1
|
Jones D, Whitehead CA, Dinevska M, Widodo SS, Furst LM, Morokoff AP, Kaye AH, Drummond KJ, Mantamadiotis T, Stylli SS. Repurposing FDA-approved drugs as inhibitors of therapy-induced invadopodia activity in glioblastoma cells. Mol Cell Biochem 2023; 478:1251-1267. [PMID: 36302993 PMCID: PMC10164021 DOI: 10.1007/s11010-022-04584-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Glioblastoma (GBM) is the most prevalent primary central nervous system tumour in adults. The lethality of GBM lies in its highly invasive, infiltrative, and neurologically destructive nature resulting in treatment failure, tumour recurrence and death. Even with current standard of care treatment with surgery, radiotherapy and chemotherapy, surviving tumour cells invade throughout the brain. We have previously shown that this invasive phenotype is facilitated by actin-rich, membrane-based structures known as invadopodia. The formation and matrix degrading activity of invadopodia is enhanced in GBM cells that survive treatment. Drug repurposing provides a means of identifying new therapeutic applications for existing drugs without the need for discovery or development and the associated time for clinical implementation. We investigate several FDA-approved agents for their ability to act as both cytotoxic agents in reducing cell viability and as 'anti-invadopodia' agents in GBM cell lines. Based on their cytotoxicity profile, three agents were selected, bortezomib, everolimus and fludarabine, to test their effect on GBM cell invasion. All three drugs reduced radiation/temozolomide-induced invadopodia activity, in addition to reducing GBM cell viability. These drugs demonstrate efficacious properties warranting further investigation with the potential to be implemented as part of the treatment regime for GBM.
Collapse
Affiliation(s)
- Dylan Jones
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Clarissa A Whitehead
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Marija Dinevska
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Samuel S Widodo
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Liam M Furst
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Hadassah University Medical Centre, 91120, Jerusalem, Israel
| | - Katharine J Drummond
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Theo Mantamadiotis
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stanley S Stylli
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
| |
Collapse
|
2
|
Das Ghosh D, Roy Chowdhury R, Dutta R, Mukhopadhyay I, Mukhopadhyay A, Roychoudhury S. In-silico analysis of TCGA data showing multiple POLE-like favourable subgroups overlapping with TP53 mutated endometrial cancer: Implications for clinical practice in low and middle-income countries. Gynecol Oncol Rep 2023; 47:101209. [PMID: 37293351 PMCID: PMC10245003 DOI: 10.1016/j.gore.2023.101209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The Cancer Genome Atlas cohort of endometrial carcinoma (TCGA-UCEC) includes almost 40% TP53-mutants encompassing missense and truncated variants. TCGA revealed 'POLE', characterized by POLE gene bearing exonuclease domain mutation (EDM), as the prognostically best molecular profile. The worst profile was characterized by TP53-mutated Type 2 cancer requiring adjuvant therapy having cost implications in low-resource settings. We aimed to find more 'POLE-like' favourable subgroups by searching TCGA cohort, especially within TP53 mutated risk group, that could eventually avoid adjuvant treatment in resource-poor settings. Method Our study was an in-silico survival analysis performed on the TCGA-UCEC dataset using SPSS statistical package. TP53 and POLE mutations, microsatellite instability (MSI), time-to-event and clinicopathological parameters were compared among 512 endometrial cancer cases. Deleterious POLE-mutations were identified by Polyphen2. Progression free survival was studied using Kaplan-Meier plots keeping original 'POLE' as comparator. Result In presence of wild type (WT)-TP53, other deleterious POLE-mutations behaved like POLE-EDM. Only truncated and not missense TP53 benefitted from POLE/MSI overlap. However, TP53 missense mutation, Y220C, was found to be as favourable as 'POLE'. Overlapping POLE, MSI and WT-TP53 also performed favourably. Truncated TP53 overlapped with POLE and/or MSI, TP53 Y220C alone and, WT-TP53 overlapped with POLE and MSI both, were named 'POLE-like' for prognostically behaving like the comparator 'POLE'. Conclusion Obesity being a lesser frequent event in low and middle-income countries (LMICs), relative proportion of women with lower BMI and Type 2 endometrial cancers may be high. Identification of 'POLE-like' groups may facilitate therapeutic de-escalation in some TP53-mutated cases - a novel option. Instead of 5% (POLE-EDM), potential beneficiary would then comprise 10% (POLE-like) of TCGA-UCEC.
