steps, and applications of scDNA, scRNA, scATAC, scCITE, and scSNARE methods, focusing on the unique clinical advantages of SCS, particularly in cancer therapy. We have explored challenging but critical areas such as circulating tumor cells (CTCs), lineage tracing, tumor heterogeneity, drug resistance, and tumor immunotherapy. Despite challenges in managing and analyzing the large amounts of data that result from SCS, this technique is expected to reveal new horizons in cancer research. This review aims to emphasize the key role of SCS in cancer research and promote the application of single-cell technologies to cancer therapy.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Weibo Jiang
- Department of Orthopaedic, The Second Hospital of Jilin University, Changchun, China
| | - Yanhui Du
- Department of Orthopaedics, Jilin Province People’s Hospital, Changchun, China
| | - Manshi Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yihan Pan
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Huan Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Mengying Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Zhou JS, Liu ZN, Chen YY, Liu YX, Shen H, Hou LJ, Ding Y. New advances in circulating tumor cell‑mediated metastasis of breast cancer (Review). Mol Clin Oncol 2023; 19:71. [PMID: 37614367 PMCID: PMC10442766 DOI: 10.3892/mco.2023.2667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/20/2023] [Indexed: 08/25/2023] Open
Abstract
Breast cancer stands as the most prevalent form of cancer affecting women, with metastasis serving as a leading cause of mortality among patients with breast cancer. Gaining a comprehensive understanding of the metastatic mechanism in breast cancer is essential for early detection and precision treatment of the disease. Circulating tumor cells (CTCs) play a vital role in this context, representing cancer cells that detach from tumor tissues and enter the bloodstream of cancer patients. These cells travel in the blood circulation as single cells or clusters. Recent research has shed light on the enhanced metastatic potential of CTC clusters compared to single CTCs, despite their limited occurrence. The aim of the present review was to explore recent findings on CTCs with a particular focus on the clustering phenomenon of CTCs observed in breast cancer. Additionally, the present review delved into the comparison between single CTCs and CTC clusters regarding their implications for the treatment and prognosis of patients diagnosed with metastatic breast cancer. By examining the role and mechanisms of CTCs in breast cancer metastasis, the present review provided an improved understanding of CTCs and their significance in early detection of breast cancer metastasis through peripheral blood analysis. Moreover, it contributed to the comprehension of cancer prognosis and prediction by highlighting the implications of CTCs in these aspects. Ultimately, the present study seeks to advance knowledge in the field and pave the way for improved approaches to breast cancer management.
Collapse
Affiliation(s)
- Jiang-Shan Zhou
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zi-Ning Liu
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yuan-Yuan Chen
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yu-Xi Liu
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Hua Shen
- Department of Mathematics and Statistics, University of Calgary, Alberta T2N 1N4, Canada
| | - Li-Jun Hou
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
- Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yi Ding
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
- Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
16
|
Grašič Kuhar C, Silvester J, Mencinger M, Ovčariček T, Čemažar M, Miceska S, Modic Ž, Kuhar A, Jesenko T, Kloboves Prevodnik V. Association of Circulating Tumor Cells, Megakaryocytes and a High Immune-Inflammatory Environment in Metastatic Breast Cancer. Cancers (Basel) 2023; 15:3397. [PMID: 37444507 DOI: 10.3390/cancers15133397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Liquid biopsy is becoming an important source of new biomarkers during the treatment of metastatic cancer patients. Using size-based microfluid technology, we isolated circulating tumor cells (CTCs) from metastatic breast cancer patients to evaluate their presence and cluster formation, as well as the presence of megakaryocytes and immune-inflammatory blood cells, and to correlate their presence with clinicopathological data and overall survival (OS). In total, 59 patients (median age 60.4 years) were included in the study: 62.7% luminal A/B-like, 20.3% HER2-positive, and 17% triple-negative. Our results showed that at least one CTC was present in 79.7% and ≥5 CTCs in 35.2% of the patients. CTC clusters were present in patients with ≥5 CTCs only (in 19.2% of them), and megakaryocytes were present in 52% of all patients. The presence of CTC clusters and megakaryocytes was positively associated with the CTC count. Patients with low pan-inflammatory value (PIV), low systemic immune-inflammatory index (SII), and low relative change from baseline (ΔPIV%, ΔSII%) were associated with significantly higher OS than their counterparts. ΔPIV%, the presence of infection in the last month, and a long duration of metastatic disease were identified as independent prognostic factors for OS. The interplay of CTCs, CTC clusters, megakaryocytes, and PIV needs to be further explored.
Collapse
Affiliation(s)
- Cvetka Grašič Kuhar
- Department Medical Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
- Faculty of Medicine Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Jernej Silvester
- Faculty of Medicine Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Marina Mencinger
- Department Medical Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Tanja Ovčariček
- Department Medical Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Maja Čemažar
- Department of Experimental Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, 6000 Izola, Slovenia
| | - Simona Miceska
- Faculty of Medicine Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Cytopathology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Živa Modic
- Faculty of Medicine Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Experimental Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Anamarija Kuhar
- Department of Cytopathology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Tanja Jesenko
- Faculty of Medicine Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Experimental Oncology, Institute of Oncology, 1000 Ljubljana, Slovenia
| | - Veronika Kloboves Prevodnik
- Department of Cytopathology, Institute of Oncology, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Ljubljana, Slovenia
| |
Collapse
|
17
|
Strati A, Economopoulou P, Lianidou E, Psyrri A. Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy. Biomedicines 2023; 11:1768. [PMID: 37371863 DOI: 10.3390/biomedicines11061768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The approval of monoclonal antibodies against programmed death-ligand 1 (PD-L1) and programmed cell death protein (PD1) has changed the landscape of cancer treatment. To date, many immune checkpoint inhibitors (ICIs) have been approved by the FDA for the treatment of metastatic cancer as well as locally recurrent advanced cancer. However, immune-related adverse events (irAEs) of ICIs highlight the need for biomarker analysis with strong predictive value. Liquid biopsy is an important tool for clinical oncologists to monitor cancer patients and administer or change appropriate therapy. CTCs frequently express PD-L1, and this constitutes a clinically useful and non-invasive method to assess PD-L1 status in real-time. This review summarizes all the latest findings about the clinical significance of CTC for the management of cancer patients during the administration of immunotherapy and mainly focuses on the assessment of PD-L1 expression in CTCs.
Collapse
Affiliation(s)
- Areti Strati
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Panagiota Economopoulou
- Department of Internal Medicine, Section of Medical Oncology, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Amanda Psyrri
- Department of Internal Medicine, Section of Medical Oncology, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| |
Collapse
|
18
|
Schuster E, Dashzeveg N, Jia Y, Golam K, Zhang T, Hoffman A, Zhang Y, Zheng C, Ramos E, Taftaf R, Shennawy LE, Scholten D, Kitata RB, Adorno-Cruz V, Reduzzi C, Spahija S, Xu R, Siziopikou KP, Platanias LC, Shah A, Gradishar WJ, Cristofanilli M, Tsai CF, Shi T, Liu H. Computational ranking-assisted identification of Plexin-B2 in homotypic and heterotypic clustering of circulating tumor cells in breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536233. [PMID: 37090580 PMCID: PMC10120645 DOI: 10.1101/2023.04.10.536233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Metastasis is the cause of over 90% of all deaths associated with breast cancer, yet the strategies to predict cancer spreading based on primary tumor profiles and therefore prevent metastasis are egregiously limited. As rare precursor cells to metastasis, circulating tumor cells (CTCs) in multicellular clusters in the blood are 20-50 times more likely to produce viable metastasis than single CTCs. However, the molecular mechanisms underlying various CTC clusters, such as homotypic tumor cell clusters and heterotypic tumor-immune cell clusters, are yet to be fully elucidated. Combining machine learning-assisted computational ranking with experimental demonstration to assess cell adhesion candidates, we identified a transmembrane protein Plexin- B2 (PB2) as a new therapeutic target that drives the formation of both homotypic and heterotypic CTC clusters. High PB2 expression in human primary tumors predicts an unfavorable distant metastasis-free survival and is enriched in CTC clusters compared to single CTCs in advanced breast cancers. Loss of PB2 reduces formation of homotypic tumor cell clusters as well as heterotypic tumor-myeloid cell clusters in triple-negative breast cancer. Interactions between PB2 and its ligand Sema4C on tumor cells promote homotypic cluster formation, and PB2 binding with Sema4A on myeloid cells (monocytes) drives heterotypic CTC cluster formation, suggesting that metastasizing tumor cells hijack the PB2/Sema family axis to promote lung metastasis in breast cancer. Additionally, using a global proteomic analysis, we identified novel downstream effectors of the PB2 pathway associated with cancer stemness, cell cycling, and tumor cell clustering in breast cancer. Thus, PB2 is a novel therapeutic target for preventing new metastasis.