Collapse
Affiliation(s)
- Damayanti Das Ghosh
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
- Kolkata Gynecological Oncology Trials and Translational Research Group, Kolkata, India
| | - Rahul Roy Chowdhury
- Department of Gynecological Oncology, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
- Kolkata Gynecological Oncology Trials and Translational Research Group, Kolkata, India
| | - Rajeswari Dutta
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | | | - Asima Mukhopadhyay
- Kolkata Gynecological Oncology Trials and Translational Research Group, Kolkata, India
| | - Susanta Roychoudhury
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
- Kolkata Gynecological Oncology Trials and Translational Research Group, Kolkata, India
| |
Collapse
|
3
|
Michishita M, Ochiai K, Nakahira R, Azakami D, Machida Y, Nagashima T, Nakagawa T, Ishiwata T. mTOR pathway as a potential therapeutic target for cancer stem cells in canine mammary carcinoma. Front Oncol 2023; 13:1100602. [PMID: 36816969 PMCID: PMC9931192 DOI: 10.3389/fonc.2023.1100602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Mammary adenocarcinoma, the most common cancer in female dogs, often exhibits the lymph node and lung metastases and has a higher mortality rate. However, mammary adenocarcinoma has no established treatment, except early surgical excision. Canine mammary carcinoma has many common features with human mammary carcinoma, including clinical characteristics, heterogeneity, and genetic aberrations, making it an excellent spontaneous tumor model for human breast cancer. Diverse cancers comprised heterogeneous cell populations originating from cancer stem cells (CSCs) with self-renewal ability. Therefore, in addition to conventional therapy, therapeutic strategies targeting CSCs are essential for cancer eradication. The present study aimed to extract inhibitors of canine mammary CSCs that suppress their self-renewal ability. Sphere-formation assay, which evaluates self-renewal ability, was performed for the canine mammary cancer cell lines CTBp and CNMp. The spheres formed in this assay were used in inhibitor library screening, which identified various signaling pathways such as proteosome, stress inducer, and mammalian target of rapamycin (mTOR). The present study focused on the mTOR signaling pathway. Western blotting showed higher levels of phosphorylated mTOR in sphere-forming CTBp and CNMp cells than in adherent cells. Drug sensitivity examination using the mTOR inhibitors everolimus and temsirolimus revealed dose-dependent reductions in viability among both sphere-forming cells and adherent cells. Expression of phosphorylated mTOR in adherent and sphere-forming cells decreased by everolimus and temsirolimus treatment. In mice transplanted with CTBp-derived spheres, everolimus treatment significantly decreased tumor volume compared to control. These results reveal that the mTOR signaling pathway may be a potential to be a therapeutic target in both cancer cells and CSCs. Novel therapeutic strategies for canine mammary carcinoma are expected to benefit to human breast carcinoma as well.
Collapse
Affiliation(s)
- Masaki Michishita
- Department of Veterinary Pathology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan,Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Tokyo, Japan,*Correspondence: Masaki Michishita,
| | - Kazuhiko Ochiai
- Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Tokyo, Japan,Department of Veterinary Hygiene, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Rei Nakahira
- Department of Veterinary Pathology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Daigo Azakami
- Laboratory of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Yukino Machida
- Department of Veterinary Pathology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tomokazu Nagashima
- Department of Veterinary Pathology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Ishiwata
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
4
|
Tabbal M, Hachim MY, Jan RK, Adrian TE. Using publicly available datasets to identify population-based transcriptomic landscape contributing to the aggressiveness of breast cancer in young women. Front Genet 2023; 13:1039037. [PMID: 36685821 PMCID: PMC9845274 DOI: 10.3389/fgene.2022.1039037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/30/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: Although the risk of breast cancer increases with advancing age, some regions have larger number of young breast cancer patients (≤45 years-old), such as the Middle East, Eastern Asia, and North Africa, with more aggressive and poorly differentiated tumors. We aimed to conduct an in-silico analysis in an attempt to understand the aggressive nature of early-onset breast cancer, and to identify potential drivers of early-onset breast cancer using gene expression profiling datasets in a population-dependent manner. Methods: Functional genomics experiments data were acquired from cBioPortal database for cancer genomics, followed by the stratification of patients based on the age at representation of breast cancer and race. Differential gene expression analysis and gene amplification status analysis were carried out, followed by hub gene, transcription factor, and signalling pathway identification. Results: PAM50 subtype analysis revealed that young patients (≤45 years-old) had four-fold more basal tumors and worst progression-free survival (median of 101 months), compared with the 45-65 years group (median of 168 months). Fourteen genes were amplified in more than 14% of patients with an early-onset breast cancer. Interestingly, FREM2, LINC00332, and LINC00366 were exclusively amplified in younger patients. Gene expression data from three different populations (Asian, White, and African) revealed a unique transcriptomic profile of young patients, which was also reflected on the PAM50 subtype analysis. Our data indicates a higher tendency of young African patients to develop basal tumors, while young Asian patients are more prone to developing Luminal A tumors. Most genes that were found to be upregulated in younger patients are involved in important signaling pathways that promote cancer progression and metastasis, such as MAPK pathway, Reelin pathway and the PI3K/Akt pathway. Conclusion: This study provides strong evidence that the molecular profile of tumors derived from young breast cancer patients of different populations is unique and may explain the aggressiveness of these tumors, stressing the need to conduct population- based multi-omic analyses to identify the potential drivers for tumorigenesis and molecular profiles of young breast cancer patients.
Collapse
Affiliation(s)
| | | | | | - Thomas E. Adrian
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| |
Collapse
|
5
|
Identifying Circulating Tumor DNA Mutations Associated with Neoadjuvant Chemotherapy Efficacy in Local Advanced Breast Cancer. Appl Biochem Biotechnol 2022; 194:3961-3973. [PMID: 35579744 DOI: 10.1007/s12010-022-03946-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Circulating tumor DNA (ctDNA) correlates with tumor burden and provides early detection of treatment response and tumor genetic alterations in breast cancer. Neoadjuvant chemotherapy (NACT) has become standard therapy for local advanced breast cancer (LABC). The aim of our study was to investigate plasma ctDNA as a prognostic marker for outcome in patients with LABC treated with NACT. A total of 56 patients with LABC were involved in this study. ctDNA mutations were investigated by using a 100 gene panel-target capture next-generation sequencing. The patients then received standard NACT therapy: adriamycin and cyclophosphamide and paclitaxel (AC-T) or AC-TH (AC-T+ Trastuzumab) regimen. The efficacy of NACT was evaluated by Miller-Payne grading system. A predictive and weight model was used to screen ctDNA point mutation biomarkers for NACT. The ctDNA mutational profile of LABC patients was identified. For nonsynonymous mutations, the top 5 mutated genes were MTHFR (51/56, 91.1%), XPC (50/56, 89.3%), ABCB1 (48/51, 94.1%), BRCA2 (38/56, 67.9%), and XRCC1 (38/56, 67.9%). In addition, the mutation frequencies of PIK3CA and TP53 were 32.1% (18/56) and 26.8% (15/56), respectively. The predictive model indicated that XRCC1 44055726 (TG>-) mutation (25/56, 44.6%) was significantly associated with Miller-Payne 4-5 and Miller-Payne 3-5 responses. While mTOR 11249132(G>C) mutation (23/56, 41.1%) was associated with Miller-Payne 1-4 or Miller-Payne 1-3 responses. Furthermore, XRCC1 44055726 (TG>-) accompanied by mTOR wild type predicted a good NACT efficacy in all response classification systems. The ROC curves to discriminate good neoadjuvant chemotherapy efficiency (Miller-Payne 4-5) and poor efficiency (Miller-Payne 1-3) were created, and AUC value was 0.77. Our results suggested that ctDNA mutation of XRCC1 44055726 (TG>-) might be a positive biomarker for NACT therapy in LABC, while mTOR 11249132(G>C) mutation was potentially associated with NACT resistance.