Collapse
|
19
|
Yang B, Lou C, Chen S, Zhang Z, Xu Q. XIAP and PHB1 Regulate Anoikis through Competitive Binding to TRAF6. Mol Cancer Res 2023; 21:127-139. [PMID: 36346305 DOI: 10.1158/1541-7786.mcr-22-0415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
Anoikis resistance is a prerequisite for circulating tumor cells to survive. However, the mechanism underlying anoikis resistance is poorly understood. In the current study, the effect of TNF receptor-associated factor 6 (TRAF6)-induced NF-kB activation on anoikis susceptibility in tumor cells was evaluated. Differential TRAF6-binding proteins in anoikis-sensitive versus anoikis-resistant tumor cells were screened by LC/MS-MS analysis. The effects of TRAF6-binding proteins on the stability of TRAF6, the activation of NF-kB signaling and anoikis susceptibility in tumor cells were detected. We found that the loss of TRAF6 expression is an important molecular event linked to anoikis. X-linked inhibitor of apoptosis protein (XIAP), an E3 ligase, can bind, ubiquitinate, and degrade TRAF6 and may lead to inactivation of NF-κB signaling and anoikis sensitivity. High expression of prohibitin 1 (PHB1) competes with XIAP for binding to TRAF6 and confers anoikis resistance to tumor cells. PHB1 and TRAF6 knockdown eliminated tumor cells from the circulation in vivo. Significant correlations between elevated PHB1 and TRAF6 expression and distant metastasis were observed in patients with oral cancer. Collectively, we elucidated a novel mechanism governing anoikis. Our data also indicated that TRAF6 and PHB1 are potential therapeutic targets for tumor cells disseminating in the circulation. IMPLICATIONS Our data implicate that PHB1 competes with XIAP for binding to TRAF6 and confers anoikis resistance to tumor cells.
Collapse
Affiliation(s)
- Bo Yang
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chao Lou
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shengkai Chen
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Qin Xu
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
20
|
van Dyk L, Verhoog NJD, Louw A. Combinatorial treatments of tamoxifen and SM6Met, an extract from Cyclopia subternata Vogel, are superior to either treatment alone in MCF-7 cells. Front Pharmacol 2022; 13:1017690. [PMID: 36210845 PMCID: PMC9535530 DOI: 10.3389/fphar.2022.1017690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Synergistic drug combinations are not only popular in antibiotic, anti-microbial, immune disease (i.e., AIDS) and viral infection studies, but has also gained traction in the field of cancer research as a multi-targeted approach. It has the potential to lower the doses needed of standard of care (SOC) therapeutic agents, whilst maintaining an effective therapeutic level. Lower dosages could ameliorate the fundamental problems such as drug resistance and metastasis associated with current SOC therapies. In the current study, we show that the combination of SM6Met with (2)-4-hydroxytamoxifen (4-OH-Tam, the active metabolite of tamoxifen) produces a strong synergistic effect in terms of inhibiting MCF7 ER-positive (ER+) breast cancer cell proliferation and that a 20 times lower dose of 4-OH-Tam in combination with SM6Met is required to produce the same inhibitory effect on cell proliferation as 4-OH-Tam on its own. Cell cycle analyses of the best combination ratios of SM6Met and 4-OH-Tam also suggests that the combination results in increased accumulation of cells in the S-phase and in the apoptotic phase. Moreover, the best combination ratio (20:1) of SM6Met with 4-OH-Tam displayed greater anti-metastatic potential in terms of inhibiting ER+ breast cancer cell migration, invasion, and colony formation than the SOC therapy alone, suggesting that SM6Met together with 4-OH-Tam could be a viable drug combination for not only delaying resistance and ameliorating the negative side-effects associated with current SOC therapies, like tamoxifen, but could also provide a novel, more affordable therapeutic alternative for treating or preventing ER+ breast cancer metastasis.