Collapse
|
6
|
Kumar SU, Wang H, Telichko AV, Natarajan A, Bettinger T, Cherkaoui S, Massoud TF, Dahl JJ, Paulmurugan R. Ultrasound Triggered Co‐Delivery of Therapeutic MicroRNAs and a Triple Suicide Gene Therapy Vector by Using Biocompatible Polymer Nanoparticles for Improved Cancer Therapy in Mouse Models. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Huaijun Wang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Arsenii V. Telichko
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Thierry Bettinger
- Novel Agents Department Bracco Suisse SA Route de la Galaise 31 1228 Plan‐les‐Ouates Geneva Switzerland
| | - Samir Cherkaoui
- Novel Agents Department Bracco Suisse SA Route de la Galaise 31 1228 Plan‐les‐Ouates Geneva Switzerland
| | - Tarik F. Massoud
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Jeremy J. Dahl
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| |
Collapse
|
7
|
Philipovskiy A, Dwivedi AK, Gamez R, McCallum R, Mukherjee D, Nahleh Z, Aguilera RJ, Gaur S. Association between tumor mutation profile and clinical outcomes among Hispanic Latina women with triple-negative breast cancer. PLoS One 2020; 15:e0238262. [PMID: 32886682 PMCID: PMC7473586 DOI: 10.1371/journal.pone.0238262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) represents 15%-20% of all breast cancer types. It is more common among African American (AA) and Hispanic-Latina (HL) women. The biology of TNBC in HL women has been poorly characterized, but some data suggest that the molecular drivers of breast cancer might differ. There are no clinical tools to aid medical oncologists with decisions regarding appropriate individualized therapy, and no way to predict long-term outcomes. The aim of this study was to characterize individual patient gene mutation profiles and to identify the relationship with clinical outcomes. We collected formalin-fixed paraffin-embedded tumors (FFPE) from women with TNBC. We analyzed the gene mutation profiles of the collected tumors and compared the results with individual patient's clinical histories and outcomes. Of 25 patients with TNBC, 24 (96%) identified as HL. Twenty-one (84%) had stage III-IV disease. The most commonly mutated genes were TP53, NOTCH1, NOTCH2, NOTCH3, AKT, MEP3K, PIK3CA, and EGFR. Compared with other international cancer databases, our study demonstrated statistically significant higher frequencies of these genes among HL women. Additionally, a worse clinical course was observed among patients whose tumors had mutations in NOTCH genes and PIK3CA. This study is the first to identify the most common genetic alterations among HL women with TNBC. Our data strongly support the notion that molecular drivers of breast cancer could differ in HL women compared with other ethnic backgrounds. Therefore, a deeper understanding of the biological mechanisms behind NOTCH gene and PIK3CA mutations may lead to a new treatment approach.