Collapse
|
21
|
Isebia KT, Dathathri E, Verschoor N, Nanou A, De Jong AC, Coumans FAW, Terstappen LWMM, Kraan J, Martens JWM, Bansal R, Lolkema MP. Characterizing Circulating Tumor Cells and Tumor-Derived Extracellular Vesicles in Metastatic Castration-Naive and Castration-Resistant Prostate Cancer Patients. Cancers (Basel) 2022; 14:4404. [PMID: 36139564 PMCID: PMC9497200 DOI: 10.3390/cancers14184404] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Circulating tumor cell (CTC)- and/or tumor-derived extracellular vesicle (tdEV) loads in the blood of metastatic castration-resistant prostate cancer (CRPC) patients are associated with worse overall survival and can be used as predictive markers of treatment response. In this study, we investigated the quantity/quality of CTCs and tdEVs in metastatic castration-naive prostate cancer (CNPC) and CRPC patients, and whether androgen deprivation therapy (ADT) affects CTCs and tdEVs. We included 104 CNPC patients before ADT initiation and 66 CRPC patients. Blood samples from 31/104 CNPC patients were obtained 6 months after ADT. CTCs and tdEVs were identified using ACCEPT software. Based on the morphology, CTCs of metastatic CNPC and CRPC patients were subdivided by manual reviewing into six subclasses. The numbers of CTCs and tdEVs were correlated in both CNPC and CRPC patients, and both CTCs (p = 0.013) and tdEVs (p = 0.005) were significantly lower in CNPC compared to CRPC patients. Qualitative differences in CTCs were observed: CTC clusters (p = 0.006) and heterogeneously CK expressing CTCs (p = 0.041) were significantly lower in CNPC patients. CTC/tdEV numbers declined 6 months after ADT. Our study showed that next to CTC-load, qualitative CTC analysis and tdEV-load may be useful in CNPC patients.
Collapse
Affiliation(s)
- Khrystany T. Isebia
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Eshwari Dathathri
- Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Noortje Verschoor
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Afroditi Nanou
- Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Anouk C. De Jong
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Frank A. W. Coumans
- Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Leon W. M. M. Terstappen
- Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Ruchi Bansal
- Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Martijn P. Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
22
|
A Laboratory-Friendly CTC Identification: Comparable Double-Immunocytochemistry with Triple-Immunofluorescence. Cancers (Basel) 2022; 14:cancers14122871. [PMID: 35740537 PMCID: PMC9221448 DOI: 10.3390/cancers14122871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/03/2022] [Accepted: 06/05/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Tumor cells that circulate in the peripheral blood of patients with solid tumors are called circulating tumor cells. Since the source of circulating tumor cells are from primary cancer sites, metastatic sites, and/or a disseminated tumor cell pool, these cells have clinical significance. The circulating tumor cells offer a rare glimpse of the evolution of the tumor and its response/resistance to treatment in a real-time non-invasive manner. Although the clinical relevance of circulating tumor cells is undeniable, the routine use of these cells remains limited due to the elusive nature of the cells, which demands highly sophisticated and costly instrumentation. We presented a specific and sensitive laboratory-friendly parallel double-detection format method for the simultaneous isolation and identification of circulating tumor cells from peripheral blood of 91 consented and enrolled patients with tumors of the lung, endometrium, ovary, esophagus, prostate, and liver. Our user-friendly cost-effective circulating tumor cells detection technique has the potency to facilitate the routine use of circulating tumor cells detection even in community-based cancer centers for prognosis, before and after surgery, which will provide a unique opportunity to move cancer diagnostics forward. Abstract The source of circulating tumor cells (CTC) in the peripheral blood of patients with solid tumors are from primary cancer, metastatic sites, and a disseminated tumor cell pool. As 90% of cancer-related deaths are caused by metastatic progression and/or resistance-associated treatment failure, the