Collapse
Affiliation(s)
- Alexander Philipovskiy
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Alok K. Dwivedi
- Division of Biostatistics & Epidemiology, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Roberto Gamez
- Department of Pathology, University Medical Center, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Richard McCallum
- Division of Gastroenterology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Debabrata Mukherjee
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Zeina Nahleh
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic, Florida, Weston, Florida, United States of America
| | - Renato J. Aguilera
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| | - Sumit Gaur
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| |
Collapse
|
8
|
Machine-learned analysis of the association of next-generation sequencing-based genotypes with persistent pain after breast cancer surgery. Pain 2020; 160:2263-2277. [PMID: 31107411 DOI: 10.1097/j.pain.0000000000001616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer and its surgical treatment are among the most important triggering events for persistent pain, but additional factors need to be present for the clinical manifestation, such as variants in pain-relevant genes. In a cohort of 140 women undergoing breast cancer surgery, assigned based on a 3-year follow-up to either a persistent or nonpersistent pain phenotype, next-generation sequencing was performed for 77 genes selected for known functional involvement in persistent pain. Applying machine-learning and item categorization techniques, 21 variants in 13 different genes were found to be relevant to the assignment of a patient to either the persistent pain or the nonpersistent pain phenotype group. In descending order of importance for correct group assignment, the relevant genes comprised DRD1, FAAH, GCH1, GPR132, OPRM1, DRD3, RELN, GABRA5, NF1, COMT, TRPA1, ABHD6, and DRD4, of which one in the DRD4 gene was a novel discovery. Particularly relevant variants were found in the DRD1 and GPR132 genes, or in a cis-eCTL position of the OPRM1 gene. Supervised machine-learning-based classifiers, trained with 2/3 of the data, identified the correct pain phenotype group in the remaining 1/3 of the patients at accuracies and areas under the receiver operator characteristic curves of 65% to 72%. When using conservative classical statistical approaches, none of the variants passed α-corrected testing. The present data analysis approach, using machine learning and training artificial intelligences, provided biologically plausible results and outperformed classical approaches to genotype-phenotype association.
Collapse
|
9
|
Chun BM, Page DB, McArthur HL. Combination Immunotherapy Strategies in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00333-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
We summarize combination immunotherapy strategies for the treatment of breast cancer, with a focus on metastatic disease. First, a general overview of combination approaches is presented according to breast cancer subtype. Second, additional review of promising combination approaches is presented.
Recent Findings
Combination strategies utilizing chemotherapy or radiotherapy with immune checkpoint inhibition are being evaluated across multiple phase III trials. Dual immunotherapy strategies, such as dual immune checkpoint inhibition or combined co-stimulation/co-inhibition, have supportive preclinical evidence and are under early clinical investigation. Modulation of the immune microenvironment via cytokines and vaccination strategies, as well as locally focused treatments to enhance antigenic responses, are active areas of research.
Summary
Pre-clinical and translational research sheds new light on numerous ways the immune system may be modulated to fight against cancer. We describe current and emerging combination approaches which may improve patient outcomes in metastatic breast cancer.
Collapse
|
10
|
Targeting Proteotoxic Stress in Cancer: A Review of the Role that Protein Quality Control Pathways Play in Oncogenesis. Cancers (Basel) 2019; 11:cancers11010066. [PMID: 30634515 PMCID: PMC6356294 DOI: 10.3390/cancers11010066] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/24/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Despite significant advances in cancer diagnostics and therapeutics the majority of cancer unfortunately remains incurable, which has led to continued research to better understand its exceptionally diverse biology. As a result of genomic instability, cancer cells typically have elevated proteotoxic stress. Recent appreciation of this functional link between the two secondary hallmarks of cancer: aneuploidy (oxidative stress) and proteotoxic stress, has therefore led to the development of new anticancer therapies targeting this emerging “Achilles heel” of malignancy. This review highlights the importance of managing proteotoxic stress for cancer cell survival and provides an overview of the integral role proteostasis pathways play in the maintenance of protein homeostasis. We further review the efforts undertaken to exploit proteotoxic stress in multiple myeloma (as an example of a hematologic malignancy) and triple negative breast cancer (as an example of a solid tumor), and give examples of: (1) FDA-approved therapies in routine clinical use; and (2) promising therapies currently in clinical trials. Finally, we provide new insights gleaned from the use of emerging technologies to disrupt the protein secretory pathway and repurpose E3 ligases to achieve targeted protein degradation.