above fact justifies the undeniable predictive and prognostic value of identifying CTC in the bloodstream at stages of the disease progression and resistance to treatment. Yet enumeration of CTC remains far from a standard routine procedure either for post-surgery follow-ups or ongoing adjuvant therapy. The most compelling explanation for this paradox is the absence of a convenient, laboratory-friendly, and cost-effective method to determine CTC. We presented a specific and sensitive laboratory-friendly parallel double-detection format method for the simultaneous isolation and identification of CTC from peripheral blood of 91 consented and enrolled patients with various malignant solid tumors of the lung, endometrium, ovary, esophagus, prostate, and liver. Using a pressure-guided method, we used the size-based isolation to capture CTC on a commercially available microfilter. CTC identification was carried out by two expression marker-based independent staining methods, double-immunocytochemistry parallel to standard triple-immunofluorescence. The choice of markers included specific markers for epithelial cells, EpCAM and CK8,18,19, and exclusion markers for WBC, CD45. We tested the method’s specificity based on the validation of the staining method, which included positive and negative spiked samples, blood from the healthy age-matched donor, healthy age-matched leucopaks, and blood from metastatic patients. Our user-friendly cost-effective CTC detection technique may facilitate the regular use of CTC detection even in community-based cancer centers for prognosis, before and after surgery.
Collapse
|
23
|
Wu Y, Fan Z. Peripheral blood RNAseq links neutrophilic inflammation to clinical glioma metastasis. BMC Med 2021; 19:296. [PMID: 34856994 PMCID: PMC8641233 DOI: 10.1186/s12916-021-02174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuanyuan Wu
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, Connecticut, 06030, USA
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, Connecticut, 06030, USA.
| |
Collapse
|
24
|
Ramos-Medina R, López-Tarruella S, del Monte-Millán M, Massarrah T, Martín M. Technical Challenges for CTC Implementation in Breast Cancer. Cancers (Basel) 2021; 13:4619. [PMID: 34572846 PMCID: PMC8466817 DOI: 10.3390/cancers13184619] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the most common neoplasm in women worldwide. Tissue biopsy, currently the gold standard to obtain tumor molecular information, is invasive and might be affected by tumor heterogeneity rendering it incapable to portray the complete dynamic picture by the absence of specific genetic changes during the evolution of the disease. In contrast, liquid biopsy can provide unique opportunities for real-time monitoring of disease progression, treatment response and for studying tumor heterogeneity combining the information of DNA that tumors spread in the blood (circulating tumor DNA) with CTCs analysis. In this review, we analyze the technical and biological challenges for isolation and characterization of circulating tumor cells from breast cancer patients. Circulating tumor cell (CTC) enumeration value is included in numerous clinical studies due to the prognostic's role of these cells. Despite this, there are so many questions pending to answer. How to manage lymphocytes background, how to distinguish the CTCs subtypes or how to work with frozen samples, are some of the issues that will discuss in this review. Based on our experience, we try to address these issues and other technical limitations that should be solved to optimize the standardization of protocols, sample extraction procedures, circulating-tumor material isolation (CTCs vs. ctDNA) and the very diverse methodologies employed, aiming to consolidate the use of CTCs in the clinic. Furthermore, we think that new approaches focusing on isolation CTCs in other body fluids such as cerebrospinal or ascitic fluid are necessary to increase the opportunities of circulating tumor cells in the practice clinic as well as to study the promising role of CTC clusters and their prognostic value in metastatic breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Miguel Martín
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Hospital General Universitario Gregorio Marañón, CIBERONC, Universidad Complutense, 28007 Madrid, Spain; (R.R.-M.); (S.L.-T.); (M.d.M.-M.); (T.M.)