Collapse
|
11
|
Salaroglio IC, Gazzano E, Abdullrahman A, Mungo E, Castella B, Abd-Elrahman GEFAE, Massaia M, Donadelli M, Rubinstein M, Riganti C, Kopecka J. Increasing intratumor C/EBP-β LIP and nitric oxide levels overcome resistance to doxorubicin in triple negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:286. [PMID: 30482226 PMCID: PMC6258159 DOI: 10.1186/s13046-018-0967-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Triple negative breast cancer (TNBC) easily develops resistance to the first-line drug doxorubicin, because of the high levels of the drug efflux transporter P-glycoprotein (Pgp) and the activation of pro-survival pathways dependent on endoplasmic reticulum (ER). Interfering with these mechanisms may overcome the resistance to doxorubicin, a still unmet need in TNBC. METHODS We analyzed a panel of human and murine breast cancer cells for their resistance to doxorubicin, Pgp expression, lysosome and proteasome activity, nitrite production, ER-dependent cell death and immunogenic cell death parameters. We evaluated the efficacy of genetic (C/EBP-β LIP induction) and pharmacological strategies (lysosome and proteasome inhibitors), in restoring the ER-dependent and immunogenic-dependent cell death induced by doxorubicin, in vitro and in syngeneic mice bearing chemoresistant TNBC. The results were analyzed by one-way analysis of variance test. RESULTS We found that TNBC cells characterized by high levels of Pgp and resistance to doxorubicin, had low induction of the ER-dependent pro-apoptotic factor C/EBP-β LIP upon doxorubicin treatment and high activities of lysosome and proteasome that constitutively destroyed LIP. The combination of chloroquine and bortezomib restored doxorubicin sensitivity by activating multiple and interconnected mechanisms. First, chloroquine and bortezomib prevented C/EBP-β LIP degradation and activated LIP-dependent CHOP/TRB3/caspase 3 axis in response to doxorubicin. Second, C/EBP-β LIP down-regulated Pgp and up-regulated calreticulin that triggered the dendritic cell (DC)-mediated phagocytosis of tumor cell, followed by the activation of anti-tumor CD8+T-lymphocytes upon doxorubicin treatment. Third, chloroquine and bortezomib increased the endogenous production of nitric oxide that further induced C/EBP-β LIP and inhibited Pgp activity, enhancing doxorubicin's cytotoxicity. In orthotopic models of resistant TNBC, intratumor C/EBP-β LIP induction - achieved by a specific expression vector or by chloroquine and bortezomib - effectively reduced tumor growth and Pgp expression, increased intra-tumor apoptosis and anti-tumor immune-infiltrate, rescuing the efficacy of doxorubicin. CONCLUSIONS We suggest that preventing C/EBP-β LIP degradation by lysosome and proteasome inhibitors triggers multiple virtuous circuitries that restore ER-dependent apoptosis, down-regulate Pgp and re-activate the DC/CD8+T-lymphocytes response against TNBC. Lysosome and proteasome inhibitors associated with doxorubicin may overcome the resistance to the drug in TNBC.
Collapse
Affiliation(s)
- Iris C Salaroglio
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Ahmad Abdullrahman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Eleonora Mungo
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Barbara Castella
- Laboratory of Blood Tumor Immunology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Gamal Eldein Fathy Abd-Ellatef Abd-Elrahman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.,Pharmaceutical and Drug Industries Research Division, Therapeutic Chemistry Department, National Research Centre, Cairo, Egypt
| | - Massimo Massaia
- Laboratory of Blood Tumor Immunology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Hematology Division, AO S Croce e Carle, Cuneo, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Menachem Rubinstein
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.