| |
Collapse
|
25
|
Schuster E, Taftaf R, Reduzzi C, Albert MK, Romero-Calvo I, Liu H. Better together: circulating tumor cell clustering in metastatic cancer. Trends Cancer 2021; 7:1020-1032. [PMID: 34481763 PMCID: PMC8541931 DOI: 10.1016/j.trecan.2021.07.001] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/30/2023]
Abstract
Circulating tumor cells (CTCs) are vital components of liquid biopsies for diagnosis of residual cancer, monitoring of therapy response, and prognosis of recurrence. Scientific dogma focuses on metastasis mediated by single CTCs, but advancement of CTC detection technologies has elucidated multicellular CTC clusters, which are associated with unfavorable clinical outcomes and a 20- to 100-fold greater metastatic potential than single CTCs. While the mechanistic understanding of CTC cluster formation is still in its infancy, multiple cell adhesion molecules and tight junction proteins have been identified that underlie the outperforming attributes of homotypic and heterotypic CTC clusters, such as cell survival, cancer stemness, and immune evasion. Future directions include high-resolution characterization of CTCs at multiomic levels for diagnostic/prognostic evaluations and targeted therapies.
Collapse
Affiliation(s)
- Emma Schuster
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rokana Taftaf
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Carolina Reduzzi
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mary K Albert
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Isabel Romero-Calvo
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Lurie Comprehensive Cancer Center and Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Reduzzi C, Di Cosimo S, Gerratana L, Motta R, Martinetti A, Vingiani A, D’Amico P, Zhang Y, Vismara M, Depretto C, Scaperrotta G, Folli S, Pruneri G, Cristofanilli M, Daidone MG, Cappelletti V. Circulating Tumor Cell Clusters Are Frequently Detected in Women with Early-Stage Breast Cancer. Cancers (Basel) 2021; 13:cancers13102356. [PMID: 34068368 PMCID: PMC8153325 DOI: 10.3390/cancers13102356] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/29/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Metastases cause the majority of breast cancer-related deaths. Circulating tumor cells (CTCs), and in particular CTC-clusters, are considered the seeds of metastasis, but their analysis in the early-stages of the disease has so far been limited by the fact that, by using conventional and epithelial-based technologies (as the FDA-approved CellSearch platform), they are more often detected in the metastatic setting. It is known, however, that cancer cells are heterogeneous and can downregulate the expression of epithelial markers, thus limiting the detection capability of epithelial-based technologies. Here, we show that it is possible to increase CTC-cluster detection by using an epithope-independent technology based on blood filtration, and in particular that this strategy allows to detect a high number of CTC-clusters in stage II-III breast cancer patients, before and during neoadjuvant treatment. Our results therefore offer a new opportunity to deepen our understanding of the cancer dissemination process in its early steps. Abstract The clinical relevance of circulating tumor cell clusters (CTC-clusters) in breast cancer (BC) has been mostly studied using the CellSearch®, a marker-dependent method detecting only epithelial-enriched clusters. However, due to epithelial-to-mesenchymal transition, resorting to marker-independent approaches can improve CTC-cluster detection. Blood samples collected from healthy donors and spiked-in with tumor mammospheres, or from BC patients, were processed for CTC-cluster detection with 3 technologies: CellSearch®, CellSieve™ filters, and ScreenCell® filters. In spiked-in samples, the 3 technologies showed similar recovery capability, whereas, in 19 clinical samples processed in parallel with CellSearch® and CellSieve™ filters, filtration allowed us to detect more CTC-clusters than CellSearch® (median number = 7 versus 1, p = 0.0038). Next, samples from 37 early BC (EBC) and 23 metastatic BC (MBC) patients were processed using ScreenCell® filters for attaining both unbiased enrichment and marker-independent identification (based on cytomorphological criteria). At baseline, CTC-clusters were detected in 70% of EBC cases and in 20% of MBC patients (median number = 2, range 0–20, versus 0, range 0–15, p = 0.0015). Marker-independent approaches for CTC-cluster assessment improve detection and show that CTC-clusters are more frequent in EBC than in MBC patients, a novel finding suggesting that dissemination of CTC-clusters is an early event in BC natural history.