| |
Collapse
|
12
|
Forster M, Mark A, Egberts F, Rosati E, Rodriguez E, Stanulla M, Bauerschlag D, Schem C, Maass N, Amallraja A, Murphy KK, Prouse BR, Sulaiman RA, Young BM, Mathiak M, Hemmrich-Stanisak G, Ellinghaus D, Weidinger S, Rosenstiel P, Arnold N, Leyland-Jones B, Williams CB, Franke A, Meißner T. RNA based individualized drug selection in breast cancer patients without patient-matched normal tissue. Oncotarget 2018; 9:32362-32372. [PMID: 30190792 PMCID: PMC6122351 DOI: 10.18632/oncotarget.25981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/04/2018] [Indexed: 11/25/2022] Open
Abstract
Background While standard RNA expression tests stratify patients into risk groups, RNA-Seq can guide personalized drug selection based on expressed mutations, fusion genes, and differential expression (DE) between tumor and normal tissue. However, patient-matched normal tissue may be unavailable. Additionally, biological variability in normal tissue and technological biases may confound results. Therefore, we present normal expression reference data for two sequencing methods that are suitable for breast biopsies. Results We identified breast cancer related and drug related genes that are expressed uniformly across our normal samples. Large subsets of these genes are identical for formalin fixed paraffin embedded samples and fresh frozen samples. Adipocyte signatures were detected in frozen compared to formalin samples, prepared by surgeons and pathologists, respectively. Gene expression confounded by adipocytes was identified using fat tissue samples. Finally, immune repertoire statistics were obtained for healthy breast, tumor and fat tissues. Conclusions Our reference data can be used with patient tumor samples that are asservated and sequenced with a matching aforementioned method. Coefficients of variation are given for normal gene expression. Thus, potential drug selection can be based on confidently overexpressed genes and immune repertoire statistics. Materials and Methods Normal expression from formalin and frozen healthy breast tissue samples using Roche Kapa RiboErase (total RNA) (19 formalin, 9 frozen) and Illumina TruSeq RNA Access (targeted RNA-Seq, aka TruSeq RNA Exome) (11 formalin, 1 frozen), and fat tissue (6 frozen Access). Tumor DE using 10 formalin total RNA tumor samples and 1 frozen targeted RNA tumor sample.
Collapse
Affiliation(s)
- Michael Forster
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Adam Mark
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany.,Current address: Center for Computational Biology and Bioinformatics, Department of Medicine, University of California, San Diego, CA, USA
| | - Friederike Egberts
- Department of Dermatology, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Elisa Rosati
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Elke Rodriguez
- Department of Dermatology, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Martin Stanulla
- Department of Pediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Dirk Bauerschlag
- Department of Gynaecology and Obstetrics, Schleswig-Holstein University Hospital, Kiel, Germany
| | | | - Nicolai Maass
- Department of Gynaecology and Obstetrics, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Anu Amallraja
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
| | | | | | | | - Brandon M Young
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Micaela Mathiak
- Department of Pathology, Schleswig-Holstein University Hospital, Kiel, Germany
| | | | - David Ellinghaus
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Stephan Weidinger
- Department of Dermatology, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Norbert Arnold
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany.,Department of Gynaecology and Obstetrics, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Brian Leyland-Jones
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Casey B Williams
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Tobias Meißner
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, SD, USA
| |
Collapse
|
13
|
Proteasome inhibition induces IKK-dependent interleukin-8 expression in triple negative breast cancer cells: Opportunity for combination therapy. PLoS One 2018; 13:e0201858. [PMID: 30089134 PMCID: PMC6082561 DOI: 10.1371/journal.pone.0201858] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) cells express increased levels of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which promotes their proliferation and migration. Because TNBC patients are unresponsive to current targeted therapies, new therapeutic strategies are urgently needed. While proteasome inhibition by bortezomib (BZ) or carfilzomib (CZ) has been effective in treating hematological malignancies, it has been less effective in solid tumors, including TNBC, but the mechanisms are incompletely understood. Here we report that proteasome inhibition significantly increases expression of IL-8, and its receptors CXCR1 and CXCR2, in TNBC cells. Suppression or neutralization of the BZ-induced IL-8 potentiates the BZ cytotoxic and anti-proliferative effect in TNBC cells. The IL-8 expression induced by proteasome inhibition in TNBC cells is mediated by IκB kinase (IKK), increased nuclear accumulation of p65 NFκB, and by IKK-dependent p65 recruitment to IL-8 promoter. Importantly, inhibition of IKK activity significantly decreases proliferation, migration, and invasion of BZ-treated TNBC cells. These data provide the first evidence demonstrating that proteasome inhibition increases the IL-8 signaling in TNBC cells, and suggesting that IKK inhibitors may increase effectiveness of proteasome inhibitors in treating TNBC.
Collapse
|