Collapse
Affiliation(s)
- Carolina Reduzzi
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.G.); (P.D.); (Y.Z.); (M.C.)
| | - Serena Di Cosimo
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
| | - Lorenzo Gerratana
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.G.); (P.D.); (Y.Z.); (M.C.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Rosita Motta
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
| | - Antonia Martinetti
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Andrea Vingiani
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giacomo Venezian 1, 20133 Milan, Italy; (A.V.); (G.P.)
- Oncology and Hemato-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Paolo D’Amico
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.G.); (P.D.); (Y.Z.); (M.C.)
- New Drugs and Early Drug Development for Innovative Therapies Division, IEO, European Institute of Oncology IRCCS, 20133 Milan, Italy
| | - Youbin Zhang
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.G.); (P.D.); (Y.Z.); (M.C.)
| | - Marta Vismara
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
| | - Catherine Depretto
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.D.); (G.S.)
| | - Gianfranco Scaperrotta
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.D.); (G.S.)
| | - Secondo Folli
- Breast Cancer Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy;
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giacomo Venezian 1, 20133 Milan, Italy; (A.V.); (G.P.)
- Oncology and Hemato-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Massimo Cristofanilli
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.G.); (P.D.); (Y.Z.); (M.C.)
| | - Maria Grazia Daidone
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
| | - Vera Cappelletti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (C.R.); (S.D.C.); (R.M.); (M.V.); (M.G.D.)
- Correspondence: ; Tel.: +39-022390-2700
| |
Collapse
|
27
|
Silvestri M, Reduzzi C, Feliciello G, Vismara M, Schamberger T, Köstler C, Motta R, Calza S, Ferraris C, Vingiani A, Pruneri G, Daidone MG, Klein CA, Polzer B, Cappelletti V. Detection of Genomically Aberrant Cells within Circulating Tumor Microemboli (CTMs) Isolated from Early-Stage Breast Cancer Patients. Cancers (Basel) 2021; 13:cancers13061409. [PMID: 33808748 PMCID: PMC8003526 DOI: 10.3390/cancers13061409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Distant metastases derive from the shedding and dissemination of single cancer cells (CTCs) or circulating tumor emboli (CTMs) into circulation. Previous studies on CTMs were mainly run in patients with metastatic disease; however, we observed that CTMs are more frequently detected in patients with early-stage breast cancer. Here, we collected single CTMs and their relative primary tumor tissue samples in early-stage patients. By studying genomic aberrations, present in tumors cells and absent in normal cells, we predicted the tumor fraction thanks to a statistical model developed from a calibration curve with breast cancer cell lines. The tumor fraction ranged from 8% to 48% and CTMs contained specific and shared alterations with respect to tissue. Thus, CTMs may derive from different regions of the primary tumor or from occult micrometastases. Moreover, CTM-private mutations may inform us about specific metastasis-associated functions of involved genes that should be further explored in follow-up and mechanistic studies. Abstract Circulating tumor microemboli (CTMs) are clusters of cancer cells detached from solid tumors, whose study can reveal mechanisms underlying metastatization. As they frequently comprise unknown fractions of leukocytes, the analysis of copy number alterations (CNAs) is challenging. To address this, we titrated known numbers of leukocytes into cancer cells (MDA-MB-453 and MDA-MB-36, displaying high and low DNA content, respectively) generating tumor fractions from 0–100%. After low-pass sequencing, ichorCNA was identified as the best algorithm to build a linear mixed regression model for tumor fraction (TF) prediction. We then isolated 53 CTMs from blood samples of six early-stage breast cancer patients and predicted the TF of all clusters. We found that all clusters harbor cancer cells between 8 and 48%. Furthermore, by comparing the identified CNAs of CTMs with their matched primary tumors, we noted that only 31–71% of aberrations were shared. Surprisingly, CTM-private alterations were abundant (30–63%), whereas primary tumor-private alterations were rare (4–12%). This either indicates that CTMs are disseminated from further progressed regions of the primary tumor or stem from cancer cells already colonizing distant sites. In both cases, CTM-private mutations may inform us about specific metastasis-associated functions of involved genes that should be explored in follow-up and mechanistic studies.
Collapse
Affiliation(s)
- Marco Silvestri
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
| | - Carolina Reduzzi
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
| | - Giancarlo Feliciello
- Division Personalized Tumor Therapy, Fraunhofer-Institute for Toxicology and Experimental Medicine, Biopark 1|Am Biopark 9, 93053 Regensburg, Germany; (G.F.); (C.K.); (C.A.K.); (B.P.)
| | - Marta Vismara
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
| | - Thomas Schamberger
- Experimental Medicine and Therapy Research, University Regensburg, Franz-Josef-Strauss Allee 11, 93040 Regensburg, Germany;
| | - Cäcilia Köstler
- Division Personalized Tumor Therapy, Fraunhofer-Institute for Toxicology and Experimental Medicine, Biopark 1|Am Biopark 9, 93053 Regensburg, Germany; (G.F.); (C.K.); (C.A.K.); (B.P.)
| | - Rosita Motta
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
| | - Stefano Calza
- Unit of Biostatistics, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25125 Brescia, Italy;
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Cristina Ferraris
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori di Milano, Via Venezian 1, 20133 Milano, Italy;
| | - Andrea Vingiani
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giacomo Venezian 1, 20133 Milan, Italy; (A.V.); (G.P.)
- Oncology and Hemato-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giacomo Venezian 1, 20133 Milan, Italy; (A.V.); (G.P.)
- Oncology and Hemato-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Maria Grazia Daidone
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
| | - Christoph A. Klein
- Division Personalized Tumor Therapy, Fraunhofer-Institute for Toxicology and Experimental Medicine, Biopark 1|Am Biopark 9, 93053 Regensburg, Germany; (G.F.); (C.K.); (C.A.K.); (B.P.)
- Experimental Medicine and Therapy Research, University Regensburg, Franz-Josef-Strauss Allee 11, 93040 Regensburg, Germany;
| | - Bernhard Polzer
- Division Personalized Tumor Therapy, Fraunhofer-Institute for Toxicology and Experimental Medicine, Biopark 1|Am Biopark 9, 93053 Regensburg, Germany; (G.F.); (C.K.); (C.A.K.); (B.P.)
| | - Vera Cappelletti
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Giovanni Antonio Amadeo 42, 20133 Milano, Italy; (M.S.); (C.R.); (M.V.); (R.M.); (M.G.D.)
- Correspondence:
| |
Collapse
|
28
|
Hurtado P, Martínez-Pena I, Piñeiro R. Dangerous Liaisons: Circulating Tumor Cells (CTCs) and Cancer-Associated Fibroblasts (CAFs). Cancers (Basel) 2020; 12:E2861. [PMID: 33027902 PMCID: PMC7599894 DOI: 10.3390/cancers12102861] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
The crosstalk between cancer cells and the tumor microenvironment (TME) is a key determinant of cancer metastasis. Cancer-associated fibroblasts (CAFs), one of the main cellular components of TME, promote cancer cell invasion and dissemination through mechanisms including cell-cell interactions and the paracrine secretion of growth factors, cytokines and chemokines. During metastasis, circulating tumor cells (CTCs) are shed from the primary tumor to the bloodstream, where they can be detected as single cells or clusters. The current knowledge about the biology of CTC clusters positions them as key actors in metastasis formation. It also indicates that CTCs do not act alone and that they may be aided by stromal and immune cells, which seem to shape their metastatic potential. Among these cells, CAFs are found associated with CTCs in heterotypic CTC clusters, and their presence seems to increase their metastatic efficiency. In this review, we summarize the current knowledge on the role that CAFs play on metastasis and we discuss their implication on the biogenesis, metastasis-initiating capacity of CTC clusters, and clinical implications. Moreover, we speculate about possible therapeutic strategies aimed to limit the metastatic potential of CTC clusters involving the targeting of CAFs as well as their difficulties and limitations.
Collapse
Affiliation(s)
- Pablo Hurtado
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Inés Martínez-Pena
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain
| |
Collapse
|